Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Toxicol Sci ; 182(2): 243-259, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34077545

ABSTRACT

Regulatory agencies are considering alternative approaches to assessing inhalation toxicity that utilizes in vitro studies with human cells and in silico modeling in lieu of additional animal studies. In support of this goal, computational fluid-particle dynamics models were developed to estimate site-specific deposition of inhaled aerosols containing the fungicide, chlorothalonil, in the rat and human for comparisons to prior rat inhalation studies and new human in vitro studies. Under bioassay conditions, the deposition was predicted to be greatest at the front of the rat nose followed by the anterior transitional epithelium and larynx corresponding to regions most sensitive to local contact irritation and cytotoxicity. For humans, simulations of aerosol deposition covering potential occupational or residential exposures (1-50 µm diameter) were conducted using nasal and oral breathing. Aerosols in the 1-5 µm range readily penetrated the deep region of the human lung following both oral and nasal breathing. Under actual use conditions (aerosol formulations >10 µm), the majority of deposited doses were in the upper conducting airways. Beyond the nose or mouth, the greatest deposition in the pharynx, larynx, trachea, and bronchi was predicted for aerosols in the 10-20 µm size range. Only small amounts of aerosols >20 µm penetrated past the pharyngeal region. Using the ICRP clearance model, local retained tissue dose metrics including maximal concentrations and areas under the curve were calculated for each airway region following repeated occupational exposures. These results are directly comparable with benchmark doses from in vitro toxicity studies in human cells leading to estimated human equivalent concentrations that reduce the reliance on animals for risk assessments.


Subject(s)
Hydrodynamics , Lung , Administration, Inhalation , Aerosols/toxicity , Animals , Computer Simulation , Humans , Models, Biological , Particle Size , Rats
2.
Toxicol Appl Pharmacol ; 417: 115463, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33631232

ABSTRACT

By extending our Paraquat (PQ) work to include primates we have implemented a modelling and simulation strategy that has enabled PQ pharmacokinetic data to be integrated into a single physiologically based pharmacokinetic (PBPK) model that enables more confident extrapolation to humans. Because available data suggested there might be differences in PQ kinetics between primates and non-primates, a radiolabelled study was conducted to characterize pharmacokinetics and excretion in Cynomolgus monkeys. Following single intravenous doses of 0.01 or 0.1 mg paraquat dichloride/kg bw, plasma PQ concentration-time profiles were dose-proportional. Excretion up to 48 h (predominantly urinary) was 82.9%, with ca. 10% remaining unexcreted. In vitro blood binding was similar across Cynomolgus monkeys, humans and rat. Our PBPK model for the rat, mouse and dog, employing a single set of PQ-specific parameters, was scaled to Cynomolgus monkeys and well represented the measured plasma concentration-time profiles over 14 days. Addition of a cartilage compartment to the model better captured the percent remaining in the monkeys at 48 h, whilst having negligible effect on model predictions for the other species. The PBPK model performed well for all four species, demonstrating there is little difference in PQ kinetics between non-primates and primates enabling a more confident extrapolation to humans. Scaling of the PBPK model to humans, with addition of a human-specific dermal submodel based on in vitro human dermal absorption data, provides a valuable tool that could be employed in defining internal dosimetry to complement human health risk assessments.


Subject(s)
Herbicides/pharmacokinetics , Models, Biological , Paraquat/pharmacokinetics , Animals , Computer Simulation , Herbicides/administration & dosage , Herbicides/blood , Herbicides/toxicity , Humans , Infusions, Intravenous , Intestinal Elimination , Macaca fascicularis , Paraquat/administration & dosage , Paraquat/blood , Paraquat/toxicity , Rats , Renal Elimination , Risk Assessment , Skin Absorption , Species Specificity , Tissue Distribution , Toxicokinetics
3.
Toxicol Appl Pharmacol ; 417: 115462, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33631233

ABSTRACT

Paraquat dichloride (PQ) is a non-selective herbicide which has been the subject of numerous toxicology studies over more than 50 years. This paper describes the development of a physiologically-based pharmacokinetic (PBPK) model of PQ kinetics for the rat, mouse and dog, firstly to aid the interpretation of studies in which no kinetic measurements were made, and secondly to enable the future extension of the model to humans. Existing pharmacokinetic data were used to develop a model for the rat and mouse. Simulations with this preliminary model were then used to identify key data gaps and to design a new blood binding study to reduce uncertainty in critical aspects of the model. The new data provided evidence to support the model structure, and its predictive performance was then assessed against dog and rat datasets not used in model development. The PQ-specific model parameters are the same for all three species, with only the physiological parameters varying between species. This consistency across species provides a strong basis for extrapolation to other species, as demonstrated here for the dog. The model enables a wide range of PQ data to be linked together to provide a broad understanding of PQ pharmacokinetics in rodents and the dog, showing that the key aspects of PQ kinetics in these species are understood and adequately encapsulated within the model.


Subject(s)
Herbicides/pharmacokinetics , Models, Biological , Paraquat/pharmacokinetics , Animals , Computer Simulation , Dogs , Herbicides/blood , Herbicides/toxicity , Intestinal Elimination , Mice , Paraquat/blood , Paraquat/toxicity , Protein Binding , Rats , Renal Elimination , Risk Assessment , Species Specificity , Tissue Distribution , Toxicokinetics
4.
Toxicol Sci ; 169(2): 365-379, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30768128

ABSTRACT

An in vitro to in vivo (IVIVE) extrapolation based-physiologically based pharmacokinetic (PBPK) modeling approach was demonstrated to understand age-related differences in kinetics and how they potentially affect age-related differences in acute neurotoxic effects of pyrethroids. To describe the age-dependent changes in pyrethroid kinetics, it was critical to incorporate age-dependent changes in metabolism into the model. As such, in vitro metabolism data were collected for 3 selected pyrethroids, deltamethrin (DLM), cis-permethrin, and trans-permethrin, using liver microsomes and cytosol, and plasma prepared from immature and adult rats. Resulting metabolism parameters, maximum rate of metabolism (Vmax) and Michaelis-Menten constant (Km), were biologically scaled to respective in vivo parameters for use in the age-specific PBPK model. Then, age-dependent changes in target tissue exposure, i.e., brain Cmax, to a given pyrethroid were simulated across ages using the model. The PBPK model recapitulated in vivo time-course plasma and brain concentrations of the 3 pyrethroids in immature and adult rats following oral administration of both low and high doses of these compounds. A single model structure developed for DLM was able to describe the kinetics of the other 2 pyrethroids when used with compound- and age-specific metabolism parameters, suggesting that one generic model for pyrethroids as a group can be used for early age-sensitivity evaluation if appropriate metabolic parameters are used. This study demonstrated the validity of applying IVIVE-based PBPK modeling to development of age-specific PBPK models for pyrethroids in support of pyrethroid risk assessment of potentially sensitive early age populations in humans.


Subject(s)
Insecticides/pharmacokinetics , Pyrethrins/pharmacokinetics , Age Factors , Animals , Inactivation, Metabolic , Intestinal Absorption , Male , Models, Biological , Permeability , Rats , Rats, Sprague-Dawley
6.
Toxicol Sci ; 150(2): 441-53, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26794140

ABSTRACT

The previously-published physiologically based pharmacokinetic model for atrazine (ATZ), deisopropylatrazine (DIA), deethylatrazine (DEA), and diaminochlorotriazine (DACT), which collectively comprise the total chlorotriazines (TCT) as represented in this study, was modified to allow for scaling to humans. Changes included replacing the fixed dose-dependent oral uptake rates with a method that represented delayed absorption observed in rats administered ATZ as a bolus dose suspended in a methylcellulose vehicle. Rate constants for metabolism of ATZ to DIA and DEA, followed by metabolism of DIA and DEA to DACT were predicted using a compartmental model describing the metabolism of the chlorotriazines by rat and human hepatocytesin vitro Overall, the model successfully predicted both the 4-day plasma time-course data in rats administered ATZ by bolus dose (3, 10, and 50 mg/kg/day) or in the diet (30, 100, or 500 ppm). Simulated continuous daily exposure of a 55-kg adult female to ATZ at a dose of 1.0 µg/kg/day resulted in steady-state urinary concentrations of 0.6, 1.4, 2.5, and 6.0 µg/L for DEA, DIA, DACT, and TCT, respectively. The TCT (ATZ + DEA + DIA + DACT) human urinary biomonitoring equivalent concentration following continuous exposure to ATZ at the chronic point of departure (POD = 1.8 mg/kg/day) was 360.6 µg/L.


Subject(s)
Atrazine/pharmacokinetics , Hepatocytes/metabolism , Models, Biological , Triazines/pharmacokinetics , Absorption, Physiological , Administration, Oral , Animals , Atrazine/blood , Atrazine/urine , Cells, Cultured , Dose-Response Relationship, Drug , Female , Hepatocytes/drug effects , Humans , Rats, Sprague-Dawley , Species Specificity , Tissue Distribution , Triazines/blood , Triazines/urine
7.
Regul Toxicol Pharmacol ; 69(3): 580-97, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24200834

ABSTRACT

Sensitivity to some chemicals in animals and humans are known to vary with age. Age-related changes in sensitivity to chlorpyrifos have been reported in animal models. A life-stage physiologically based pharmacokinetic and pharmacodynamic (PBPK/PD) model was developed to predict disposition of chlorpyrifos and its metabolites, chlorpyrifos-oxon (the ultimate toxicant) and 3,5,6-trichloro-2-pyridinol (TCPy), as well as B-esterase inhibition by chlorpyrifos-oxon in humans. In this model, previously measured age-dependent metabolism of chlorpyrifos and chlorpyrifos-oxon were integrated into age-related descriptions of human anatomy and physiology. The life-stage PBPK/PD model was calibrated and tested against controlled adult human exposure studies. Simulations suggest age-dependent pharmacokinetics and response may exist. At oral doses ⩾0.6mg/kg of chlorpyrifos (100- to 1000-fold higher than environmental exposure levels), 6months old children are predicted to have higher levels of chlorpyrifos-oxon in blood and higher levels of red blood cell cholinesterase inhibition compared to adults from equivalent doses. At lower doses more relevant to environmental exposures, simulations predict that adults will have slightly higher levels of chlorpyrifos-oxon in blood and greater cholinesterase inhibition. This model provides a computational framework for age-comparative simulations that can be utilized to predict chlorpyrifos disposition and biological response over various postnatal life stages.


Subject(s)
Chlorpyrifos/pharmacokinetics , Environmental Exposure/adverse effects , Environmental Exposure/analysis , Adult , Age Factors , Carboxylesterase/blood , Carboxylesterase/metabolism , Carboxylesterase/pharmacokinetics , Carboxylesterase/urine , Child, Preschool , Chlorpyrifos/analogs & derivatives , Chlorpyrifos/blood , Chlorpyrifos/metabolism , Chlorpyrifos/urine , Cholinesterase Inhibitors/blood , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/pharmacokinetics , Cholinesterase Inhibitors/urine , Female , Humans , Infant , Male , Models, Biological , Pyridones/blood , Pyridones/metabolism , Pyridones/pharmacokinetics , Pyridones/urine
8.
Regul Toxicol Pharmacol ; 65(1): 12-28, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23099439

ABSTRACT

The pharmacokinetics of nicotine, the pharmacologically active alkaloid in tobacco responsible for addiction, are well characterized in humans. We developed a physiologically based pharmacokinetic/pharmacodynamic model of nicotine pharmacokinetics, brain dosimetry and brain nicotinic acetylcholine receptor (nAChRs) occupancy. A Bayesian framework was applied to optimize model parameters against multiple human data sets. The resulting model was consistent with both calibration and test data sets, but in general underestimated variability. A pharmacodynamic model relating nicotine levels to increases in heart rate as a proxy for the pharmacological effects of nicotine accurately described the nicotine related changes in heart rate and the development and decay of tolerance to nicotine. The PBPK model was utilized to quantitatively capture the combined impact of variation in physiological and metabolic parameters, nicotine availability and smoking compensation on the change in number of cigarettes smoked and toxicant exposure in a population of 10,000 people presented with a reduced toxicant (50%), reduced nicotine (50%) cigarette Across the population, toxicant exposure is reduced in some but not all smokers. Reductions are not in proportion to reductions in toxicant yields, largely due to partial compensation in response to reduced nicotine yields. This framework can be used as a key element of a dosimetry-driven risk assessment strategy for cigarette smoke constituents.


Subject(s)
Cotinine/pharmacokinetics , Models, Biological , Nicotine/pharmacokinetics , Receptors, Nicotinic/metabolism , Animals , Bayes Theorem , Brain/metabolism , Heart Rate/drug effects , Humans , Nicotine/administration & dosage , Nicotine/pharmacology , Nicotinic Agonists/pharmacokinetics , Nicotinic Agonists/pharmacology , Risk Assessment/methods , Smoking/adverse effects , Smoking/metabolism , Tissue Distribution
9.
Regul Toxicol Pharmacol ; 61(1): 82-92, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21722690

ABSTRACT

Probabilistic models of interindividual variation in exposure and response were linked to create a source-to-outcome population model. This model was used to investigate cholinesterase inhibition from dietary exposures to an insecticide (chlorpyrifos) in populations of adults and 3 year old children. A physiologically based pharmacokinetic and pharmacodynamic (PBPK/PD) model was used to calculate the variation in sensitivity occurring from interindividual variability in physiology, metabolism, and physical activity levels. A dietary intake model characterizes the variation in dietary insecticide exposures and variation in anthropometry in the populations. Published equations were used to describe the necessary physiology for each simulated individual based on the anthropometry from the dietary intake model. The model of the interindividual variation in response to chlorpyrifos was developed by performing a sensitivity analysis on the PBPK/PD model to determine the parameters that drive variation in pharmacodynamics outcomes (brain and red blood cell acetylcholinesterase inhibition). Distributions of interindividual variation were developed for parameters with the largest impact; the probabilistic model sampled from these distributions. The impact of age and interindividual variation on sensitivity at the doses that occur from dietary exposures, typically orders of magnitude lower than exposures assessed in toxicological studies, was assessed using the source-to-outcome model. The resulting simulations demonstrated that metabolic detoxification capacity was sufficient to prevent significant brain and red blood cell acetylcholinesterase inhibition, even in individuals with the lowest detoxification potential. Age-specific pharmacokinetic and pharmacodynamic parameters did not predict differences in susceptibility between adults and children. In the future, the approach of this case study could be used to assess the risks from low level exposures to other chemicals.


Subject(s)
Chlorpyrifos , Cholinesterase Inhibitors , Computer Simulation , Cytochrome P-450 Enzyme System/metabolism , Food Contamination , Insecticides , Pesticide Residues , Software , Acetylcholinesterase/metabolism , Adipose Tissue/metabolism , Adult , Brain/metabolism , Child, Preschool , Chlorpyrifos/pharmacokinetics , Chlorpyrifos/pharmacology , Chlorpyrifos/toxicity , Cholinesterase Inhibitors/blood , Cholinesterase Inhibitors/pharmacokinetics , Cholinesterase Inhibitors/pharmacology , Diet , Dose-Response Relationship, Drug , Female , Forecasting , Humans , Insecticides/blood , Insecticides/pharmacokinetics , Insecticides/pharmacology , Insecticides/toxicity , Liver/metabolism , Male , Models, Biological , Models, Statistical , Pesticide Residues/pharmacokinetics , Pesticide Residues/toxicity
10.
Regul Toxicol Pharmacol ; 61(1): 23-31, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21651950

ABSTRACT

The paper presents a case study of the application of a "source-to-outcome" model for the evaluation of the health outcomes from dietary exposures to an insecticide, chlorpyrifos, in populations of adults (age 30) and children (age 3). The model is based on publically-available software programs that characterize the longitudinal dietary exposure and anthropometry of exposed individuals. These predictions are applied to a validated PBPK/PD model to estimate interindividual and longitudinal variation in brain and RBC AChE inhibition (key events) and chlorpyrifos concentrations in blood and TCPy in urine (biomarkers of exposure). The predicted levels of chlorpyrifos and TCPy are compared to published measurements of the biomarkers. Predictions of RBC AChE are compared to levels of inhibition associated with reported exposure-related effects in humans to determine the potential for the occurrence of adverse cholinergic effects. The predicted distributions of chlorpyrifos in blood and TCPy in urine were found to be reasonably consistent with published values, supporting the predictive value of the exposure and PBPK portions of the source-to-outcome model. Key sources of uncertainty in predictions of dietary exposures were investigated and found to have a modest impact on the model predictions. Future versions of this source-to-outcome model can be developed that consider advances in our understanding of metabolism, to extend the approach to other age groups (infants), and address intakes from other routes of exposure.


Subject(s)
Acetylcholinesterase/metabolism , Chlorpyrifos/toxicity , Environmental Exposure , Insecticides/toxicity , Pesticide Residues/toxicity , Software , Adult , Biomarkers/blood , Biomarkers/urine , Child, Preschool , Chlorpyrifos/metabolism , Computer Simulation , Diet , Food Contamination , Humans , Insecticides/metabolism , Longitudinal Studies , Nutrition Policy , Pesticide Residues/metabolism , Predictive Value of Tests , Risk Assessment/methods
11.
Part Fibre Toxicol ; 7(1): 36, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21118529

ABSTRACT

BACKGROUND: The difficulty of directly measuring cellular dose is a significant obstacle to application of target tissue dosimetry for nanoparticle and microparticle toxicity assessment, particularly for in vitro systems. As a consequence, the target tissue paradigm for dosimetry and hazard assessment of nanoparticles has largely been ignored in favor of using metrics of exposure (e.g. µg particle/mL culture medium, particle surface area/mL, particle number/mL). We have developed a computational model of solution particokinetics (sedimentation, diffusion) and dosimetry for non-interacting spherical particles and their agglomerates in monolayer cell culture systems. Particle transport to cells is calculated by simultaneous solution of Stokes Law (sedimentation) and the Stokes-Einstein equation (diffusion). RESULTS: The In vitro Sedimentation, Diffusion and Dosimetry model (ISDD) was tested against measured transport rates or cellular doses for multiple sizes of polystyrene spheres (20-1100 nm), 35 nm amorphous silica, and large agglomerates of 30 nm iron oxide particles. Overall, without adjusting any parameters, model predicted cellular doses were in close agreement with the experimental data, differing from as little as 5% to as much as three-fold, but in most cases approximately two-fold, within the limits of the accuracy of the measurement systems. Applying the model, we generalize the effects of particle size, particle density, agglomeration state and agglomerate characteristics on target cell dosimetry in vitro. CONCLUSIONS: Our results confirm our hypothesis that for liquid-based in vitro systems, the dose-rates and target cell doses for all particles are not equal; they can vary significantly, in direct contrast to the assumption of dose-equivalency implicit in the use of mass-based media concentrations as metrics of exposure for dose-response assessment. The difference between equivalent nominal media concentration exposures on a µg/mL basis and target cell doses on a particle surface area or number basis can be as high as three to six orders of magnitude. As a consequence, in vitro hazard assessments utilizing mass-based exposure metrics have inherently high errors where particle number or surface areas target cells doses are believed to drive response. The gold standard for particle dosimetry for in vitro nanotoxicology studies should be direct experimental measurement of the cellular content of the studied particle. However, where such measurements are impractical, unfeasible, and before such measurements become common, particle dosimetry models such as ISDD provide a valuable, immediately useful alternative, and eventually, an adjunct to such measurements.


Subject(s)
Computer Simulation , Ferric Compounds/pharmacokinetics , Models, Biological , Nanoparticles , Polystyrenes/pharmacokinetics , Silicon Dioxide/pharmacokinetics , Animal Testing Alternatives , Biological Transport , Biomarkers, Pharmacological , Cells, Cultured , Chemical Precipitation , Diffusion , Dose-Response Relationship, Drug , Humans , Particle Size , Risk Assessment , Toxicity Tests
12.
Food Chem Toxicol ; 48(11): 3085-92, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20692312

ABSTRACT

A physiologically based pharmacokinetic (PBPK) model for hydroquinone (HQ) was refined to include an expanded description of HQ-glucuronide metabolites and a description of dermal exposures to support route-to-route and cross-species extrapolation. Total urinary excretion of metabolites from in vivo rat dermal exposures was used to estimate a percutaneous permeability coefficient (K(p); 3.6×10(-5) cm/h). The human in vivo K(p) was estimated to be 1.62×10(-4) cm/h, based on in vitro skin permeability data in rats and humans and rat in vivo values. The projected total multi-substituted glutathione (which was used as an internal dose surrogate for the toxic glutathione metabolites) was modeled following an exposure scenario based on submersion of both hands in a 5% aqueous solution of HQ (similar to black and white photographic developing solution) for 2 h, a worst-case exposure scenario. Total multi-substituted glutathione following this human dermal exposure scenario was several orders of magnitude lower than the internal total glutathione conjugates in rats following an oral exposure to the rat NOEL of 20 mg/kg. Thus, under more realistic human dermal exposure conditions, it is unlikely that toxic glutathione conjugates (primarily the di- and, to a lesser degree, the tri-glutathione conjugate) will reach significant levels in target tissues.


Subject(s)
Antioxidants/pharmacokinetics , Hydroquinones/pharmacokinetics , Occupational Exposure/adverse effects , Skin/metabolism , Administration, Cutaneous , Animals , Female , Glutathione/metabolism , Humans , Male , Models, Biological , Permeability/drug effects , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Risk Assessment , Skin/drug effects , Species Specificity
13.
Toxicol Sci ; 113(2): 468-82, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19875680

ABSTRACT

Establishing an occupational exposure limit (OEL) for N-methyl pyrrolidone (NMP) is important due to its widespread use as a solvent. Based on studies in rodents, the most sensitive toxic end point is a decrease in fetal/pup body weights observed after oral, dermal, and inhalation exposures of dams to NMP. Evidence indicates that the parent compound is the causative agent. To reduce the uncertainty in rat to human extrapolations, physiologically based pharmacokinetic (PBPK) models were developed to describe the pharmacokinetics of NMP in both species. Since in utero exposures are of concern, the models considered major physiological changes occurring in the dam or mother over the course of gestation. The rat PBPK model was used to determine the relationship between NMP concentrations in maternal blood and decrements in fetal/pup body weights following exposures to NMP vapor. Body weight decrements seen after vapor exposures occurred at lower NMP blood levels than those observed after oral and dermal exposures. Benchmark dose modeling was used to better define a point of departure (POD) for fetal/pup body weight changes based on dose-response information from two inhalation studies in rats. The POD and human PBPK model were then used to estimate the human equivalent concentrations (HECs) that could be used to derive an OEL value for NMP. The geometric mean of the PODs derived from the rat studies was estimated to be 350 mg h/l (expressed in terms of internal dose), a value which corresponds to an HEC of 480 ppm (occupational exposure of 8 h/day, 5 days/week). The HEC is much higher than recently developed internationally recognized OELs for NMP of 10-20 ppm, suggesting that these OELs adequately protect workers exposed to NMP vapor.


Subject(s)
Environmental Exposure/standards , Pyrrolidinones/pharmacokinetics , Adult , Animals , Benchmarking , Dose-Response Relationship, Drug , Environmental Exposure/analysis , Female , Humans , Male , Mice , Models, Animal , Models, Chemical , Pregnancy , Pyrrolidinones/blood , Pyrrolidinones/toxicity , Rats , Risk Assessment , Stress, Physiological , Young Adult
14.
Toxicology ; 244(2-3): 87-97, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-18096291

ABSTRACT

The thermoregulatory system of laboratory rodents is susceptible to a variety of chemical toxicants. Because temperature directly affects the reaction of virtually all biological processes, it is critical to consider how changes in the thermoregulatory response to a toxicant may affect physiological, behavioral, and pathological endpoints. Researchers in industry and government laboratories are often faced with addressing how changes in body temperature of their experimental subjects may affect the outcome of a particular toxicity test and/or screening panel. However, many toxicologists are either unaware of the importance or ignore the potential impact of a toxic-induced change in body temperature. This paper endeavors to summarize the importance of thermoregulation in the study of toxicology and propose recommendations for thermometry that researchers may utilize in their toxicological studies.


Subject(s)
Body Temperature Regulation/drug effects , Body Temperature Regulation/physiology , Toxicity Tests , Animals , Animals, Newborn , Humans , Hypothermia/chemically induced , Hypothermia/physiopathology , Thermometers
15.
Toxicol Sci ; 95(2): 300-12, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17098817

ABSTRACT

The rapid growth in the use of in vitro methods for nanoparticle toxicity assessment has proceeded with limited consideration of the unique kinetics of these materials in solution. Particles in general and nanoparticles specifically, diffuse, settle, and agglomerate in cell culture media as a function of systemic and particle properties: media density and viscosity and particle size, shape, charge and density, for example. Cellular dose then is also a function of these factors as they determine the rate of transport of nanoparticles to cells in culture. Here we develop and apply the principles of dosimetry in vitro and outline an approach for simulation of nanoparticle particokinetics in cell culture systems. We illustrate that where equal mass concentrations (mug/ml) imply equal doses for dissimilar materials, the corresponding particle number or surface area concentration doses differ by orders of magnitude. More importantly, when rates of diffusional and gravitational particle delivery are accounted for, trends and magnitude of the cellular dose as a function of particle size and density differ significantly from those implied by "concentration" doses. For example, 15-nm silver nanoparticles appear approximately 4000 times more potent than micron-sized cadmium oxide particles on a cm(2)/ml media basis, but are only approximately 50 times more potent when differences in delivery to adherent cells are considered. We conclude that simple surrogates of dose can cause significant misinterpretation of response and uptake data for nanoparticles in vitro. Incorporating particokinetics and principles of dosimetry would significantly improve the basis for nanoparticle toxicity assessment, increasing the predictive power and scalability of such assays.


Subject(s)
Nanoparticles/toxicity , Cells, Cultured , Diffusion , Dose-Response Relationship, Drug , Gravitation , Models, Biological , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Particle Size
16.
Toxicol Sci ; 91(2): 341-55, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16543293

ABSTRACT

The absorption, distribution, metabolism, and elimination of [3-14C] 8-2 fluorotelomer alcohol (8-2 FTOH, C7F1514CF2CH2CH2OH) following a single oral dose at 5 and 125 mg/kg in male and female rats have been determined. Following oral dosing, the maximum concentration of 8-2 FTOH in plasma occurred by 1 h postdose and cleared rapidly with a half-life of less than 5 h. The internal dose to 8-2 FTOH, as measured by area under the concentration-time curve to infinity, was similar for male and female rats and was observed to increase in a dose-dependent fashion. The majority of the 14C 8-2 FTOH (> 70%) was excreted in feces, and 37-55% was identified as parent. Less than 4% of the administered dose was excreted in urine, which contained low concentrations of perfluorooctanoate (approximately 1% of total 14C). Metabolites identified in bile were principally composed of glucuronide and glutathione conjugates, and perfluorohexanoate was identified in excreta and plasma, demonstrating the metabolism of the parent FTOH by sequential removal of multiple CF2 groups. At 7 days postdose, 4-7% of the administered radioactivity was present in tissues, and for the majority, 14C concentrations were greater than whole blood with the highest concentration in fat, liver, thyroid, and adrenals. Distribution and excretion of a single 125-mg/kg [3-14C] 8-2 FTOH dermal dose following a 6-h exposure in rats was also determined. The majority of the dermal dose either volatilized from the skin (37%) or was removed by washing (29%). Following a 6-h dermal exposure and a 7-day collection period, excretion of total radioactivity via urine (< 0.1%) and feces (< 0.2%) was minor, and radioactivity concentrations in most tissues were below the limit of detection. Systemic availability of 8-2 FTOH following dermal exposure was negligible.


Subject(s)
Fatty Alcohols/pharmacokinetics , Absorption , Administration, Cutaneous , Administration, Oral , Animals , Bile/chemistry , Cells, Cultured , Fatty Alcohols/administration & dosage , Fatty Alcohols/blood , Fatty Alcohols/urine , Feces/chemistry , Female , Hepatocytes/metabolism , Male , Metabolic Clearance Rate , Rats , Rats, Inbred Strains , Tissue Distribution
17.
Toxicology ; 211(1-2): 139-48, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15863257

ABSTRACT

This study was conducted to develop a quantitative understanding of the potential for gestational and lactational transfer of perfluorooctanoate (PFOA) in the rat. Time-mated female rats were dosed by oral gavage once daily at concentrations of 3, 10, or 30 mg/kg/day of the ammonium salt of PFOA (APFO) starting on gestation (G) day 4 and continuing until sacrifice. On days 10, 15, and 21G, five rats per dose level were sacrificed and blood samples were collected 2h post-dose. Embryos were collected on day 10G, amniotic fluid, placentas, and embryos/fetuses were collected on days 15 and 21G, and fetal blood samples were collected on day 21G. Five rats per dose level were allowed to deliver and nurse their litters, and on days 3, 7, 14, and 21 post-partum (PP) milk and blood samples of maternal and pup were collected 2h post-dose. All samples were analyzed by high-performance liquid chromatography-mass spectrometry (HPLC-MS) for PFOA concentration. Concentrations of PFOA in maternal plasma and milk attained steady state during the sampling interval. The steady-state concentrations in maternal plasma were 10-15, 25-30, and 60-75 microg/mL in rats receiving 3, 10, and 30 mg/kg, respectively. Steady-state concentrations in milk were approximately 10 times less than those in maternal plasma. The concentration of PFOA in fetal plasma on day 21G was approximately half the steady-state concentration in maternal plasma. The milk concentrations appeared to be generally comparable to the concentrations in pup plasma. Pup plasma concentrations decreased from day 3PP to day 7PP, and were similar on days 7, 14, and 21PP at all dose levels. PFOA was detected in placenta (days 15 and 21G), amniotic fluid (days 15 and 21G), embryo (days 10 and 15G), and fetus (day 21G). These pharmacokinetics allow estimation of the dose to developing and nursing rat offspring following maternal exposure.


Subject(s)
Caprylates/pharmacokinetics , Fluorocarbons/pharmacokinetics , Lactation/physiology , Placenta/metabolism , Animals , Biotransformation , Body Weight/drug effects , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Female , Fetus/metabolism , Litter Size/drug effects , Mass Spectrometry , Maternal-Fetal Exchange , Milk/metabolism , Pregnancy , Rats
18.
Drug Chem Toxicol ; 27(4): 341-60, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15573471

ABSTRACT

Perfluorooctanoic acid (PFOA) is an organic fluorochemical and is reported to have a long half-life in human blood. Its urinary elimination in rats is markedly sex-dependent, and characterized by significantly longer plasma half-life of PFOA in male rats than in females. It has been postulated that male-specific PFOA binding protein(s) is responsible for the long half-life of PFOA in male rats. In this paper, two male rat specific proteins, liver- and kidney-form alpha2u-globulins (A2U(L) and A2U(K)), were purified from male rat urine and kidney, respectively. The binding of these two nroteins to PFOA was investigated using ligand blotting, electrospray ionization mass spectrometry and fluorescence competitive binding assay. The results revealed that both A2U(L) and A2U(K) were able to bind PFOA in vitro under physiological conditions, and that PFOA and a fluorescent-labeled fatty acid shared the same binding site on both A2U(L) and A2U(K). The binding affinities, however, are relatively weak. The estimated dissociation constants are in the 10(-3) M range, indicating that bindings of PFOA to either A2U(L) or A2U(K) cannot adequately explain the sex-dependent elimination of PFOA in rats, and it is unlikely that PFOA-A2U(K) binding would induce A2U nephropathy as seen with, for example, 1,4-dichlorobenzene.


Subject(s)
Alpha-Globulins/metabolism , Caprylates/metabolism , Fluorocarbons/metabolism , Alpha-Globulins/isolation & purification , Animals , Female , Kidney/metabolism , Kidney Diseases/etiology , Liver/metabolism , Male , Organic Anion Transporters, Sodium-Independent/physiology , Protein Binding , Rats , Sex Characteristics , Spectrometry, Mass, Electrospray Ionization
19.
Toxicol Sci ; 79(1): 28-37, 2004 May.
Article in English | MEDLINE | ID: mdl-14976335

ABSTRACT

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.


Subject(s)
Chloroprene/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Administration, Inhalation , Animals , Atmosphere Exposure Chambers , Chloroprene/chemistry , Chloroprene/pharmacology , Cricetinae , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Forecasting , Humans , Kidney/drug effects , Kidney/metabolism , Kinetics , Liver/drug effects , Liver/metabolism , Lung/cytology , Lung/drug effects , Lung/metabolism , Male , Mesocricetus , Mice , Mice, Inbred Strains , Models, Biological , Muscles/drug effects , Muscles/metabolism , Rats , Rats, Inbred F344 , Rats, Wistar , Tissue Distribution/drug effects
20.
Toxicol Sci ; 79(1): 18-27, 2004 May.
Article in English | MEDLINE | ID: mdl-14976339

ABSTRACT

Beta-chloroprene (2-chloro-1,3-butadiene, CD) is carcinogenic by inhalation exposure to B6C3F1 mice and Fischer F344 rats but not to Wistar rats or Syrian hamsters. The initial step in metabolism is oxidation, forming a stable epoxide (1-chloroethenyl)oxirane (1-CEO), a genotoxicant that might be involved in rodent tumorigenicity. This study investigated the species-dependent in vitro kinetics of CD oxidation and subsequent 1-CEO metabolism by microsomal epoxide hydrolase and cytosolic glutathione S-transferases in liver and lung, tissues that are prone to tumor induction. Estimates for Vmax and Km for cytochrome P450-dependent oxidation of CD in liver microsomes ranged from 0.068 to 0.29 micromol/h/mg protein and 0.53 to 1.33 microM, respectively. Oxidation (Vmax/Km) of CD in liver was slightly faster in the mouse and hamster than in rats or humans. In lung microsomes, Vmax/Km was much greater for mice compared with the other species. The Vmax and Km estimates for microsomal epoxide hydrolase activity toward 1-CEO ranged from 0.11 to 3.66 micromol/h/mg protein and 20.9 to 187.6 microM, respectively, across tissues and species. Hydrolysis (Vmax/Km) of 1-CEO in liver and lung microsomes was faster for the human and hamster than for rat or mouse. The Vmax/Km in liver was 3 to 11 times greater than in lung. 1-CEO formation from CD was measured in liver microsomes and was estimated to be 2-5% of the total CD oxidation. Glutathione S-transferase-mediated metabolism of 1-CEO in cytosolic tissue fractions was described as a pseudo-second order reaction; rates were 0.0016-0.0068/h/mg cytosolic protein in liver and 0.00056-0.0022 h/mg in lung. The observed differences in metabolism are relevant to understanding species differences in sensitivity to CD-induced liver and lung tumorigenicity.


Subject(s)
Chloroprene/metabolism , Ethylene Oxide/analogs & derivatives , Liver/metabolism , Lung/metabolism , Subcellular Fractions/metabolism , Animals , Chloroprene/chemistry , Chloroprene/pharmacology , Cricetinae , Cytochrome P-450 Enzyme System/metabolism , Cytosol/enzymology , Epoxide Hydrolases/metabolism , Ethylene Oxide/chemical synthesis , Ethylene Oxide/metabolism , Glutathione Transferase/metabolism , Humans , Hydrolysis/drug effects , Kinetics , Liver/cytology , Liver/drug effects , Lung/cytology , Lung/drug effects , Male , Mesocricetus , Mice , Mice, Inbred Strains , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Biological , Oxidation-Reduction/drug effects , Rats , Rats, Inbred F344 , Rats, Wistar , Species Specificity , Subcellular Fractions/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...