key: cord-0002958-udbd5ip2 authors: Cichocki, Joseph A.; Furuya, Shinji; Venkatratnam, Abhishek; McDonald, Thomas J.; Knap, Anthony H.; Wade, Terry; Sweet, Stephen; Chiu, Weihsueh A.; Threadgill, David W.; Rusyn, Ivan title: Characterization of Variability in Toxicokinetics and Toxicodynamics of Tetrachloroethylene Using the Collaborative Cross Mouse Population date: 2017-05-30 journal: Environ Health Perspect DOI: 10.1289/ehp788 sha: 31fae2807695521804442fedcd3f7b9f17d0f9ea doc_id: 2958 cord_uid: udbd5ip2 BACKGROUND: Evaluation of interindividual variability is a challenging step in risk assessment. For most environmental pollutants, including perchloroethylene (PERC), experimental data are lacking, resulting in default assumptions being used to account for variability in toxicokinetics and toxicodynamics. OBJECTIVE: We quantitatively examined the relationship between PERC toxicokinetics and toxicodynamics at the population level to test whether individuals with increased oxidative metabolism are be more sensitive to hepatotoxicity following PERC exposure. METHODS: Male mice from 45 strains of the Collaborative Cross (CC) were orally administered a single dose of PERC ([Formula: see text]) or vehicle (Alkamuls-EL620) and euthanized at various time points ([Formula: see text] /strain/time). Concentration–time profiles were generated for PERC and its primary oxidative metabolite trichloroacetate (TCA) in multiple tissues. Toxicodynamic phenotyping was also performed. RESULTS: Significant variability among strains was observed in toxicokinetics of PERC and TCA in every tissue examined. Based on area under the curve (AUC), the range of liver TCA levels spanned nearly an order of magnitude ([Formula: see text]-fold). Expression of liver cytochrome P4502E1 did not correlate with TCA levels. Toxicodynamic phenotyping revealed an effect of PERC on bodyweight loss, induction of peroxisome proliferator activated receptor-alpha (PPAR [Formula: see text])-regulated genes, and dysregulation of hepatic lipid homeostasis. Clustering was observed among a) liver levels of PERC, TCA, and triglycerides; b) TCA levels in liver and kidney; and c) TCA levels in serum, brain, fat, and lung. CONCLUSIONS: Using the CC mouse population model, we have demonstrated a complex and highly variable relationship between PERC and TCA toxicokinetics and toxicodynamics at the population level. https://doi.org/10.1289/EHP788 Interindividual variability in susceptibility to toxicity has been observed or assumed to exist for all environmental chemicals and drugs; however, little empirical data are available to quantify such differences (Zeise et al. 2013) . Traditional in vivo toxicity testing relies on the use of a single strain of rodent (typically an inbred or hybrid strain), with multiple identical animals being used at each time point or in each dose group; thus, extrapolation from these data to a diverse human population is difficult and necessitates the use of default assumptions. In the past decade, the utility of a mouse population-based approaches to in vivo toxicity testing has been demonstrated for a number of toxic agents and the appreciation of genetics as an important dimension in science and practice of investigative pharmacology and toxicology is increasing (Rusyn et al. 2010) . Importantly, genetically heterogeneous mouse models have been combined with the limited data for interindividual differences in chemical toxicity from human studies to characterize the extent of the variability , discover genetic determinants (Harrill et al. 2009 ), and understand the molecular underpinnings of toxicity (French et al. 2015) . Recent advances in mouse genetics have led to the development of highly diverse mouse populations, one of which is known as the Collaborative Cross (CC) (Churchill et al. 2004 ). The CC is a large panel of recombinant inbred strains that were derived from eight inbred founder strains (Collaborative Cross Consortium 2012; Threadgill et al. 2011 ). This new resource has been used to make seminal discoveries in biomedical science (Crowley et al. 2015; Phillippi et al. 2014 ) and enabled creation of mouse models for diseases that were thought not to exist in the mouse (Gralinski et al. 2015; Rasmussen et al. 2014) . One of the most ubiquitous environmental pollutants without solid experimental data on variability in toxicokinetics and toxicodynamics is tetrachloroethylene (perchloroethylene, PERC). PERC is a chlorinated olefin solvent with a variety of industrial applications, a ubiquitous contaminant of groundwater, soil, ambient and urban air, and is one of the most common pollutants present in many hazardous waste sites (National Research Council 2009a) . The most well-known application of PERC is in dry-cleaning. Of the approximately 32,000 dry cleaners in the United States operating as of 2006, about 28,000 ( ∼ 88%) were using PERC as a solvent (Goehl and O'Neil 2005) . Not only are workers exposed to PERC via dry-cleaning, but consumers are also potentially exposed via "off-gassing" of PERC from treated clothing (Sherlach et al. 2011) . PERC is therefore of great concern to public health protection agencies, including the U.S Environmental Protection Agency (EPA) and the International Agency for Research on Cancer (IARC). PERC has been classified by IARC (2013) and the U.S. EPA (2011b) as a possible human carcinogen, mainly based on evidence for carcinogenesis from chronic studies in rodents (National Toxicology Program 1986) . PERC exposure is also associated with noncancer toxicity in a number of tissues (U.S. EPA 2011b) . The organ-specific adverse effects associated with exposure to PERC and other structurally similar olefins, such as trichloroethylene (TCE), are consequences of tissue-specific metabolism (Cichocki et al. 2016; Lash and Parker 2001; Lash et al. 2014) , which involves both cytochrome P450s (CYPs) and glutathione (GSH) S-transferases (GSTs). The oxidative metabolism of PERC results in the formation of trichloroacetate (TCA), a chemical with suggestive evidence of carcinogenic potential based on significantly increased incidences of liver tumors in mice (U.S. EPA 2011a) . Because TCA is a major metabolite of PERC, it is thought to be critical to establishing an exposure-response relationship for PERC and toxicity (Lash and Parker 2001) . Furthermore, it is estimated that the contribution of TCA to PERC-associated liver cancer in rodents can be as little as 12% to as much as 100% (U.S. EPA 2011b) . TCA itself provides a useful marker of oxidative metabolism, which has been used as a dose metric for risk assessment extrapolation from mice to humans (U.S. EPA 2011b) . Numerous recent hazard and risk assessments pointed to a critical need for better characterization of toxicokinetics, toxicodynamics, and population variability of PERC in order to improve public health protection (Guha et al. 2012; National Research Council 2010; U.S. EPA 2011b) . Although there have been previous attempts at characterizing toxicokinetic variability of PERC Covington et al. 2007; Gelman et al. 1996) . Chiu and Ginsberg (2011) found that all these efforts were hampered by inadequate experimental data, leading to substantial uncertainty in the ultimate risk assessments. Moreover, variability exists in both toxicokinetics and toxicodynamics, and may be a consequence of multiple factors, including genetics. Therefore, the contribution of such factors to population level variability in both toxicokinetic and toxicodynamic responses to PERC deserves further attention. We used a genetically diverse mouse population of 45 CC mouse strains to quantify the extent of interstrain variability in response to exposure to a single high dose of PERC. We determined whether individuals with increased hepatic TCA levels are more sensitive to the hepatotoxic effects of PERC. Mice were exposed to a single dose of PERC and were sampled at multiple time points to generate population-level concentration-time profiles for PERC and TCA across multiple tissues. Toxicodynamic phenotyping included study of the expression of CYPs, dysregulation of hepatic lipid metabolism, and histopathological assessment of the liver. PERC was purchased from Sigma Aldrich (St. Louis, MO; Catalog #270,393-100ML; Lot #SHBD9374V). Alkamuls-EL620 was acquired from Solvay (Deptford, NJ; Lot #SP8J26X07). All reagents used were of the highest purity available. Adult (6-8 weeks of age) male mice from 45 strains of the Collaborative Cross were acquired from the Systems Genetics Core at the University of North Carolina (Chapel Hill, NC) or from a colony maintained at Texas A&M University. All animals were acquired between 6 January 2015 and 20 February 2015. Exact dates of birth, dates of acquisition, and original IDs (if applicable) are available online on the Mouse Phenome Database (see "Data Availability" subsection, below). Strain nomenclature is used as suggested by the nomenclature committee (Collaborative Cross Consortium 2012). Of the 45 strains exposed to PERC, one animal from one strain (CC059/TauUnc) died in the 24-hr treatment group as a result of a gavage error. Comparisons between time-matched vehicle-and PERC-exposed individuals within a strain was required for data interpretation, toxicodynamic data are only presented for 44/45 strains. Animals were housed in disposable HEPA-filtered polycarbonate cages with hardword chip bedding (Sanichip, P.J. Murphy Forest Products, Bowling Green, KY) with access to food (Teklad #8,604, Envigo, Indianapolis, IN) and purified water ad libitum. The animal room was maintained on a 12-hr light/dark cycle. Animals were allowed to acclimate to the room for at least 10 days prior to beginning experimentation. All experiments were approved by the Institutional Animal Care and Use Committee of Texas A&M University. For all animal studies, exposures were conducted between 0800 and 1100 hours to limit potential diurnal variation in PERC metabolism. The in-life portion of the study was conducted over a 6-week time frame to limit seasonal variations. Figure 1 shows a schematic of the overall experimental design. Mice were administered a single dose of PERC (1,000 mg/kg) or vehicle (5% Alkamuls EL-620 in saline) by oral gavage (i.g.; 10 mL/kg). All dosing solutions were prepared fresh daily (and used within 1.5 hr of preparation) in amber glass vials with foil-lined septa to minimize adsorption of PERC to the container walls and/ or septum. For PERC-treated animals, the time points for tissue collection were 1, 2, 4, 12, and 24 hr postgavage (n = 1= strain=time point); vehicle-treated mice were euthanized only at the 24-hr time point (n = 1=strain). Prior to necropsy, animals were weighed and were deeply anesthetized by intraperitoneal (i.p.) injection of EuthasolE (3 mL/kg; Med-Vet International, Mettawa, IL) and euthanasia was performed via exsanguination through the vena cava. Serum was prepared by centrifugation using Z-gel tubes (Starstedt; Numbrecht, Germany). All other tissues were removed, rinsed in phosphate-buffered saline, and weighed. The left lobe of the liver was separated from the other Figure 1 . Study design consisted of two groups (PERC and vehicle) and five time points (1, 2, 4, 12, and 24 hr postdosing). lobes after weighing. The kidneys, lungs, brain, and epididymal fat pads were collected and snap-frozen in liquid nitrogen. For animals in the 24-hr group, small sections of left and median liver lobes and kidney were fixed in 10% neutral-buffered formalin, embedded in paraffin, sectioned at 5 lm, and stained with hematoxylin & eosin (H&E). A detailed protocol for the analysis of PERC and TCA in tissues is provided in the Supplemental Material, "Materials and Methods." Briefly, PERC was analyzed in methanolic tissue extracts via headspace GC/MS. TCA was analyzed via GC/MS after derivatization to its methyl ester based on a modified U.S. EPA method (EPA 815-B-03-002; Domino et al. 2003 ). Based upon TCA analysis in liver, kidney, and serum, tissues from nine additional strains were analyzed for TCA levels in brain, gonadal fat pad, and lung tissue. These strains were selected by stratifying strains in quartiles based on TCA AUC levels in liver, kidney, and serum, and then using the randomLHS function of the lhs R package. Unique strains were then selected from each box of the Latin hypercube. A detailed protocol for qRT-PCR is provided in the Supplemental Material, "Materials and Methods." Briefly, qRT-PCR was performed on 50 ng of cDNA from the liver (left lobe) with Taqman V R probes targeting Acyl-CoA oxidase 1 (Acox1) and Cyp4a10. b-glucuronidase (GusB) was used as the housekeeping gene. A detailed protocol for Western blotting of hepatic CYP2E1 is provided in the Supplemental Material, "Materials and Methods." Briefly, 45 lg of hepatic protein was immunoblotted for CYP2E1 and b-actin (loading control) using standard sodium dodecylsulfate polyacrylamide gel electrophoresis techniques. Triglycerides were measured in liver (left lobe) and serum using a commercially available colorimetric kit (Dako, Carpinteria, CA) according to the manufacturer's instructions. GraphPad Prism (La Jolla, CA) was used to calculate areas under the curves (AUCs) for all analytes and to perform paired and unpaired t-tests between vehicle-and PERC-treated groups. R (v.3.1.2) was used for fitting toxicokinetic data to a nonlinear function (nls), to generate heatmaps (gplots) and boxplots (ggplot2). For all tests, a p < 0:05 was required for statistical significance. All raw data have been made publically available by uploading files into the Mouse Phenome Database (http://phenome.jax.org; MPD: Rusyn7). A single dose of PERC resulted in bodyweight loss in ∼ 80% of strains, whereas liver-to-bodyweight ratios remained largely unaffected by PERC exposure (Figure 2 ). On average, a significant loss of body weight by ∼ 6% was observed (p < 0:0001, t-test), although the response was variable between strains. For instance, strains CC043/GeniUnc, CC065/Unc, and CC001/Unc lost over 15% of their body weight, whereas other strains (e.g., CC012/GeniUnc) maintained their body weight. The liver-tobodyweight ratio was highly variable in both vehicle-and PERCtreated animals. The median liver-to-bodyweight ratio was lower in PERC-treated animals compared with vehicle-treated animals, although this was not a statistically significant observation (p = 0:282, unpaired t-test). Significant accumulation (p = 0:0006, t-test) of triglycerides was observed in the livers of PERC-exposed animals ( Figure 3A , B); this finding was confirmed histopathologically (see Figure S1 ). About one-third of the strains tested exhibited various degrees of hepatosteatosis 24 hr following PERC exposure. Interestingly, the degree and type of steatosis varied among strains; some mice displayed zonal steatosis, others displayed azonal, and both macro-and micro-vesicular (and mixed) steatosis was observed. The accumulation of hepatic lipids was coincident with a decrease (p = 0:013, t-test) in serum triglyceride levels ( Figure 3C ,D), providing additional evidence that PERC exposure, even at a single dose, is coincident with dysregulation of liver lipid metabolism. The levels of PERC and its primary oxidative metabolite TCA were assessed in a standard toxicokinetic study design. Levels of PERC in the liver and kidney (Figure 4 ; see also Table S1) were highly variable across the population (geometric standard deviation ðGSDÞ = 1:65 and 1.74, respectively), as highlighted by the broad 95% confidence intervals (gray shadow) around the nonlinear least squares regression line (solid black line). It is also evident that in both tissues, the shape of the concentration-time profiles in the individual strains was also variable. Similar to PERC, tissue levels of TCA and shapes of the concentration-time profiles were strain-dependent ( Figure 5 ; see also Table S2 ). On average, TCA levels were about 3-fold higher than PERC on a molar basis. Levels of TCA were highest in serum, followed by the liver and kidney. In the liver, the geometric mean (GM) was ∼ 13,600 nmol à hr=g liver (GSD = 1:64). Although most of the strains either exhibit a plateau or clearance from all tissue compartments by the 24 hr time point, some strains appear to accumulate TCA with time. This type of toxicokinetic behavior yields under-prediction of the maximum TCA concentration. Due to this limitation, there is greater uncertainty in peak TCA concentrations for these strains and therefore the actual GM and GSD statistics are likely greater than those reported here. PERC is structurally similar to trichloroethylene (TCE) and these compounds share qualitatively similar metabolism pathways. Thus, we compared the interstrain differences in serum TCA levels of this study (after exposure to PERC) and the serum TCA levels from (Bradford et al. 2011) , in which a single oral dose of 2,100 mg/kg TCE in corn oil was administered to 16 strains of male inbred mice. Interstrain variability in TCA levels was similar between two studies (Bradford et al. 2011 : GM ∼ 9,400; GSD = 1:42; this study: GM ∼ 29,500; GSD = 1:37); however, TCA AUCs in the serum of PERCexposed mice was much greater as compared with TCE-exposed mice even though the dose of TCE was over 2.5-fold higher on a molar basis. Thus, in spite of qualitatively similar metabolism, the conversion of PERC and TCE to TCA in vivo is quantitatively different. A subset of nine strains was selected for an in-depth profiling of TCA levels across additional tissues, as described in "Methods." In addition to liver, kidney, and serum, TCA was analyzed in gonadal (epididymal) fat pads, lung, and brain (see Figure S2 ). Although a large degree of variability existed among individual strains, the general trend for TCA levels was serum