Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Front Pharmacol ; 14: 1223808, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37663267

RESUMEN

Introduction: ß-chloroprene (2-chloro-1,3-butadiene; CP) causes lung tumors after inhalation exposures in rats and mice. Mice develop these tumors at lower exposures than rats. In rats CP exposures cause depletion of lung glutathione (GSH). Methods: PBPK models developed to relate the appearance of mouse lung tumors with rates of CP metabolism to reactive metabolites or total amounts metabolized during exposures have been expanded to include production of reactive metabolites from CP. The extended PBPK model describes both the unstable oxirane metabolite, 2-CEO, and metabolism of the more stable oxirane, 1-CEO, to reactive metabolites via microsomal oxidation to a diepoxide, and linked production of these metabolites to a PK model predicting GSH depletion with increasing CP exposure. Key information required to develop the model were available from literature studies identifying: 1) microsomal metabolites of CP, and 2) in vitro rates of clearance of CP and 1-CEO from active microsomal preparations from mice, rats, hamsters and humans. Results: Model simulation of concentration dependence of disproportionate increases in reactive metabolite concentrations as exposures increases and decreases in tissue GSH are consistent with the dose-dependence of tumor formation. At the middle bioassay concentrations with a lung tumor incidence, the predicted tissue GSH is less than 50% background. These simulations of reduction in GSH are also consistent with the gene expression results showing the most sensitive pathways are Nrf2-regulation of oxidative stress and GSH metabolism. Discussion: The PBPK model is used to correlate predicted tissue exposure to reactive metabolites with toxicity and carcinogenicity of CP.

3.
Toxicol Appl Pharmacol ; 380: 114695, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31394159

RESUMEN

A previously published human PBPK model for manganese (Mn) in infants and children has been updated with Mn in drinking water as an additional exposure source. Built upon the ability to capture differences in Mn source-specific regulation of intestinal uptake in nursing infants who are breast-fed and formula-fed, the updated model now describes the bioavailability of Mn from drinking water in children of ages 0-18. The age-related features, including the recommended age-specific Mn dietary intake, age-specific water consumption rates, and age-specific homeostasis of Mn, are based on the available human data and knowledge of the biology of essential-metal homeostasis. Model simulations suggest that the impact of adding drinking-water exposure to daily Mn exposure via dietary intake and ambient air inhalation in children is not greater than the impacts in adults, even at a drinking-water concentration that is 2 times higher than the USEPA's lifetime health advisory value. This conclusion was also valid for formula-fed infants who are considered at the highest potential exposure to Mn from drinking water compared to all other age groups. Our multi-route, multi-source Mn PBPK model for infants and children provides insights about the potential for Mn-related health effects on growing children and will thereby improve the level of confidence in properly interpreting Mn exposure-health effects relationships in children in human epidemiological studies.


Asunto(s)
Exposición Dietética/análisis , Agua Potable , Manganeso/farmacocinética , Modelos Biológicos , Contaminantes Químicos del Agua/farmacocinética , Adolescente , Niño , Preescolar , Femenino , Humanos , Lactante , Fórmulas Infantiles , Recién Nacido , Masculino , Leche Humana
4.
Environ Int ; 125: 291-299, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30735960

RESUMEN

An association between serum concentrations of persistent organic pollutants (POPs), such as 2,2',4,4',5,5'-hexachlorobiphenyl (PCB-153), and risk of type 2 diabetes mellitus (T2DM) has been reported. Conditional on body mass index (BMI) and waist circumference (WC), a higher serum PCB-153 concentration may be a marker of T2DM risk because it reflects other aspects of obesity that are related to T2DM risk and to PCB-153 clearance. To estimate the amount of residual confounding by other aspects of obesity, we performed a quantitative bias analysis on the results of a specific study. A physiologically-based pharmacokinetic (PBPK) model was developed to predict serum levels of PCB-153 for a simulated population. T2DM status was assigned to simulated subjects based on age, sex, BMI, WC, and visceral adipose tissue mass. The distributions of age, BMI, WC, and T2DM prevalence of the simulated population were tailored to closely match the target population. Analysis of the simulated data showed that a small part of the observed association appeared to be due to residual confounding. For example, the predicted odds ratio of T2DM that would have been obtained had the results been adjusted for visceral adipose tissue mass, for the ≥90th percentile of PCB-153 serum concentration, was 6.60 (95% CI 2.46-17.74), compared with an observed odds ratio of 7.13 (95% CI 2.65-19.13). Our results predict that the association between PCB-153 and risk of type 2 diabetes mellitus would not be substantially changed by additional adjustment for visceral adipose tissue mass in epidemiologic analyses. Confirmation of these predictions with longitudinal data would be reassuring.


Asunto(s)
Diabetes Mellitus Tipo 2/inducido químicamente , Contaminantes Ambientales/toxicidad , Bifenilos Policlorados/toxicidad , Adulto , Anciano , Sesgo , Índice de Masa Corporal , Simulación por Computador , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/epidemiología , Contaminantes Ambientales/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Obesidad/sangre , Obesidad/complicaciones , Bifenilos Policlorados/sangre , Prevalencia , Circunferencia de la Cintura , Adulto Joven
5.
Environ Mol Mutagen ; 58(8): 607-618, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28862355

RESUMEN

While insoluble nickel subsulfide (Ni3 S2 ) was carcinogenic in the lung in a 2-year rat bioassay, soluble nickel sulfate hexahydrate (NiSO4* 6H2 O) was not. To investigate whether differences in the cellular responses to these two nickel compounds could underlie their differential activities, we conducted parallel studies to determine the gene expression changes in micro-dissected lung distal airway cells from Fischer 344 rats following inhalation of the two compounds for one and four weeks (6 hr per day, 5 days per week). The results of the Ni3 S2 study have been reported previously; this paper reports the results for NiSO4 and provides a comparative analysis. The cellular responses to NiSO4 were highly similar to those previously reported for Ni3 S2 , and a set of genes was identified whose expression could be used as biomarkers for comparing cellular nickel effects from in vitro or in vivo studies with soluble NiSO4 and particulate Ni3 S2 . Evaluation of the genomic concentration-responses for the two compounds suggests that the highest inhaled concentration in the tumor bioassay for NiSO4 , which was limited by toxicity, may not have achieved the Ni concentrations at which tumors were observed in the Ni3 S2 bioassay. However, several key differences in the immune responses to NiSO4 and Ni3 S2 were identified that may result from the differential intracellular disposition of Ni from NiSO4 entering the cell as an ion rather than as a slowly soluble Ni3 S2 particle. These differences may also contribute to the observation of tumors in the bioassay for Ni3 S2 but not NiSO4 . Environ. Mol. Mutagen. 58:607-618, 2017. © 2017 The Authors Environmental and Molecular Mutagenesis published by Wiley Periodicals, Inc. on behalf of Environmental Mutagen Society.


Asunto(s)
Carcinógenos/toxicidad , Pulmón/efectos de los fármacos , Níquel/toxicidad , Administración por Inhalación , Animales , Líquido del Lavado Bronquioalveolar , Relación Dosis-Respuesta a Droga , Humanos , Inmunidad Celular/efectos de los fármacos , Pulmón/patología , Mutágenos/toxicidad , Ratas
6.
Toxicol Appl Pharmacol ; 280(1): 78-85, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24976557

RESUMEN

Male and female Fischer 344 rats were exposed to naphthalene vapors at 0 (controls), 0.1, 1, 10, and 30ppm for 6h/d, 5 d/wk, over a 90-day period. Following exposure, the respiratory epithelium and olfactory epithelium from the nasal cavity were dissected separately, RNA was isolated, and gene expression microarray analysis was conducted. Only a few significant gene expression changes were observed in the olfactory or respiratory epithelium of either gender at the lowest concentration (0.1ppm). At the 1.0ppm concentration there was limited evidence of an oxidative stress response in the respiratory epithelium, but not in the olfactory epithelium. In contrast, a large number of significantly enriched cellular pathway responses were observed in both tissues at the two highest concentrations (10 and 30ppm, which correspond to tumorigenic concentrations in the NTP bioassay). The nature of these responses supports a mode of action involving oxidative stress, inflammation and proliferation. These results are consistent with a dose-dependent transition in the mode of action for naphthalene toxicity/carcinogenicity between 1.0 and 10ppm in the rat. In the female olfactory epithelium (the gender/site with the highest incidences of neuroblastomas in the NTP bioassay), the lowest concentration at which any signaling pathway was significantly affected, as characterized by the median pathway benchmark dose (BMD) or its 95% lower bound (BMDL) was 6.0 or 3.7ppm, respectively, while the lowest female olfactory BMD values for pathways related to glutathione homeostasis, inflammation, and proliferation were 16.1, 11.1, and 8.4ppm, respectively. In the male respiratory epithelium (the gender/site with the highest incidences of adenomas in the NTP bioassay), the lowest pathway BMD and BMDL were 0.4 and 0.3ppm, respectively, and the lowest male respiratory BMD values for pathways related to glutathione homeostasis, inflammation, and proliferation were 0.5, 0.7, and 0.9ppm, respectively. Using a published physiologically based pharmacokinetic (PBPK) model to estimate target tissue dose relevant to the proposed mode of action (total naphthalene metabolism per gram nasal tissue), the lowest transcriptional BMDLs from this analysis equate to human continuous naphthalene exposure at approximately 0.3ppm. It is unlikely that significant effects of naphthalene or its metabolites will occur at exposures below this concentration.


Asunto(s)
Exposición por Inhalación , Naftalenos/administración & dosificación , Mucosa Nasal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Administración por Inhalación , Animales , Relación Dosis-Respuesta a Droga , Femenino , Exposición por Inhalación/efectos adversos , Masculino , Mucosa Nasal/patología , Mucosa Nasal/fisiología , Ratas , Ratas Endogámicas F344 , Transcripción Genética/fisiología
7.
Regul Toxicol Pharmacol ; 70(1): 203-13, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25010378

RESUMEN

ß-Chloroprene (2-chloro-1,3-butadiene, CD) is used in the manufacture of polychloroprene rubber. Chronic inhalation studies have demonstrated that CD is carcinogenic in B6C3F1 mice and Fischer 344 rats. However, epidemiological studies do not provide compelling evidence for an increased risk of mortality from total cancers of the lung. Differences between the responses observed in animals and humans may be related to differences in toxicokinetics, the metabolism and detoxification of potentially active metabolites, as well as species differences in sensitivity. The purpose of this study was to develop and apply a novel method that combines the results from available physiologically based kinetic (PBK) models for chloroprene with a statistical maximum likelihood approach to test commonality of low-dose risk across species. This method allows for the combined evaluation of human and animal cancer study results to evaluate the difference between predicted risks using both external and internal dose metrics. The method applied to mouse and human CD data supports the hypothesis that a PBK-based metric reconciles the differences in mouse and human low-dose risk estimates and further suggests that, after PBK metric exposure adjustment, humans are equally or less sensitive than mice to low levels of CD exposure.


Asunto(s)
Carcinógenos/toxicidad , Cloropreno/toxicidad , Neoplasias/inducido químicamente , Medición de Riesgo/métodos , Animales , Carcinógenos/administración & dosificación , Carcinógenos/farmacocinética , Cloropreno/administración & dosificación , Cloropreno/farmacocinética , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Funciones de Verosimilitud , Masculino , Ratones , Neoplasias/epidemiología , Ratas , Ratas Endogámicas F344 , Especificidad de la Especie
8.
Toxicol In Vitro ; 28(7): 1196-205, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24937311

RESUMEN

In the recent National Research Council report on conducting a dose-response assessment for inorganic arsenic, the committee remarked that mode of action data should be used, to the extent possible, to extrapolate below the observed range for epidemiological studies to inform the shape of the dose-response curve. Recent in vitro mode of action studies focused on understanding the development of bladder cancer following exposure to inorganic arsenic provide data to inform the dose-response curve. These in vitro data, combined with results of bladder cancer epidemiology studies, inform the dose-response curve in the low-dose region, and include values for both pharmacokinetic and pharmacodynamic variability. Integration of these data provides evidence of a range of concentrations of arsenic for which no effect on the bladder would be expected. Specifically, integration of these results suggest that arsenic exposures in the range of 7-43 ppb in drinking water are exceedingly unlikely to elicit changes leading to key events in the development of cancer or noncancer effects in bladder tissue. These findings are consistent with the lack of evidence for bladder cancer following chronic ingestion of arsenic water concentrations <100 ppb in epidemiological studies.


Asunto(s)
Arsénico/toxicidad , Carcinógenos/toxicidad , Neoplasias de la Vejiga Urinaria/inducido químicamente , Contaminantes Químicos del Agua/toxicidad , Animales , Arsénico/farmacocinética , Arsénico/normas , Carcinógenos/farmacocinética , Carcinógenos/normas , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Humanos , Ratones , Medición de Riesgo , Neoplasias de la Vejiga Urinaria/epidemiología , Neoplasias de la Vejiga Urinaria/metabolismo , Contaminantes Químicos del Agua/farmacocinética , Contaminantes Químicos del Agua/normas
9.
Toxicol Appl Pharmacol ; 279(3): 441-454, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24952340

RESUMEN

OBJECTIVE: To provide insights into the mode of action for Ni3S2 lung carcinogenicity by examining gene expression changes in target cells after inhalation exposure. METHODS: Gene expression changes were determined in micro-dissected lung broncho-alveolar cells from Fischer 344 rats following inhalation of Ni3S2 at 0.0, 0.04, 0.08, 0.15, and 0.60 mg/m(3) (0.03, 0.06, 0.11, and 0.44 mgNi/m(3)) for one and four weeks (6h/day, 5 days/week). RESULTS: Broncho-alveolar lavage fluid evaluation and lung histopathology provided evidence of inflammation only at the two highest concentrations, which were similar to those tested in the 2-year bioassay. The number of statistically significant up- and down-regulated genes decreased markedly from one to four weeks of exposure, suggesting adaptation. Cell signal pathway enrichment at both time-points primarily reflected responses to toxicity, including inflammatory and proliferative signaling. While proliferative signaling was up-regulated at both time points, some inflammatory signaling reversed from down-regulation at 1 week to up-regulation at 4 weeks. CONCLUSIONS: These results support a mode of action for Ni3S2 carcinogenicity driven by chronic toxicity, inflammation and proliferation, leading to mis-replication, rather than by direct genotoxicity. Benchmark dose (BMD) analysis identified the lowest pathway transcriptional BMD exposure concentration as 0.026 mgNi/m(3), for apoptosis/survival signaling. When conducted on the basis of lung Ni concentration the lowest pathway BMD was 0.64 µgNi/g lung, for immune/inflammatory signaling. IMPLICATIONS: These highly conservative BMDs could be used to derive a point of departure in a nonlinear risk assessment for Ni3S2 toxicity and carcinogenicity.


Asunto(s)
Carcinógenos/toxicidad , Mutágenos , Níquel/toxicidad , Animales , Apoptosis/efectos de los fármacos , Benchmarking , Peso Corporal/efectos de los fármacos , Líquido del Lavado Bronquioalveolar/citología , Carcinógenos/administración & dosificación , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/patología , Exposición por Inhalación , Pulmón/metabolismo , Pulmón/patología , Masculino , Análisis por Micromatrices , Microscopía Electrónica de Transmisión , Níquel/administración & dosificación , Níquel/metabolismo , Ratas , Ratas Endogámicas F344 , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
10.
Toxicol Sci ; 123(2): 421-32, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21795629

RESUMEN

Inorganic arsenic (As(i)) is a known human bladder carcinogen. The objective of this study was to examine the concentration dependence of the genomic response to As(i) in the urinary bladders of mice. C57BL/6J mice were exposed for 1 or 12 weeks to arsenate in drinking water at concentrations of 0.5, 2, 10, and 50 mg As/l. Urinary bladders were analyzed using gene expression microarrays. A consistent reversal was observed in the direction of gene expression change: from predominantly decreased expression at 1 week to predominantly increased expression at 12 weeks. These results are consistent with evidence from in vitro studies of an acute adaptive response that is suppressed on longer exposure due to downregulation of Fos. Pathways with the highest enrichment in gene expression changes were associated with epithelial-to-mesenchymal transition, inflammation, and proliferation. Benchmark dose (BMD) analysis determined that the lowest median BMD values for pathways were above 5 mg As/l, despite the fact that pathway enrichment was observed at the 0.5 mg As/l exposure concentration. This disparity may result from the nonmonotonic nature of the concentration-responses for the expression changes of a number of genes, as evidenced by the much fewer gene expression changes at 2 mg As/l compared with lower or higher concentrations. Pathway categories with concentration-related gene expression changes included cellular morphogenesis, inflammation, apoptosis/survival, cell cycle control, and DNA damage response. The results of this study provide evidence of a concentration-dependent transition in the mode of action for the subchronic effects of As(i) in mouse bladder cells in the vicinity of 2 mg As(i)/l.


Asunto(s)
Arseniatos/toxicidad , Carcinógenos Ambientales/toxicidad , Epitelio/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Vejiga Urinaria/efectos de los fármacos , Animales , Benchmarking , Relación Dosis-Respuesta a Droga , Ingestión de Líquidos , Epitelio/metabolismo , Epitelio/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Medición de Riesgo , Factores de Tiempo , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Abastecimiento de Agua
11.
Toxicol Appl Pharmacol ; 232(3): 448-55, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18706920

RESUMEN

The relationship of exposure and tissue concentration of parent chemical and metabolites over prolonged exposure is a critical issue for chronic toxicities mediated by metabolite(s) rather than parent chemical alone. This is an issue for AsV because its trivalent metabolites have unique toxicities and relatively greater potency compared to their pentavalent counterparts for many endpoints. In this study, dose-dependency in tissue distribution and urinary excretion for inorganic arsenic and its methylated metabolites was assessed in female C57Bl/6 mice exposed to 0, 0.5, 2, 10 or 50 ppm arsenic (as arsenate, AsV) in their drinking water for 12 weeks. No adverse effects were observed and body weight gain did not differ significantly among groups. Urinary excretion of arsenite monomethylarsonous acid (MMA(III)), dimethylarsinous acid (DMA(III)), dimethylarsinic acid (DMAV), and trimethylarsine oxide (TMAO) increased linearly with dose, whereas AsV and monomethylarsonic acid (MMAV) excretion was non-linear with respect to dose. Total tissue arsenic accumulation was greatest in kidney > lung > urinary bladder >>> skin > blood > liver. Monomethyl arsenic (MMA, i.e. MMA(III)+MMAV) was the predominant metabolite in kidney, whereas dimethylarsenic (DMA, i.e., DMA(III)+DMAV) was the predominant metabolite in lung. Urinary bladder tissue had roughly equivalent levels of inorganic arsenic and dimethylarsenic, as did skin. These data indicate that pharmacokinetic models for arsenic metabolism and disposition need to include mechanisms for organ-specific accumulation of some arsenicals and that urinary metabolite profiles are not necessarily reflective of target tissue dosimetry.


Asunto(s)
Arseniatos/farmacocinética , Arsénico/orina , Animales , Arsenicales/orina , Ácido Cacodílico/análogos & derivados , Ácido Cacodílico/orina , Relación Dosis-Respuesta a Droga , Femenino , Ratones , Ratones Endogámicos C57BL , Distribución Tisular
12.
Toxicol Sci ; 85(1): 429-46, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15703268

RESUMEN

The metabolic series approach for risk assessment uses a dosimetry-based analysis to develop toxicity information for a group of metabolically linked compounds using pharmacokinetic (PK) data for each compound and toxicity data for the parent compound. The metabolic series approach for n-butyl acetate and its subsequent metabolites, n-butanol and n-butyric acid (the butyl series), was first demonstrated using a provisional physiologically based pharmacokinetic (PBPK) model for the butyl series. The objective of this work was to complete development of the PBPK model for the butyl series. Rats were administered test compounds by iv bolus dose, iv infusion, or by inhalation in a recirculating closed chamber. Hepatic, vascular, and extravascular metabolic constants for metabolism were estimated by fitting the model to the blood time course data from these experiments. The respiratory bioavailability of n-butyl acetate (100% of alveolar ventilation) and n-butanol (50% of alveolar ventilation) was estimated from closed chamber inhalation studies and measured ventilation rates. The resulting butyl series PBPK model successfully reproduces the blood time course of these compounds following iv administration and inhalation exposure to n-butyl acetate and n-butanol in rats and arterial blood n-butanol kinetics following inhalation exposure to n-butanol in humans. These validated inhalation route models can be used to support species and dose-route extrapolations required for risk assessment of butyl series family of compounds. Human equivalent concentrations of 169 ppm and 1066 ppm n-butanol corresponding to the rat n-butyl acetate NOAELs of 500 and 3000 ppm were derived using the models.


Asunto(s)
1-Butanol/farmacocinética , Acetatos/farmacocinética , Ácido Butírico/farmacocinética , Modelos Biológicos , 1-Butanol/sangre , Acetatos/sangre , Administración por Inhalación , Animales , Ácido Butírico/sangre , Humanos , Infusiones Intravenosas , Inyecciones Intravenosas , Masculino , Ratas , Ratas Sprague-Dawley , Medición de Riesgo , Distribución Tisular
13.
Regul Toxicol Pharmacol ; 35(2 Pt 1): 177-97, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12052003

RESUMEN

Increasing sophistication in methods used to account for human variability in susceptibility to toxicants has been one of the success stories in the continuing evolution of risk assessment science. Genetic polymorphisms have been suggested as an important contributor to overall human variability. Recently, data on polymorphisms in metabolic enzymes have been integrated with physiologically based pharmacokinetic (PBPK) modeling as an approach to determining the resulting overall variability. We present an analysis of the potential contribution of polymorphisms in enzymes modulating the disposition of four diverse compounds: methylene chloride, warfarin, parathion, and dichloroacetic acid. Through these case studies, we identify key uncertainties likely to be encountered in the use of polymorphism data and highlight potential simplifying assumptions that might be required to test the hypothesis that genetic factors are a substantive source of human variability in susceptibility to environmental toxicants. These uncertainties include (1) the relative contribution of multiple enzyme systems, (2) the extent of induction/inhibition through coexposure, (3) allelic frequencies of major ethnic groups, (4) the absence of chemical-specific data on the kinetic parameters for the different allelic forms of key enzymes, (5) large numbers of low-frequency alleles, and (6) uncertainty regarding differences between in vitro and in vivo kinetic data. Our effort sets the stage for the acquisition of critical data and further integration of polymorphism data with PBPK modeling as a means to quantitate population variability.


Asunto(s)
Enzimas/genética , Polimorfismo Genético , Medición de Riesgo/métodos , Xenobióticos/farmacocinética , Animales , Ácido Dicloroacético/farmacocinética , Relación Dosis-Respuesta a Droga , Enzimas/metabolismo , Humanos , Técnicas In Vitro , Cloruro de Metileno/farmacocinética , Paratión/farmacocinética , Reproducibilidad de los Resultados , Incertidumbre , Warfarina/farmacocinética
14.
Toxicol Sci ; 63(2): 160-72, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11568359

RESUMEN

A physiologically based pharmacokinetic (PBPK) model for isopropanol (IPA) and its major metabolite, acetone, is described. The structure of the parent chemical model, which can be used for either IPA or acetone by choosing the appropriate chemical-specific parameters, is similar to previously published models of volatile organic chemicals such as styrene. However, in order to properly simulate data on the exhalation of IPA and acetone during inhalation exposures, it was necessary to expand the description of the lung compartment to include a subcompartment for the upper respiratory tract mucus layer. This elaboration is consistent with published PBPK models of other water-soluble vapors in which the mucus layer serves to absorb the chemical during inhalation and then release it during exhalation. In the case of IPA exposure, a similar PBPK structure is used to describe the kinetics of the acetone produced from the metabolism of IPA. The resulting model is able to provide a coherent description of IPA and acetone kinetics in the rat and human for exposures to IPA by several routes: intravenous, intraperitoneal, oral, inhalation, and dermal. It is also able to consistently reproduce kinetic data for exposures of rats or humans to acetone. Thus, the model provides a validated framework for performing chemical-specific route-to-route extrapolation and cross-species dosimetry, which can be used in place of generic default calculations in support of risk assessments for IPA and acetone.


Asunto(s)
2-Propanol/farmacocinética , Acetona/farmacocinética , Encéfalo/efectos de los fármacos , Tasa de Depuración Metabólica/efectos de los fármacos , Modelos Biológicos , 2-Propanol/metabolismo , Absorción , Administración Oral , Animales , Encéfalo/metabolismo , Simulación por Computador , Relación Dosis-Respuesta a Droga , Humanos , Exposición por Inhalación , Cinética , Hígado/metabolismo , Pulmón/metabolismo , Matemática , Permeabilidad , Planificación de la Radioterapia Asistida por Computador , Ratas , Sistema Respiratorio/metabolismo , Solubilidad , Distribución Tisular , Agua
15.
Sci Total Environ ; 274(1-3): 37-66, 2001 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-11453305

RESUMEN

Vinyl chloride (VC) is a trans-species carcinogen, producing tumors in a variety of tissues, from both inhalation and oral exposures, across a number of species. In particular, exposure to VC has been associated with a rare tumor, liver angiosarcoma, in a large number of studies in mice, rats, and humans. The mode of action for the carcinogenicity of VC appears to be a relatively straightforward example of DNA adduct formation by a reactive metabolite, leading to mutation, mistranscription, and neoplasia. The objective of the present analysis was to investigate the comparative potency of a classic genotoxic carcinogen across species, by performing a quantitative comparison of the carcinogenic potency of VC using data from inhalation and oral rodent bioassays as well as from human epidemiological studies. A physiologically-based pharmacokinetic (PBPK) model for VC was developed to support the target tissue dosimetry for the cancer risk assessment. Unlike previous models, the initial metabolism of VC was described as occurring via two saturable pathways, one representing low capacity-high affinity oxidation by CYP2E1 and the other (in the rodent) representing higher capacity-lower affinity oxidation by other isozymes of P450, producing in both cases chloroethylene oxide (CEO) and chloroacetaldehyde (CAA) as intermediate reactive products. Depletion of glutathione by reaction with CEO and CAA was also described. Animal-based risk estimates for human inhalation exposure to VC using total metabolism estimates from the PBPK model were consistent with risk estimates based on human epidemiological data, and were lower than those currently used in environmental decision-making by a factor of 80.


Asunto(s)
Carcinógenos/toxicidad , Neoplasias/inducido químicamente , Neoplasias/epidemiología , Cloruro de Vinilo/farmacocinética , Cloruro de Vinilo/toxicidad , Contaminantes Atmosféricos/farmacocinética , Contaminantes Atmosféricos/toxicidad , Animales , Carcinógenos/farmacocinética , Aductos de ADN , Humanos , Ratones , Modelos Biológicos , Modelos Estadísticos , Método de Montecarlo , Mutagénesis , Ratas , Medición de Riesgo , Factores de Riesgo , Transcripción Genética
16.
Environ Health Perspect ; 108 Suppl 2: 283-305, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10807559

RESUMEN

A physiologically based pharmacokinetic (PBPK) model was developed that provides a comprehensive description of the kinetics of trichloroethylene (TCE) and its metabolites, trichloroethanol (TCOH), trichloroacetic acid (TCA), and dichloroacetic acid (DCA), in the mouse, rat, and human for both oral and inhalation exposure. The model includes descriptions of the three principal target tissues for cancer identified in animal bioassays: liver, lung, and kidney. Cancer dose metrics provided in the model include the area under the concentration curve (AUC) for TCA and DCA in the plasma, the peak concentration and AUC for chloral in the tracheobronchial region of the lung, and the production of a thioacetylating intermediate from dichlorovinylcysteine in the kidney. Additional dose metrics provided for noncancer risk assessment include the peak concentrations and AUCs for TCE and TCOH in the blood, as well as the total metabolism of TCE divided by the body weight. Sensitivity and uncertainty analyses were performed on the model to evaluate its suitability for use in a pharmacokinetic risk assessment for TCE. Model predictions of TCE, TCA, DCA, and TCOH concentrations in rodents and humans are in good agreement with a variety of experimental data, suggesting that the model should provide a useful basis for evaluating cross-species differences in pharmacokinetics for these chemicals. In the case of the lung and kidney target tissues, however, only limited data are available for establishing cross-species pharmacokinetics. As a result, PBPK model calculations of target tissue dose for lung and kidney should be used with caution.


Asunto(s)
Carcinógenos Ambientales/farmacocinética , Modelos Biológicos , Medición de Riesgo , Tricloroetileno/farmacocinética , Animales , Carcinógenos Ambientales/metabolismo , Humanos , Sensibilidad y Especificidad , Tricloroetileno/metabolismo
17.
Environ Health Perspect ; 108 Suppl 2: 323-34, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10807562

RESUMEN

Alternatives for developing chronic exposure limits for noncancer effects of trichloroethylene (TCE) were evaluated. These alternatives were organized within a framework for dose-response assessment--exposure:dosimetry (pharmacokinetics):mode of action (pharmacodynamics): response. This framework provides a consistent structure within which to make scientific judgments about available information, its interpretation, and use. These judgments occur in the selection of critical studies, internal dose metrics, pharmacokinetic models, approaches for interspecies extrapolation of pharmacodynamics, and uncertainty factors. Potentially limiting end points included developmental eye malformations, liver effects, immunotoxicity, and kidney toxicity from oral exposure and neurological, liver, and kidney effects by inhalation. Each end point was evaluated quantitatively using several methods. Default analyses used the traditional no-observed adverse effect level divided by uncertainty factors and the benchmark dose divided by uncertainty factors methods. Subsequently, mode-of-action and pharmacokinetic information were incorporated. Internal dose metrics were estimated using a physiologically based pharmacokinetic (PBPK) model for TCE and its major metabolites. This approach was notably useful with neurological and kidney toxicities. The human PBPK model provided estimates of human exposure doses for the internal dose metrics. Pharmacodynamic data or default assumptions were used for interspecies extrapolation. For liver and neurological effects, humans appear no more sensitive than rodents when internal dose metrics were considered. Therefore, the interspecies uncertainty factor was reduced, illustrating that uncertainty factors are a semiquantitative approach fitting into the organizational framework. Incorporation of pharmacokinetics and pharmacodynamics can result in values that differ significantly from those obtained with the default methods.


Asunto(s)
Sustancias Peligrosas/efectos adversos , Tricloroetileno/efectos adversos , Administración por Inhalación , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Anomalías del Ojo/inducido químicamente , Sustancias Peligrosas/administración & dosificación , Humanos , Riñón/efectos de los fármacos , Sistema Nervioso/efectos de los fármacos , Medición de Riesgo , Tricloroetileno/administración & dosificación
18.
Toxicol Ind Health ; 16(9-10): 335-438, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11762928

RESUMEN

Environmental risk-management decisions in the U.S. involving potential exposures to methylmercury currently use a reference dose (RfD) developed by the U.S. Environmental Protection Agency (USEPA). This RfD is based on retrospective studies of an acute poisoning incident in Iraq in which grain contaminated with a methylmercury fungicide was inadvertently used in the baking of bread. The exposures, which were relatively high but lasted only a few months, were associated with neurological effects in both adults (primarily paresthesia) and infants (late walking, late talking, etc.). It is generally believed that the developing fetus represents a particularly sensitive subpopulation for the neurological effects of methylmercury. The USEPA derived an RfD of 0.1 microg/kg/day based on benchmark dose (BMD) modeling of the combined neurological endpoints reported for children exposed in utero. This RfD included an uncertainty factor of 10 to consider human pharmacokinetic variability and database limitations (lack of data on multigeneration effects or possible long-term sequelae of perinatal exposure). Alcoa signed an Administrative Order of Consent for the conduct of a remedial investigation/feasibility study (RI/FS) at their Point Comfort Operations and the adjacent Lavaca Bay in Texas to address the effects of historical discharges of mercury-containing wastewater. In cooperation with the Texas Natural Resource Conservation Commission and USEPA Region VI, Alcoa conducted a baseline risk assessment to assess potential risk to human health and the environment. As a part of this assessment. Alcoa pursued the development of a site-specific RfD for methylmercury to specifically address the potential human health effects associated with the ingestion of contaminated finfish and shellfish from Lavaca Bay. Application of the published USEPA RfD to this site is problematic; while the study underlying the RfD represented acute exposure to relatively high concentrations of methylmercury, the exposures of concern for the Point Comfort site are from the chronic consumption of relatively low concentrations of methylmercury in fish. Since the publication of the USEPA RfD, several analyses of chronic exposure to methylmercury in fish-eating populations have been reported. The purpose of the analysis reported here was to evaluate the possibility of deriving an RfD for methylmercury, specifically for the case of fish ingestion, on the basis of these new studies. In order to better support the risk-management decisions associated with developing a remediation approach for the site in question, the analysis was designed to provide information on the distribution of acceptable ingestion rates across a population, which could reasonably be expected to be consistent with the results of the epidemiological studies of other fish-eating populations. Based on a review of the available literature on the effects of methylmercury, a study conducted with a population in the Seychelles Islands was selected as the critical study for this analysis. The exposures to methylmercury in this population result from chronic, multigenerational ingestion of contaminated fish. This prospective study was carefully conducted and analyzed, included a large cohort of mother-infant pairs, and was relatively free of confounding factors. The results of this study are essentially negative, and a no-observed-adverse-effect level (NOAEL) derived from the estimated exposures has recently been used by the Agency for Toxic Substances and Disease Registry (ATSDR) as the basis for a chronic oral minimal risk level (MRL) for methylmercury. In spite of the fact that no statistically significant effects were observed in this study, the data as reported are suitable for dose-response analysis using the BMD method. Evaluation of the BMD method used in this analysis, as well as in the current USEPA RfD, has demonstrated that the resulting 95% lower bound on the 10% benchmark dose (BMDL) represents a conservative estimate of the traditional NOAEL, and that it is superior to the use of "average" or "grouped" exposure estimates when dose-response information is available, as is the case for the Seychelles study. A more recent study in the Faroe Islands, which did report statistically significant associations between methylmercury exposure and neurological effects, could not be used for dose-response modeling due to inadequate reporting of the data and confounding from co-exposure to polychlorinated biphenyls (PCBs). BMD modeling over the wide range of neurological endpoints reported in the Seychelles study yielded a lowest BMDL for methylmercury in maternal hair of 21 ppm. This BMDL was then converted to an expected distribution of daily ingestion rates across a population using Monte Carlo analysis with a physiologically based pharmacokinetic (PBPK) model to evaluate the impact of interindividual variability. The resulting distribution of ingestion rates at the BMDL had a geometric mean of 1.60 microg/kg/day with a geometric standard deviation of 1.33; the 1st, 5th, and 10th percentiles of the distribution were 0.86, 1.04, and 1.15 microg/kg/day. In place of the use of an uncertainty factor of 3 for pharmacokinetic variability, as is done in the current RfD, one of these lower percentiles of the daily ingestion rate distribution provides a scientifically based, conservative basis for taking into consideration the impact of pharmacokinetic variability across the population. On the other hand, it was felt that an uncertainty factor of 3 for database limitations should be used in the current analysis. Although there can be high confidence in the benchmark-estimated NOAEL of 21 ppm in the Seychelles study, some results in the New Zealand and Faroe Islands studies could be construed to suggest the possibility of effects at maternal hair concentrations below 10 ppm. In addition, while concerns regarding the possibility of chronic sequelae are not supported by the available data, neither can they be absolutely ruled out. The use of an uncertainty factor of 3 is equivalent to using a NOAEL of 7 ppm in maternal hair, which provides additional protection against the possibility that effects could occur at lower concentrations in some populations. Based on the analysis described above, the distribution of acceptable daily ingestion rates (RfDs) recommended to serve as the basis for site-specific risk-management decisions at Alcoa's Point Comfort Operations ranges from approximately 0.3 to 1.1 microg/kg/day, with a population median (50th percentile) of 0.5 microg/kg/day. By analogy with USEPA guidelines for the use of percentiles in applications of distributions in exposure assessments, the 10th percentile provides a reasonably conservative measure. On this basis, a site-specific RfD of 0.4 microg/kg/day is recommended.


Asunto(s)
Benchmarking , Exposición a Riesgos Ambientales , Peces , Contaminación de Alimentos , Compuestos de Metilmercurio/análisis , Modelos Teóricos , Contaminantes Químicos del Agua/análisis , Adulto , Animales , Estudios de Cohortes , Femenino , Geografía , Humanos , Recién Nacido , Masculino , Intercambio Materno-Fetal , Compuestos de Metilmercurio/efectos adversos , Compuestos de Metilmercurio/farmacocinética , Nivel sin Efectos Adversos Observados , Embarazo , Salud Pública , Valores de Referencia , Medición de Riesgo , Contaminantes Químicos del Agua/efectos adversos , Contaminantes Químicos del Agua/farmacocinética
19.
Neurotoxicology ; 20(2-3): 161-71, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10385880

RESUMEN

Manganese (Mn)-deficiency or Mn-excess can lead to adverse biological consequences. Central nervous system tissues, rich in dopaminergic neurons, are the targets whether the Mn gains entrance by inhalation, oral ingestion, or intravenous administration. Risk assessments with Mn need to ensure that brain concentrations in the globus pallidus and striatum stay within the range of normal. This paper first provides a critical review of the biological factors that determine the disposition of Mn in tissues within the body. Secondly, it outlines specific data needs for developing a physiologically based pharmacokinetic (PBPK) model for Mn to assist in conducting risk assessments for inhaled and ingested Mn. Uptake of dietary Mn appears to be controlled by several dose-dependent processes: biliary excretion, intestinal absorption, and intestinal elimination. Mn absorbed in the divalent form from the gut via the portal blood is complexed with plasma proteins that are efficiently removed by the liver. Absorption of Mn via inhalation, intratracheal instillation or intravenous infusions bypasses the control processes in the gastrointestinal tract. After absorption into the blood system by these alternate routes, Mn is apparently oxidized by ceruloplasmin and the trivalent Mn binds to the iron carrying protein, transferrin. Brain uptake of Mn occurs via transferrin receptors located in various brain regions. Transferrin-bound trivalent Mn is not as readily removed by the liver, as are protein complexes with divalent Mn. Thus, Mn delivered by these other dose routes would be available for uptake into tissues for a longer period of time than the orally administered Mn, leading to quantitative differences in tissue uptake for different dose routes. Several important data gaps impede organizing these various physiological factors into a multi-dose route PK model for Mn. They include knowledge of (1) oxidation rates of Mn in blood, (2) uptake rates of protein-bound forms of Mn by the liver, (3) neuronal transfer rates within the CNS, and (4) quantitative analyses of the control processes that regulate uptake of ingested Mn by the intestines and liver. These data gaps are the main obstacles to developing a risk assessment strategy for Mn that considers contributions of both inhalation and ingestion of this essential nutrient in determining brain Mn concentrations.


Asunto(s)
Exposición a Riesgos Ambientales/efectos adversos , Exposición por Inhalación/efectos adversos , Manganeso/farmacocinética , Administración Oral , Animales , Humanos , Medición de Riesgo/métodos , Distribución Tisular
20.
Risk Anal ; 19(4): 547-58, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10765421

RESUMEN

An analysis of the uncertainty in guidelines for the ingestion of methylmercury (MeHg) due to human pharmacokinetic variability was conducted using a physiologically based pharmacokinetic (PBPK) model that describes MeHg kinetics in the pregnant human and fetus. Two alternative derivations of an ingestion guideline for MeHg were considered: the U.S. Environmental Protection Agency reference dose (RfD) of 0.1 microgram/kg/day derived from studies of an Iraqi grain poisoning episode, and the Agency for Toxic Substances and Disease Registry chronic oral minimal risk level (MRL) of 0.5 microgram/kg/day based on studies of a fish-eating population in the Seychelles Islands. Calculation of an ingestion guideline for MeHg from either of these epidemiological studies requires calculation of a dose conversion factor (DCF) relating a hair mercury concentration to a chronic MeHg ingestion rate. To evaluate the uncertainty in this DCF across the population of U.S. women of child-bearing age, Monte Carlo analyses were performed in which distributions for each of the parameters in the PBPK model were randomly sampled 1000 times. The 1st and 5th percentiles of the resulting distribution of DCFs were a factor of 1.8 and 1.5 below the median, respectively. This estimate of variability is consistent with, but somewhat less than, previous analyses performed with empirical, one-compartment pharmacokinetic models. The use of a consistent factor in both guidelines of 1.5 for pharmacokinetic variability in the DCF, and keeping all other aspects of the derivations unchanged, would result in an RfD of 0.2 microgram/kg/day and an MRL of 0.3 microgram/kg/day.


Asunto(s)
Compuestos de Metilmercurio/administración & dosificación , Compuestos de Metilmercurio/farmacocinética , Adulto , Femenino , Cabello/metabolismo , Humanos , Masculino , Compuestos de Metilmercurio/envenenamiento , Modelos Biológicos , Método de Montecarlo , Nivel sin Efectos Adversos Observados , Embarazo , Medición de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...