Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Regul Toxicol Pharmacol ; 81: 468-479, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27756559

RESUMEN

To help develop a comprehensive, quantitative understanding of the hazards of 1,2-dichloroethane (ethylene dichloride, EDC, CAS No. 107-06-2) exposure by the inhalation route, the results of existing subchronic studies and an extended one-generation reproductive toxicity (EOGRT) study recently conducted by the oral route in rats were extrapolated using a physiologically based pharmacokinetic (PBPK) model. The no observed adverse effects levels (NOAELs) for the endpoints of neurotoxicity and reproductive/developmental toxicity were the highest tested doses of 169 and 155 mg/kg-day, respectively. These NOAELs were equivalent to continuous exposure of rats to minimums of 76 ppm and 62 ppm EDC, respectively, using total metabolism of EDC as the dose metric that is equivalent in the oral and inhalation scenarios. In contrast, the subchronic study NOAEL of 37.5 mg/kg-day corresponded to continuous inhalation of 4.4 ppm EDC, based on equivalent extrahepatic metabolism. The selection of the internal metric which serves to establish route-to-route equivalency was found to profoundly influence the NOAEL-equivalent inhalation exposure concentration and thus will be a key determinant of inhalation toxicity reference criteria developed on the basis of EDC studies conducted by the oral route.


Asunto(s)
Dicloruros de Etileno/administración & dosificación , Dicloruros de Etileno/farmacocinética , Administración por Inhalación , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Dicloruros de Etileno/toxicidad , Femenino , Cinética , Masculino , Ratas , Ratas Sprague-Dawley , Reproducción/efectos de los fármacos
2.
Crit Rev Toxicol ; 45(8): 662-726, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25997510

RESUMEN

Potential chronic health risks for children and prospective parents exposed to ethylbenzene were evaluated in response to the Voluntary Children's Chemical Evaluation Program. Ethylbenzene exposure was found to be predominately via inhalation with recent data demonstrating continuing decreases in releases and both outdoor and indoor concentrations over the past several decades. The proportion of ethylbenzene in ambient air that is attributable to the ethylbenzene/styrene chain of commerce appears to be relatively very small, less than 0.1% based on recent relative emission estimates. Toxicity reference values were derived from the available data, with physiologically based pharmacokinetic models and benchmark dose methods used to assess dose-response relationships. An inhalation non-cancer reference concentration or RfC of 0.3 parts per million (ppm) was derived based on ototoxicity. Similarly, an oral non-cancer reference dose or RfD of 0.5 mg/kg body weight/day was derived based on liver effects. For the cancer assessment, emphasis was placed upon mode of action information. Three of four rodent tumor types were determined not to be relevant to human health. A cancer reference value of 0.48 ppm was derived based on mouse lung tumors. The risk characterization for ethylbenzene indicated that even the most highly exposed children and prospective parents are not at risk for non-cancer or cancer effects of ethylbenzene.


Asunto(s)
Derivados del Benceno/toxicidad , Exposición a Riesgos Ambientales/efectos adversos , Exposición por Inhalación/efectos adversos , Animales , Derivados del Benceno/administración & dosificación , Niño , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Monitoreo del Ambiente/métodos , Humanos , Padres , Valores de Referencia , Medición de Riesgo/métodos
3.
Inhal Toxicol ; 27(2): 83-99, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25600219

RESUMEN

The inhalation toxicity of submarine contaminants is of concern to ensure the health of men and women aboard submarines during operational deployments. Due to a lack of adequate prior studies, potential general, neurobehavioral, reproductive and developmental toxicity was evaluated in male and female rats exposed to mixtures of three critical submarine atmospheric components: carbon monoxide (CO) and carbon dioxide (CO2; levels elevated above ambient), and oxygen (O2; levels decreased below ambient). In a 14-day, 23 h/day, whole-body inhalation study of exposure to clean air (0.4 ppm CO, 0.1% CO2 and 20.6% O2), low-dose, mid-dose and high-dose gas mixtures (high dose of 88.4 ppm CO, 2.5% CO2 and 15.0% O2), no adverse effects on survival, body weight or histopathology were observed. Reproductive, developmental and neurobehavioral performance were evaluated after a 28-day exposure in similar atmospheres. No adverse effects on estrus phase, mating, gestation or parturition were observed. No developmental or functional deficits were observed in either exposed parents or offspring related to motor activity, exploratory behavior or higher-level cognitive functions (learning and memory). Only minimal effects were discovered in parent-offspring emotionality tests. While statistically significant increases in hematological parameters were observed in the offspring of exposed parents compared to controls, these parameters remained within normal clinical ranges for blood cells and components and were not considered adverse. In summary, subacute exposures to elevated concentrations of the submarine atmosphere gases did not affect the ability of rats to reproduce and did not appear to have any significant adverse health effects.


Asunto(s)
Dióxido de Carbono/efectos adversos , Monóxido de Carbono/efectos adversos , Oxígeno/análisis , Reproducción , Medicina Submarina , Pruebas de Toxicidad Subaguda/métodos , Administración por Inhalación , Aire , Animales , Conducta Animal , Peso Corporal , Dióxido de Carbono/análisis , Monóxido de Carbono/análisis , Conducta Exploratoria , Femenino , Masculino , Nivel sin Efectos Adversos Observados , Ratas , Ratas Sprague-Dawley , Valores de Referencia
4.
Inhal Toxicol ; 27(3): 121-37, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25687554

RESUMEN

Carbon monoxide (CO), carbon dioxide (CO2) and low-level oxygen (O2) (hypoxia) are submarine atmosphere components of highest concern because of a lack of toxicological data available to address the potential effects from long-duration, combined exposures on female reproductive and developmental health. In this study, subchronic toxicity of mixed atmospheres of these three submarine air components was evaluated in rats. Male and female rats were exposed via inhalation to clean air (0.4 ppm CO; 0.13% CO2; 20.6% O2) (control), a low-dose (5.0 ppm CO; 0.41% CO2; 17.1% O2), a mid-dose (13.9 ppm CO; 1.19 or 1.20% CO2; 16.1% O2) and a high-dose (89.9 ppm CO; 2.5% CO2; 15.0% O2) gas mixture for 23 h per day for 70 d premating and a 14-d mating period. Impregnated dams continued exposure to gestation day 19. Adverse reproductive effects were not identified in exposed parents (P0) or first (F1) and second generation (F2) offspring during mating, gestation or parturition. No adverse changes to the estrous cycle or in reproductive hormone concentrations were identified. The exposure-related effects were reduced weight gains and adaptive up-regulation of erythropoiesis in male rats from the high-dose group. No adverse, dose-related health effects on clinical data or physiological data were observed. Neurobehavioral tests identified no apparent developmental deficits at the tested levels of exposure. In summary, subchronic exposures to the submarine atmosphere gases did not affect the ability of the exposed rats or their offspring to reproduce and did not appear to have any significant adverse health effects.


Asunto(s)
Aire , Conducta Animal/efectos de los fármacos , Dióxido de Carbono/toxicidad , Monóxido de Carbono/toxicidad , Reproducción/efectos de los fármacos , Medicina Submarina , Administración por Inhalación , Aire/análisis , Aire/normas , Animales , Peso Corporal , Dióxido de Carbono/administración & dosificación , Monóxido de Carbono/administración & dosificación , Femenino , Masculino , Ratas
5.
Regul Toxicol Pharmacol ; 64(2): 205-24, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22841928

RESUMEN

Hexahydro-1,3,5-trinitro-1,3,5-triazine (RDX, CAS No. 121-82-4) is a component of munitions formulations, and has been detected in groundwater samples collected at various US military sites. Clean up target levels for RDX may be derived based on consideration of acceptable cumulative human exposure as expressed in toxicity reference values. Evaluations of the cancer weight of evidence and possible modes of action (MOA) for RDX-induced cancer were conducted. It was concluded that the available data provide suggestive evidence of human carcinogenic potential for RDX. While a mutagenic/genotoxic MOA for RDX is unlikely, no alterative MOA is strongly supported by the available data. A nonlinear (threshold) approach to the assessment of human cancer risk was recommended, and a recommended chronic cancer reference dose of 0.08mg/kg/day was derived. For comparison only, computations using a linear approach were also conducted, yielding a cancer risk specific dose of 0.000235mg/kg/day for 1 in 10(5) risk; this value is 2.6-fold higher the current US EPA risk specific dose for 1 in 10(5) risk. Thus, cleanup standards based on human health risk from RDX exposure could potentially depend on the willingness of risk managers to accept a nonlinear MOA and nonlinear toxicity risk value derivation.


Asunto(s)
Carcinógenos/toxicidad , Contaminantes Ambientales/toxicidad , Sustancias Explosivas/toxicidad , Neoplasias Hepáticas/inducido químicamente , Triazinas/toxicidad , Animales , Carcinógenos/farmacocinética , Contaminantes Ambientales/farmacocinética , Sustancias Explosivas/farmacocinética , Femenino , Masculino , Ratones , Pruebas de Mutagenicidad , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Valores de Referencia , Medición de Riesgo , Triazinas/farmacocinética
6.
Regul Toxicol Pharmacol ; 62(1): 107-14, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22197625

RESUMEN

RDX (hexahydro-1,3,5-trinitro-1,3,5-triazine) is an explosive used in military applications. It has been detected in ground water surrounding US military installations and at manufacturing facilities. RDX has been shown to produce hepatotoxicity, testicular, and neurological effects in animals, the latter also in humans. The current chronic oral reference dose (RfD) of 0.003 mg/kg/day was derived based on prostate effects in rats. Here, we provide a reevaluation of the risk associated with RDX exposure by examining old and new data and using physiologically based pharmacokinetic (PBPK) modeling approaches. Candidate non-cancer endpoints in rodents were evaluated and the most plausible mode(s) of action were determined. A PBPK model was used to derive appropriate internal doses based on the mode of action, and then a benchmark dose (BMD) and the lower confidence limit on the BMD (BMDL) were determined using these internal doses in animals. Uncertainty factors (UF) were applied to the animal BMDL or no-observed effect level and a human PBPK model was used to determine a human equivalent dose resulting in the candidate RfDs (cRfDs). A proposed chronic RfD of 0.07 mg/kg/day, based on multiple effects observed in rats, was selected from among the cRfDs.


Asunto(s)
Sustancias Explosivas/toxicidad , Modelos Biológicos , Medición de Riesgo/métodos , Triazinas/toxicidad , Animales , Sustancias Explosivas/farmacocinética , Humanos , Triazinas/farmacocinética
7.
Crit Rev Toxicol ; 40 Suppl 1: 74-92, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20868268

RESUMEN

1,3-Butadiene (BD) is a multisite carcinogen in laboratory rodents following lifetime exposure, with greater potency in the mouse than the rat, and is associated with an increase in leukemia mortality in highly exposed workers. Species differences in the formation of reactive metabolites underlie observed species differences in sensitivity to the carcinogenic effects of BD. The modes of action (MOAs) for human leukemia and rodent tumors are both likely related to mutagenic potencies of one or more of these metabolites. However, differences in the nature of genotoxic lesions associated with human leukemia and rodent tumors, along with their implications for risk assessment, require that they be discussed separately. The MOAs for BD are assessed in this review using the modified Hill criteria and human relevance framework. Key events in MOAs for human and rodent cancers are identified, along with important species differences and sources of nonlinearity for each event that can affect extrapolations made from high- to low-dose exposures. Because occupational exposures to BD have also included co-exposures to styrene and dimethyldithiocarbamide (DMDTC), potential interactions with BD carcinogenicity are also discussed. The MOAs for BD carcinogenesis will be used to guide key decisions made in the quantitative cancer dose-response assessment.


Asunto(s)
Butadienos/metabolismo , Butadienos/toxicidad , Leucemia/inducido químicamente , Mutágenos/metabolismo , Mutágenos/toxicidad , Animales , Pruebas de Carcinogenicidad , Humanos , Masculino , Ratones , Exposición Profesional , Ratas , Medición de Riesgo
8.
Crit Rev Toxicol ; 40 Suppl 1: 1-11, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20868266

RESUMEN

1,3-Butadiene (BD) is a multisite carcinogen in laboratory rodents following lifetime exposure, with mice demonstrating greater sensitivity than rats. In epidemiology studies of men in the styrene-butadiene rubber industry, leukemia mortality is associated with butadiene exposure, and this association is most pronounced for high-intensity BD exposures. Metabolism is an important determinant of BD carcinogenicity. BD is metabolized to several electrophilic intermediates, including epoxybutene (EB), diepoxybutane (DEB), and epoxybutane diol (EBD), which differ considerably in their genotoxic potency (DEB >> EB > EBD). Important species differences exist with respect to the formation of reactive metabolites and their subsequent detoxification, which underlie observed species differences in sensitivity to the carcinogenic effects of BD. The modes of action for human leukemia and for the observed solid tumors in rodents are both likely related to the genotoxic potencies for one or more of these metabolites. A number of factors related to metabolism can also contribute to nonlinearity in the dose-response relationship, including enzyme induction and inhibition, depletion of tissue glutathione, and saturation of oxidative metabolism. A quantitative risk assessment of BD needs to reflect these species differences and sources of nonlinearity if it is to reflect the current understanding of the disposition of BD.


Asunto(s)
Butadienos/metabolismo , Butadienos/toxicidad , Elastómeros/toxicidad , Estudios Epidemiológicos , Estirenos/toxicidad , Animales , Pruebas de Carcinogenicidad , Elastómeros/metabolismo , Compuestos Epoxi/metabolismo , Compuestos Epoxi/toxicidad , Femenino , Humanos , Leucemia/inducido químicamente , Masculino , Ratones , Salud Laboral , Ratas , Medición de Riesgo , Estirenos/metabolismo
9.
Crit Rev Toxicol ; 40 Suppl 1: 12-73, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20868267

RESUMEN

1,3-Butadiene’s (BD’s) major electrophilic metabolites 1,2-epoxy-3-butene (EB), 1,2-dihydroxy-3,4-epoxybutane (EBD), and 1,2,3,4-diepoxybutane (DEB) are responsible for both its mutagenicity and carcinogenicity. EB, EBD, and DEB are DNA reactive, forming a variety of adducts. All three metabolites are genotoxic in vitro and in vivo, with relative mutagenic potencies of DEB >> EB > EBD. DEB also effectively produces gene deletions and chromosome aberrations. BD’s greater mutagenicity and carcinogenicity in mice over rats as well as its failure to induce chromosome-level mutations in vivo in rats appear to be due to greater production of DEB in mice. Concentrations of EB and DEB in vivo in humans are even lower than in rats. Although most studies of BD-exposed humans have failed to find increases in gene mutations, one group has reported positive findings. Reasons for these discordant results are examined. BD-related chromosome aberrations have never been demonstrated in humans except for the possible production of micronuclei in lymphocytes of workers exposed to extremely high levels of BD in the workplace. The relative potencies of the BD metabolites, their relative abundance in the different species, and the kinds of mutations they can induce are major considerations in BD’s overall genotoxicity profile.


Asunto(s)
Butadienos/metabolismo , Butadienos/toxicidad , Aberraciones Cromosómicas , Animales , Biomarcadores , Pruebas de Carcinogenicidad , Aductos de ADN/genética , Daño del ADN , Drosophila/genética , Compuestos Epoxi/metabolismo , Compuestos Epoxi/toxicidad , Femenino , Humanos , Masculino , Ratones , Mutación , Ratas
10.
Crit Rev Toxicol ; 39(6): 462-86, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19545197

RESUMEN

Propylene oxide (PO) is an important industrial chemical used primarily in the synthesis of other compounds. Inhalation carcinogenesis studies in rodents, with no-observed-adverse-effect levels (NOAELs) of 100 and 200 ppm, have revealed that chronic, high exposure to PO can induce tumors at the site of contact. Despite these characteristics, there is no evidence that typical environmental or occupational exposures to PO constitute a health risk for humans. The nongenotoxic effects of PO (glutathione depletion and cell proliferation) that augment its DNA-reactive and non-DNA-reactive genotoxicity are expected to be similar in humans and rodents. Available evidence on mode-of-action suggests that cancer induction by PO at the site of contact in rodents is characterized by a practical threshold. Human toxicity reference values for potential carcinogenic effects of PO were derived based on nasal tumors identified in rodent studies and specified uncertainty factors. The 95% lower confidence limit on the dose producing a 10% increase in additional tumor risk (LED10) was calculated using the rat and mouse data sets. The human reference values derived from the rat and mouse LED10 values were 0.7 and 0.5 ppm PO, respectively. A similar noncancer reference value, 0.4 ppm, was derived on the basis of non-neoplastic nasal effects in rats.


Asunto(s)
Carcinógenos/análisis , Exposición a Riesgos Ambientales/análisis , Compuestos Epoxi/análisis , Valores Limites del Umbral , Animales , Carcinógenos/metabolismo , Carcinógenos/toxicidad , Exposición a Riesgos Ambientales/efectos adversos , Exposición a Riesgos Ambientales/normas , Compuestos Epoxi/metabolismo , Compuestos Epoxi/toxicidad , Humanos , Valores de Referencia , Medición de Riesgo
11.
Inhal Toxicol ; 21(6): 537-51, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19519154

RESUMEN

Recent studies have indicated that exposures to methyl iodide (MeI) produce a number of effects in laboratory animals, including fetal toxicity, neurotoxicity, and degeneration of the nasal epithelium. An understanding of the mode of action by which the effects of MeI are produced is useful in guiding critical decisions used in risk assessment. These decisions include the selection of the appropriate internal dose measure(s) calculated using physiologically based pharmacokinetic (PBPK) modeling, and evaluating the relevance of the observations in animals to human health. Modified Hill criteria were used to evaluate several possible mode(s) of action through which MeI produces toxicity in animals. For each endpoint, the key studies were summarized and several possible modes of action were compared to the modified Hill criteria. The available data best support the hypothesis that the fetal effects were likely associated with modulation of the thyroid hormones by iodide during development. This mode of action dictates the use of an internal dose measure in the risk assessment that is indicative of fetal iodide status, such as cumulative iodide concentration (area-under-the-curve or AUC) for iodide in fetal blood. The acute transient neurotoxicity observed in rats exposed to MeI is best supported by a mode of action involving modification of ion currents by the parent chemical in nerve cells. In the case of assessing the potential acute neurotoxicity of MeI, the peak concentration of MeI in the brain would be the appropriate internal dose measure. Finally, the nasal lesions associated with exposure to high concentrations of MeI in rats are best supported by a mode of action that involves glutathione (GSH) depletion in the nasal epithelial tissue. The daily minimum GSH level in olfactory epithelium is the most appropriate internal dose measure for use in risk assessment for this endpoint. Confidence in these modes of action is considered low for the neurotoxic effects, medium for the nasal effects, and high for the fetal effects.


Asunto(s)
Hidrocarburos Yodados/administración & dosificación , Hidrocarburos Yodados/metabolismo , Modelos Animales , Animales , Evaluación Preclínica de Medicamentos/métodos , Femenino , Humanos , Masculino , Embarazo , Conejos , Ratas , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
12.
Inhal Toxicol ; 21(6): 583-605, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19519156

RESUMEN

Iodomethane is a new pre-plant soil fumigant approved in the United States. Human exposure may occur via inhalation due to the high vapor pressure of iodomethane. A quantitative human health risk assessment was conducted for inhalation exposure. The critical effects of acute duration iodomethane exposure are: (1) fetal losses in rabbits, (2) lesions in rat nasal epithelium, and (3) transient neurotoxicity in rats. Chronic exposure of rats resulted in increased thyroid follicular cell tumors from sustained perturbation of thyroid hormone homeostasis. A physiologically based pharmacokinetic (PBPK) model for iodomethane was developed to characterize potential human health effects from iodomethane exposure. The model enabled calculation of human equivalent concentrations (HECs) to the animal no-observed-adverse-effect levels (NOAELs) using chemical-specific parameters to determine the internal dose instead of default assumptions. Iodomethane HECs for workers and bystanders were derived using the PBPK model and NOAELs for acute exposure endpoints of concern. The developmental endpoint NOAEL was 10 ppm and corresponding bystander HEC was 7.4 ppm. The nasal endpoint NOAEL was 21 ppm and the HEC was 4.5 ppm. The transient neurotoxicity endpoint NOAEL was 27 ppm and the HEC was10 ppm. Data demonstrated that humans are less sensitive to the effect that causes developmental toxicity in rabbits and the PBPK model incorporated this information, resulting in a higher HEC for the developmental endpoint than for the nasal endpoint. Nasal olfactory degeneration is the primary endpoint for risk assessment of acute exposure to iodomethane.


Asunto(s)
Estado de Salud , Hidrocarburos Yodados/efectos adversos , Exposición por Inhalación/efectos adversos , Animales , Células CHO , Cricetinae , Cricetulus , Cobayas , Humanos , Hidrocarburos Yodados/administración & dosificación , Hidrocarburos Yodados/farmacocinética , Ratones , Conejos , Ratas , Medición de Riesgo , Especificidad de la Especie
13.
Inhal Toxicol ; 21(6): 505-11, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19519150

RESUMEN

Laboratory animals exposed to methyl iodide (MeI) have previously demonstrated lesions of the olfactory epithelium that were associated with local metabolism in the nasal tissues. Interactions of MeI in the nasal passage may, therefore, alter systemic toxicokinetics. The current study used unrestrained plethysmographs to determine the MeI effect on the breathing frequency and minute volume (MV) in rats and rabbits. Groups of 4 rats each were exposed to 0, 25, or 100 ppm and groups of 4 rabbits each were exposed to 0 and 20 ppm MeI for 6 h. Breathing frequency and MV were measured and recorded during the exposure. Blood samples were collected for inorganic serum iodide and the globin adduct S-methylcysteine (SMC) as biomarkers of systemic kinetics immediately following exposure. No significant reductions in breathing frequency were observed for either rats or rabbits. Significant changes in minute volume were demonstrated by both rats and rabbits; however, the changes observed in rats were not concentration dependent. The MeI-induced changes in MV resulted in significant differences in the total volume of test substance atmosphere inhaled over the 6-h period. Rats demonstrated a concentration-dependent increase in both inorganic serum iodide and SMC. Rabbits exposed to 20 ppm MeI demonstrated a significant increase of inorganic serum iodide; SMC was also increased but was not statistically significant. The results of this study are consistent with previous kinetic studies with MeI, and the data presented here can be integrated into a computational fluid dynamics physiologically based pharmacokinetic model for both rats and rabbits.


Asunto(s)
Hidrocarburos Yodados/administración & dosificación , Hidrocarburos Yodados/toxicidad , Exposición por Inhalación/efectos adversos , Mecánica Respiratoria/efectos de los fármacos , Animales , Evaluación Preclínica de Medicamentos/métodos , Femenino , Hidrocarburos Yodados/sangre , Masculino , Conejos , Ratas , Ratas Sprague-Dawley , Mecánica Respiratoria/fisiología
14.
Inhal Toxicol ; 21(6): 552-82, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19519155

RESUMEN

Methyl iodide (MeI) has been proposed as an alternative to methyl bromide as a pre-plant soil fumigant that does not deplete stratospheric ozone. In inhalation toxicity studies performed in animals as part of the registration process, three effects have been identified that warrant consideration in developing toxicity reference values for human risk assessment: nasal lesions (rat), acute neurotoxicity (rat), and fetal loss (rabbit). Uncertainties in the risk assessment can be reduced by using an internal measure of target tissue dose that is linked to the likely mode of action (MOA) for the toxicity of MeI, rather than the external exposure concentration. Physiologically based pharmacokinetic (PBPK) models have been developed for MeI and used to reduce uncertainties in the risk assessment extrapolations (e.g. interspecies, high to low dose, exposure scenario). PBPK model-derived human equivalent concentrations comparable to the animal study NOAELs (no observed adverse effect levels) for the endpoints of interest were developed for a 1-day, 24-hr exposure of bystanders or 8 hr/day exposure of workers. Variability analyses of the PBPK models support application of uncertainty factors (UF) of approximately 2 for intrahuman pharmacokinetic variability for the nasal effects and acute neurotoxicity.


Asunto(s)
Hidrocarburos Yodados/farmacocinética , Modelos Biológicos , Tejido Adiposo/metabolismo , Adulto , Anciano , Animales , Encéfalo/metabolismo , Niño , Femenino , Humanos , Lactante , Riñón/metabolismo , Masculino , Mucosa Nasal/metabolismo , Embarazo , Conejos , Ratas , Ratas Sprague-Dawley , Fenómenos Fisiológicos Respiratorios
15.
Inhal Toxicol ; 21(6): 531-6, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19519153

RESUMEN

Nasal dosimetry models that combine computational fluid dynamics and physiologically based pharmacokinetic modeling incorporate information on species-specific anatomical differences, including nasal airflow, mucosal diffusion, clearance-extraction, and metabolism specific to different epithelial layers. As such, these hybrid models have the potential to improve interspecies dosimetric comparisons, and may ultimately reduce uncertainty associated with calculation of reference concentrations. Validation of these models, however, will require unique experimental data. To this end, a method for evaluating the uptake of a prototypical compound, methyl iodide (MeI), in the nasal cavity of the intact animal was developed. The procedure involved insertion of a small-diameter air-sampling probe in the depth of the nasal cavity to the nasopharynx region in anesthetized animals. The exterior portion of the probe was connected directly to a mass spectrometer to provide a continual real-time analysis of concentrations of MeI in the nasal cavity. A plethysmography system was used to monitor breathing parameters, including frequency and tidal volume for each animal. Animals were placed in a sealed glass chamber and exposed to MeI at initial chamber concentrations ranging from 1 to 50 ppm. Studies were conducted on n = 3 rabbits per exposure concentration for a total of nine animals and n = 6 rats at a single exposure concentration of 1 ppm. In the rabbit, the percent of MeI absorbed in the nasal cavity ranged from 57 to 92% (average 72 +/- 11) regardless of exposure concentration. Similarly, the percent of MeI absorbed in the nasal cavity of the rat ranged from 51 to 71% (average 63 +/- 8).


Asunto(s)
Anestesia , Sistemas de Computación/normas , Hidrocarburos Yodados/metabolismo , Mucosa Nasal/metabolismo , Compuestos Orgánicos Volátiles/metabolismo , Absorción/efectos de los fármacos , Absorción/fisiología , Anestesia/métodos , Animales , Femenino , Hidrocarburos Yodados/administración & dosificación , Exposición por Inhalación , Masculino , Mucosa Nasal/efectos de los fármacos , Conejos , Ratas , Ratas Sprague-Dawley , Compuestos Orgánicos Volátiles/administración & dosificación
16.
Inhal Toxicol ; 21(6): 519-23, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19337875

RESUMEN

Methyl iodide (MeI) is a water soluble monohalomethane that is metabolized in vivo to release iodide (I-). A physiologically based pharmacokinetic (PBPK) model exists for iodide in adult rats, pregnant rats and fetuses, and lactating rats and neonates, but not for pregnant rabbits and fetuses, which have been used extensively for toxicity testing with MeI. Thus, this study was conducted to determine the blood and tissue distribution kinetics of radioiodide in pregnant rabbits and fetuses. Timed-pregnant New Zealand White rabbits received a single intravenous injection of the sodium salt of iodine-131 (Na131I) at either a high (10 mg/kg body weight) or low (0.75 mg/kg body weight) dose on gestation day 25. At various intervals ranging from 0.5 to 24h post- injection, blood and tissues (thyroid, stomach contents, and skin) were collected from each doe, and blood, stomach contents, thyroid, trachea, and amniotic fluid were collected from a random sampling of three fetuses per doe per time point. Radioiodide accumulated as expected in the thyroid of maternal animals, where concentrations were the highest of any maternal tissues measured in both dose groups. Radioiodide also accumulated in fetal blood and tissues; levels were consistently higher than maternal levels and, unlike maternal tissues, showed no evidence of clearance over the 24-h sampling period. In contrast to observations in the maternal animals, fetal stomach contents showed the highest accumulation of radioiodide for both dose groups by 1-2h after dosing, followed by the trachea and thyroid tissues, with the lowest concentrations of radioiodide in the amniotic fluid and blood. There was no evidence for preferential accumulation of radioiodide in fetal thyroid tissues.


Asunto(s)
Hidrocarburos Yodados/farmacocinética , Radioisótopos de Yodo/farmacocinética , Modelos Biológicos , Yoduro de Sodio/farmacocinética , Animales , Femenino , Embarazo , Conejos , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
17.
Birth Defects Res ; 111(1): 26-33, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30511437

RESUMEN

BACKGROUND: To protect crewmember health, the U.S. Navy sets exposure limits for more than 200 components of submarine atmospheres. The addition of females to nuclear submarines required a reevaluation of these exposure limits, originally established for all-male crews. In the case of carbon dioxide (CO2 ), the only available data suitable for deriving an exposure limit were from a 2010 study sponsored by the British Royal Navy that reported a debatable interpretation casting doubt on whether current U.S. Navy exposure limits served to protect fetal developmental health. METHODS: About 120 time-mated female Sprague-Dawley rats (Crl: CD[SD]) were exposed to CO2 at levels of 1.5%, 2.0%, 2.5%, and 3.0% from gestation days 6 to 20. Dams were euthanized and fetuses were examined. RESULTS: Findings with implications for exposure limits for CO2 during pregnancy were an increased mean litter proportion of early resorptions and a lower mean litter proportion of viable fetuses in the 3.0% CO2 group. CONCLUSION: The results yield a No Observed Adverse Effect Level (NOAEL) of 2.5% and a Lowest Observed Adverse Effect Level (LOAEL) of 3.0%. The results reasonably allow a point of departure of 2.5% CO2 for deriving an exposure recommendation. An interspecies uncertainty factor was applied to derive a recommended 90-day continuous exposure limit (CEL) of 0.8% for CO2 . As reproductive endpoints that are developmental in nature must be assumed to result from a single exposure at a critical point during gestation, it is further recommended that the 24-hr emergency exposure limit (EEL) also be 0.8%.


Asunto(s)
Dióxido de Carbono/toxicidad , Medicina Submarina/normas , Animales , Peso Corporal/efectos de los fármacos , Dióxido de Carbono/metabolismo , Modelos Animales de Enfermedad , Femenino , Desarrollo Fetal/efectos de los fármacos , Feto/efectos de los fármacos , Personal Militar , Nivel sin Efectos Adversos Observados , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Sprague-Dawley , Reproducción/efectos de los fármacos , Medicina Submarina/métodos
18.
Toxicol Sci ; 101(1): 32-50, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17897969

RESUMEN

1,4-Dioxane (CAS No. 123-91-1) is used primarily as a solvent or as a solvent stabilizer. It can cause lung, liver, and kidney damage at sufficiently high exposure levels. Two physiologically based pharmacokinetic (PBPK) models of 1,4-dioxane and its major metabolite, hydroxyethoxyacetic acid (HEAA), were published in 1990. These models have uncertainties and deficiencies that could be addressed and the model strengthened for use in a contemporary cancer risk assessment for 1,4-dioxane. Studies were performed to fill data gaps and reduce uncertainties pertaining to the pharmacokinetics of 1,4-dioxane and HEAA in rats, mice, and humans. Three types of studies were performed: partition coefficient measurements, blood time course in mice, and in vitro pharmacokinetics using rat, mouse, and human hepatocytes. Updated PBPK models were developed based on these new data and previously available data. The optimized rate of metabolism for the mouse was significantly higher than the value previously estimated. The optimized rat kinetic parameters were similar to those in the 1990 models. Only two human studies were identified. Model predictions were consistent with one study, but did not fit the second as well. In addition, a rat nasal exposure was completed. The results confirmed water directly contacts rat nasal tissues during drinking water under bioassay conditions. Consistent with previous PBPK models, nasal tissues were not specifically included in the model. Use of these models will reduce the uncertainty in future 1,4-dioxane risk assessments.


Asunto(s)
Dioxanos/farmacocinética , Algoritmos , Animales , Fenómenos Químicos , Química Física , Cromatografía de Gases , Dioxanos/química , Dioxanos/toxicidad , Cromatografía de Gases y Espectrometría de Masas , Hepatocitos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos , Microscopía Fluorescente , Modelos Estadísticos , Exposición Profesional , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Programas Informáticos
19.
Toxicol Lett ; 178(1): 1-8, 2008 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-18384980

RESUMEN

Styrene-acrylonitrile trimer (SAN Trimer), a mixture of six isomers (four isomers of 4-cyano-1,2,3,4-tetrahydro-alpha-methyl-1-naphthaleneacetonitrile [THAN] and two isomers of 4-cyano-1,2,3,4-tetrahydro-1-naphthaleneproprionitrile [THNP]), is a by-product of a specific production process of styrene-acrylonitrile polymer. Disposition studies in female rats were conducted to evaluate the pharmacokinetic behavior of [3H]SAN Trimer following a single intravenous administration (26 mg/kg) to nonpregnant rats; a single gavage administration (nominal doses of 25 mg/kg, 75 mg/kg, or 200 mg/kg in corn oil) to nonpregnant rats; and a single gavage administration (nominal dose of 200 mg/kg in corn oil) to pregnant and lactating rats. SAN Trimer was rapidly eliminated from blood (T1/2 approximately 1h) following a single intravenous dose and following single oral doses (T1/2 approximately 3-4h). SAN Trimer was also rapidly excreted in the urine and feces following single oral doses, while total radioactivity was cleared more slowly. In pregnant rats, the concentrations of both radioactivity and SAN Trimer 2h after dosing were highest in the blood, followed by the placenta, with the lowest levels in the fetus. In lactating rats, the concentrations of both radioactivity and SAN Trimer were higher in milk than in maternal blood. Total radioactivity and SAN Trimer blood concentrations in nonpregnant, pregnant, and lactating rats were both higher in lactating rats compared to nonpregnant and pregnant rats.


Asunto(s)
Intercambio Materno-Fetal , Nitrilos/farmacocinética , Administración Oral , Animales , Heces/química , Femenino , Feto/metabolismo , Inyecciones Intravenosas , Isomerismo , Lactancia/metabolismo , Leche/química , Nitrilos/sangre , Nitrilos/orina , Placenta/química , Embarazo , Ratas , Ratas Endogámicas F344 , Estireno
20.
Reprod Toxicol ; 25(1): 129-32, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18333297

RESUMEN

OBJECTIVES: To characterize the matched maternal and cord plasma and the amniotic fluid concentrations of iodide in preterm and term human pregnancies. METHODS: Specimens were collected at the delivery of 121 singleton pregnancies (92 at term, 29 preterm) with no pre-existing medical complications. Plasma unbound iodide concentrations were measured by the difference between the protein bound iodine and the total iodine measured spectrophotometrically. Total iodide was measured in amniotic fluid. RESULTS: Maternal plasma iodide concentrations were 1.6 +/- 0.4 mcg/dL (mean +/- S.D.) for preterm deliveries and 1.5+/ -0.5 mcg/dL for term deliveries. Cord plasma iodide concentrations were 1.4 +/- 0.5 mcg/dL for preterm deliveries and 1.7 +/- 0.7 mcg/dL for term deliveries. Cord plasma iodide concentrations at birth correlated highly with maternal levels (p < 0.001). The cord:maternal plasma iodide ratio for all pairs was 1.2+/- 0.7. The average cord:maternal plasma iodide ratio was not significantly different between the preterm (0.9+/- 0.4) and term (1.3+/- 0.8) deliveries. Amniotic fluid iodide concentrations did not correlate significantly with cord plasma concentrations. CONCLUSION: Cord plasma concentrations of iodide correlate with paired maternal levels, indicating that, unlike the rabbit and other species, the human conceptus does not highly concentrate iodide.


Asunto(s)
Líquido Amniótico/química , Sangre Fetal/química , Recien Nacido Prematuro/sangre , Yoduros/análisis , Yoduros/sangre , Femenino , Edad Gestacional , Humanos , Recién Nacido , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA