Elsevier

Toxicology

Volume 409, 1 November 2018, Pages 33-43
Toxicology

Comparative analysis of metabolism of trichloroethylene and tetrachloroethylene among mouse tissues and strains

https://doi.org/10.1016/j.tox.2018.07.012Get rights and content

Abstract

Trichloroethylene (TCE) and tetrachloroethylene (PCE) are structurally similar chemicals that are metabolized through oxidation and glutathione conjugation pathways. Both chemicals have been shown to elicit liver and kidney toxicity in rodents and humans; however, TCE has been studied much more extensively in terms of both metabolism and toxicity. Despite their qualitative similarities, quantitative comparison of tissue- and strain-specific metabolism of TCE and PCE has not been performed. To fill this gap, we conducted a comparative toxicokinetic study where equimolar single oral doses of TCE (800 mg/kg) or PCE (1000 mg/kg) were administered to male mice of C57BL/6J, B6C3F1/J, and NZW/LacJ strains. Samples of liver, kidney, serum, brain, and lung were obtained for up to 36 h after dosing. For each tissue, concentrations of parent compounds, as well as their oxidative and glutathione conjugation metabolites were measured and concentration-time profiles constructed. A multi-compartment toxicokinetic model was developed to quantitatively compare TCE and PCE metabolism. As expected, the flux through oxidation metabolism pathway predominated over that through conjugation across all mouse strains examined, it is 1,200–3,800 fold higher for TCE and 26–34 fold higher for PCE. However, the flux through glutathione conjugation, albeit a minor metabolic pathway, was 21-fold higher for PCE as compared to TCE. The degree of inter-strain variability was greatest for oxidative metabolites in TCE-treated and for glutathione conjugation metabolites in PCE-treated mice. This study provides critical data for quantitative comparisons of TCE and PCE metabolism, and may explain the differences in organ-specific toxicity between these structurally similar chemicals.

Introduction

Trichloroethylene (TCE) and tetrachloroethylene (PCE) are structurally similar chlorinated olefins that are used in chemical manufacture, metal degreasing, and other industrial applications (U.S. EPA, 2011a, U.S. EPA, 2011b). TCE and PCE are high production volume chemicals and are ubiquitous in the environment (IARC, 2014). Humans can be exposed to these chemicals via inhalation and ingestion (ATSDR, 1997; Wu and Schaum, 2000). In a National Health and Nutrition Examination Survey (2013–2014), the rate of detection for TCE and PCE in blood was 0.6% for TCE and 7.4% for PCE (CDC, 2017). TCE and PCE are still prioritized for evaluation of the risks to human health and environment (U.S. EPA, 2017).

There are differences in toxic effects of TCE and PCE in liver, kidney and other tissues (Cichocki et al., 2016). TCE is classified as “carcinogenic to humans” by US EPA (U.S. EPA, 2011b) and IARC (Guha et al., 2012); while PCE is classified as “likely to be a human carcinogen” by US EPA (U.S. EPA, 2011a) and as “probably carcinogenic to humans” by IARC (Guha et al., 2012). A comparative study of toxicodynamics of TCE and PCE in mice showed differences in the effects on liver and kidney, PCE perturbed more molecular pathways in mouse liver and kidney as compared to TCE (Zhou et al., 2017). However, there are no published reports of comparative analysis of toxicokinetics of TCE and PCE.

Upon absorption, TCE and PCE are metabolized through oxidative and glutathione conjugation pathways (Cichocki et al., 2016). Initial oxidation occurs on the double bond by cytochrome P450 s (CYPs) to generate an epoxide, which can be further metabolized. Trichloroacetic acid (TCA) is a major oxidative metabolite of both TCE and PCE, and is a common urinary biomarker of exposure (Forkert et al., 2003; Volkel et al., 1998). The other oxidative metabolite, trichloroethanol (TCOH), is a TCE-specific metabolite that is formed through oxidation of TCE to chloral hydrate (CH), while PCE oxidation occurs through trichloroacetyl chloride (Chiu et al., 2007). Both TCE and PCE can enzymatically conjugate with glutathione to form dichloro- or trichloro-glutathione conjugates (DCVG or TCVG) (Lash et al., 2000). These can be further metabolized via hepatic or renal gamma-glutamyl transferase and di-peptidase to form corresponding cysteine conjugates, DCVC or TCVC, which are then n-acetylated via N-acetyltransferase to generate NAcDCVC or NAcTCVC, respectively. In addition, both NAcDCVC and NAcTCVC can be deacetylated via acylase to yield DCVC or TCVC, respectively. Apart from N-acetylation, DCVC and TCVC can be further bio-activated via cysteine conjugate β lyase to generate reactive thioketenes, or flavin-containing monooxygenase to form corresponding sulfoxides (Lash et al., 2014). These and other reactive species derived from glutathione conjugation are thought to be significant contributors to the nephrotoxicity of TCE and PCE (Lash et al., 2001a, Lash et al., 2003).

Quantitative estimation of inter-individual variability in metabolism is also a critical challenge in human health assessments of TCE and PCE (Cichocki et al., 2017b, Cichocki et al., 2016; Venkatratnam et al., 2017). The inter-strain variability in TCE metabolism has been characterized by using a multi-strain panel of inbred mice (Bradford et al., 2011) and the Collaborative Cross mouse population (Luo et al., 2018a; Venkatratnam et al., 2017, Venkatratnam et al., 2018). Mouse population-derived variability estimates for TCE metabolism closely matched population variability estimates previously derived from human toxicokinetic studies with TCE (Chiu et al., 2014). Likewise, the inter-individual variability in PCE metabolism has also been studied in the Collaborative Cross mouse population (Cichocki et al., 2017b), and a nonalcoholic steatohepatitis mouse model (Cichocki et al., 2017a). However, these studies of inter-strain variability in metabolism and toxicity were conducted separately for TCE or PCE and using doses that were not equivalent. Because of the close structural similarity of these chemicals and paucity of the available toxicokinetic data, a comparative study of equimolar doses was conducted concurrently using three inbred strains, selected based on variability observed across strains with respect to oxidative and glutathione conjugation metabolism for TCE (Bradford et al., 2011). The data from this study fill critical gaps in our understanding of the quantitative differences in TCE and PCE toxicokinetics.

Section snippets

Chemicals

TCE (PN: 24254, ≥99%), PCE (PN: 270393, ≥99%), TCA (PN: T6399, ≥ 99%), TCOH (PN: T54801, ≥99%), 2-bromobutyric acid (PN: 147877, 97%), ethylbenzene (PN: E12508, 99%), methyl tert-butyl ether (PN: 443808, ≥99%), chloroform (PN: 650498, ≥99%), sulfuric acid (PN: 339741, ≥99%), sodium sulfate (PN: 239313, ≥ 99%), β-glucuronidase (PN: G0751, ≥300,000 units/ g solid), and sodium bicarbonate (PN: S6014, ≥99%) were obtained from Sigma Aldrich (St Louis, MO). Methanol (HPLC grade) was from Fischer

Results

Levels of parent compounds (TCE and PCE), as well as their oxidative (TCA and TCOH) and glutathione conjugation metabolites (DCVG, DCVC, NAcDCVC, TCVG, TCVC, and NAcTCVC) [see comparative schematics of metabolism for TCE and PCE in (Cichocki et al., 2016; Luo et al., 2018b)] were quantified in liver, kidney, brain, lung and serum of male mice of three strains across a range of time points. These data were used to develop toxicokinetic models for TCE and PCE metabolism.

Discussion

The most notable finding of this comparative study of toxicokinetics of TCE and PCE in mice is the observation of considerable differences in metabolism between these structurally-similar compounds. A larger portion of the parent compound undergoes metabolism in TCE-treated mice (15.7%–38.3%) as compared to PCE-treated mice (6.6%–9.7%), a finding that provides empirical data in strong support of the estimates from PBPK models (Chiu et al., 2014; Chiu and Ginsberg, 2011). The more efficient

Conclusions

In summary, this study provides a comparative analysis for the metabolisms between TCE and PCE. We show that one atom replacement of chlorine can substantially affect the metabolism via both oxidative and glutathione conjugation pathways. The qualitative and quantitative differences between TCE and PCE metabolites, as well as the tissue-specific distribution of metabolites, can shed light on the differences between TCE- and PCE-induced toxicities.

Acknowledgements

The authors wish to show gratitude to Drs. Anthony H. Knap, Terry Wade, Stephen Sweet, and Thomas J. McDonald at Texas A&M University for providing access to the analytical instrumentation. The authors also thank Dr. Chimeddulam Dalaijamts for fruitful discussions. This work was supported, in part, by grants from the U.S. EPA (STAR RD83561202) and National Institutes of Health (F32 ES026005), and institutional support from Texas A&M University.

References (61)

  • L.H. Lash et al.

    Apoptosis, necrosis, and cell proliferation induced by S-(1,2-dichlorovinyl)-l-cysteine in primary cultures of human proximal tubular cells

    Toxicol. Appl. Pharmacol.

    (2001)
  • L.H. Lash et al.

    Renal toxicity of perchloroethylene and S-(1,2,2-trichlorovinyl)glutathione in rats and mice: sex- and species-dependent differences

    Toxicol. Appl. Pharmacol.

    (2002)
  • L.H. Lash et al.

    Modulation of hepatic and renal metabolism and toxicity of trichloroethylene and perchloroethylene by alterations in status of cytochrome P450 and glutathione

    Toxicology

    (2007)
  • L.H. Lash et al.

    Trichloroethylene biotransformation and its role in mutagenicity, carcinogenicity and target organ toxicity

    Mutat. Res. Rev. Mutat. Res.

    (2014)
  • E.A. Lock et al.

    Trichloroethylene-induced formic aciduria in the male C57 Bl/6 mouse

    Toxicology

    (2017)
  • B.K. Philip et al.

    Impact of repeated exposure on toxicity of perchloroethylene in Swiss webster mice

    Toxicology

    (2007)
  • A.M. Schumann et al.

    The pharmacokinetics and macromolecular interactions of perchloroethylene in mice and rats as related to oncogenicity

    Toxicol. Appl. Pharmacol.

    (1980)
  • J.Z. Song et al.

    Simultaneous detection of trichloroethylene alcohol and acetate in rat urine by gas chromatography-mass spectrometry

    J. Chromatogr. B Analyt. Technol. Biomed. Life Sci.

    (2003)
  • V.S. Vaidya et al.

    Renal injury and repair following S-1, 2 dichlorovinyl-l-cysteine administration to mice

    Toxicol. Appl. Pharmacol.

    (2003)
  • W. Volkel et al.

    Biotransformation of perchloroethene: dose-dependent excretion of trichloroacetic acid, dichloroacetic acid, and N-acetyl-S-(trichlorovinyl)-l-cysteine in rats and humans after inhalation

    Toxicol. Appl. Pharmacol.

    (1998)
  • N. Yaqoob et al.

    Trichloroethylene-induced formic aciduria: effect of dose, sex and strain of rat

    Toxicology

    (2013)
  • N. Yaqoob et al.

    Trichloroethylene and trichloroethanol-induced formic aciduria and renal injury in male F-344 rats following 12 weeks exposure

    Toxicology

    (2014)
  • ATSDR

    Toxicological Profile for Trichloroethylene (TCE), Agency for Toxic Substances and Disease Registry

    (1997)
  • U. Bernauer et al.

    Biotransformation of trichloroethene: dose-dependent excretion of 2,2,2-trichloro-metabolites and mercapturic acids in rats and humans after inhalation

    Arch. Toxicol.

    (1996)
  • G. Birner et al.

    Biotransformation, excretion and nephrotoxicity of haloalkene-derived cysteine S-conjugates

    Arch. Toxicol.

    (1997)
  • B.U. Bradford et al.

    Interstrain differences in the liver effects of trichloroethylene in a multistrain panel of inbred mice

    Toxicol. Sci.

    (2011)
  • J.C. Caldwell et al.

    Key issues in the modes of action and effects of trichloroethylene metabolites for liver and kidney tumorigenesis

    Environ. Health Perspect.

    (2006)
  • CDC

    National Health and Nutrition Examination Survey 2013–2014

    (2017)
  • W.A. Chiu et al.

    Issues in the pharmacokinetics of trichloroethylene and its metabolites

    Environ. Health Perspect.

    (2006)
  • W.A. Chiu et al.

    Toxicokinetics of inhaled trichloroethylene and tetrachloroethylene in humans at 1 ppm: empirical results and comparisons with previous studies

    Toxicol. Sci.

    (2007)
  • Cited by (13)

    • Model systems and organisms for addressing inter- and intra-species variability in risk assessment

      2022, Regulatory Toxicology and Pharmacology
      Citation Excerpt :

      For the parent molecule and its primary oxidative metabolite trichloroacetic acid, the observed range (max to min) of variability across ∼50 strains was about 8-fold (Cichocki et al., 2017), but for glutathione conjugation metabolites it was up to 50-fold (Luo et al., 2019). In addition, Collaborative Cross population provided a better characterization for the population variability in toxicokinetics of tetrachloroethylene as compared to study of only three inbred strains (Luo et al., 2018b). The three strains exhibited oxidative metabolism of tetrachloroethylene at the lower end of the broader Collaborative Cross population, while glutathione conjugation was in the middle; these data demonstrate the validity of a concern that a single or few strains may not be representative of a larger population responses (Chiu and Rusyn, 2018).

    • Toxicity assessments of selected trichloroethylene and perchloroethylene metabolites in three in vitro human placental models

      2022, Reproductive Toxicology
      Citation Excerpt :

      Since DCA has not been measured in human serum or urine [35,67], we used the same large range of concentrations for DCA as TCA. Although more recent studies have measured TCE and PERC metabolites in mouse samples using advanced technologies [66], [82,83], differences between rodent and human metabolism of TCE and PERC [6,34] limit the direct relevance of metabolite concentrations derived from mice to our study. The current study is limited by the use of in vitro cultured cell lines and tissues to model the placenta, which may not fully represent biological interactions that occur in vivo.

    • Bioactivation of trichloroethylene to three regioisomeric glutathione conjugates by liver fractions and recombinant human glutathione transferases: Species differences and implications for human risk assessment

      2021, Toxicology Letters
      Citation Excerpt :

      The reason why the previous studies failed to detect different regioisomers of DCVG may be the fact that different chromatographic conditions are used with different HPLC-columns and steep gradients resulting in relatively short retention times. For example, using the LCMS/MS method used by Rusyn’s group DCVC, DCVG and DCV-NAC elute at 2.50, 3.35 and 3.75 min, respectively (Luo et al., 2018a,b). These short retention times probably do not allow chromatographic separation of the regioisomers.

    View all citing articles on Scopus
    1

    These authors contributed equally to this manuscript.

    View full text