Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jul 21.
Published in final edited form as: Curr Opin Environ Sci Health. 2022 Aug 15;29:100389. doi: 10.1016/j.coesh.2022.100389

Oxidative stress, glutathione, and CYP2E1 in 1,4-dioxane liver cytotoxicity and genotoxicity: insights from animal models

Yewei Wang 1, Georgia Charkoftaki 1, Emily Davidson 1,2, David J Orlicky 3, Robyn L Tanguay 4, David C Thompson 1, Vasilis Vasiliou 1, Ying Chen 1,*
PMCID: PMC10361651  NIHMSID: NIHMS1865657  PMID: 37483863

Abstract

1,4-Dioxane (DX) is an emerging drinking water contaminant worldwide, which poses a threat to public health due to its demonstrated liver carcinogenicity and potential for human exposure. The lack of drinking water standards for DX is attributed to undetermined mechanisms of DX carcinogenicity. This mini-review provides a brief discussion of a series of mechanistic studies, wherein unique mouse models were exposed to DX in drinking water to elucidate redox changes associated with DX cytotoxicity and genotoxicity. The overall conclusions from these studies support a direct genotoxic effect by high dose DX and imply that oxidative stress involving CYP2E1 activation may play a causal role in DX liver genotoxicity and potentially carcinogenicity. The mechanistic data derived from these studies can serve as important references to refine the assessment of carcinogenic pathways that may be triggered at environmentally relevant low doses of DX in future animal and human studies.

Keywords: Water contaminants, liver carcinogenicity, mechanism of action, oxidative DNA damage

Introduction

1,4-Dioxane (DX) is a synthetic chemical stabilizer of chlorinated solvents, such as 1,1,1-trichloroethane (TCA) and trichloroethylene (TCE) [1]. Improper disposal of these industrial solvents is the primary source of DX release into the environment [15]. Due to its high water solubility, low degradability and poor adsorption, DX is persistent in the environment, being transported predominantly via water. Consequently, DX contamination of water bodies, including surface water and ground water, has been reported across the US and worldwide [6,7]. To date, there is a lack of human studies assessing the toxicity associated with oral DX exposure; however, studies in laboratory animals demonstrate that chronic DX exposure in drinking water induces liver adenomas and adenocarcinomas [8,9].

Despite concerns regarding its carcinogenic potential and the widespread contamination in the US, DX in drinking water is not regulated by the federal government and state-level regulatory guidelines vary greatly. This is, in part, due to the undetermined mechanisms of action (MOAs) of DX carcinogenicity, which represents a significant knowledge gap in DX risk assessment [10]. The mutagenic MOA of DX has been investigated and remains inconclusive, due to a lack of mutagenic activity in vitro and evidence of liver genotoxicity in vivo [10,11]. Utilizing several unique mouse models, our group performed a series of DX exposure studies involving low to high DX doses and short-term to subchronic exposures [12,13]. The objective of this mini-review is to summarize the main findings from these mechanistic studies and discuss new lines of experimental evidence that support a direct genotoxic effect by high dose DX and a potential causal role of oxidative stress in this process.

Multi-omics study in BDF-1 female mice reveals altered oxidative stress defense systems accompanying DNA damage as early changes by oral exposure to high dose DX

Oxidative stress occurs when reactive oxygen species (ROS) are produced at levels exceeding those capable of being sequestered by cellular antioxidant defense systems, which comprise small molecule antioxidants (e.g. glutathione (GSH)), antioxidant enzymes (e.g. glutathione S-transferases (GSTs)) and their regulators (e.g. nuclear factor erythroid 2-related factor 2 (NRF2)) [14]. Many drugs, environmental toxicants and selected dietary components cause liver toxicity by inducing oxidative stress [1517]. Hepatic oxidative damage can result in non-neoplastic lesions (cytotoxicity), such as fatty liver (steatosis), hepatocyte death and inflammation [18], as well as preneoplastic and neoplastic lesions (carcinogenicity) [19]. Oxidative stress is an important mutagenic mechanism for numerous chemical carcinogens [20,21]. Prior to our studies, the involvement of oxidative stress in DX liver toxicity had been investigated in a limited number of in vivo studies (Table 1). Mnaa et al. reported increased lipid peroxidation in the livers of rats exposed to DX at 100 mg/kg/day in drinking water for 6 weeks [22]. Gi et al. reported no difference in liver 8-hydroxy-2’ -deoxyguanosine (8-OHdG) levels (an index of oxidative DNA damage) measured by HPLC-ECD in male gpt delta transgenic F344 rats following DX exposures up to 5000 ppm in drinking water for 16 weeks [23]. However, the same research group later identified that 8-oxo-dG (a form of oxidative DNA modification) was one of the three DNA adducts characteristic of DX treatment in livers from male F344 rats by 200 ppm or 5000 ppm DX for 16 weeks [24]. In this later study, DNA adducts were analyzed using more advanced HPLC-QTOF-MS.

Table 1.

In Vivo 1,4-Dioxane Exposure Studies Investigating Oxidative Stress and Genotoxicity in the Liver

Animal Model DX Exposure Regime Main Findings Reference
Female Sprague-Dawley rats 100 mg/kg in drinking water for 6 weeks Liver cytotoxicity
 • Histopathology (portal edema, Kupffer cells activation, small focal hepatic necrosis associated with inflammatory cells infiltration)
 • Increased serum ALP
Liver genotoxicity
 • No data reported
Oxidative stress
 • Increased marker of lipid peroxidation (MDA)
 • Reduced catalase activity
Mnaa et al. (2016) [22]
Male gpt delta transgenic F344 rats 0.2, 2, 20, 200, 1000 or 5000 ppm in drinking water for 16 weeks Liver cytotoxicity
 • No treatment-related histopathology
Liver genotoxicity
 • Increased A:T- to -T:A transversion frequency by 1000 ppm
 • Increased gpt mutation frequency, A:T- to -G:C transition and A:T- to -T:A transversion frequencies by 5000 ppm
 • Increased mRNA level of DNA repair enzyme MGMT by 5000 ppm
Liver Proliferation/preneoplastic lesion
 • Increased mRNA level of PCNA by 5000 ppm
 • Increased GST-P-positive foci by 5000 ppm
Oxidative Stress
 • No change in mRNA levels of 12 CYP enzymes
 • No difference in 8-OHdG levels (measured by HPLC-ECD)
Gi et al. (2018) [23]
Male WT F344 rats 2, 20, 200, 2000, or 5000 ppm in drinking water for 16 weeks Liver cytotoxicity
 • No treatment-related histopathology
Liver Proliferation/preneoplastic lesion
 • Increased GST-P-positive foci by 2000 and 5000 ppm
 • Increased BrdU labeling index by 5000 ppm
Gi et al. (2018) [23]
Male WT F344 rats 20, 200, or 5000 ppm in drinking water for 16 weeks (Liver samples from Gi et al. 2018 were used for analyses)Liver genotoxicity
 • Identified three DNA adducts (measured by HPLC-QTOF-MS) characteristic of DX treatment by 200 and 5000 ppm
Oxidative Stress
 • 8-oxo-dG is one of the three treatment-specific DNA adducts by 200 and 5000 ppm
Totsuka et al. (2020) [24]
Female BDF-1 mice 50, 500, and 5000 ppm in drinking water for 1 or 4 weeks Liver cytotoxicity
 • No gross histopathological change
Liver genotoxicity
 • Increased γH2AX-positive hepatocytes (DNA damage marker) by 5000 ppm at 1 and 4 weeks
Oxidative Stress
 • DEGs (measured by RNASeq analysis) enriched in GSH-mediated detoxification and NRF2-mediated oxidative stress response pathways by 5000 ppm at 4 weeks
 • DAMs (measured by metabolomics analysis) enriched in metabolic pathways of nucleotide salvage synthesis and oxidative stress response by 5000 ppm at 4 weeks
Charkoftaki et al. (2021) [12]
Male B6 GclmKO and
WT mice
1000 mg/kg/day by oral gavage for 1 week or 5000 ppm in drinking water for 12 weeks Liver cytotoxicity (no difference between GclmKO and WT mice)
 • Mild histopathology (single cell death and associated inflammation, ductal reactive change, and steatosis) at 12 weeks
 • 2-fold increase in plasma ALT and AST activities at 12 weeks
Liver genotoxicity (enhanced in GclmKO relative to WT mice)
 • Increased 8-OHdG levels (by ELISA assay) at 12 weeks
 • Elevated γH2AX/H2AX ratio (DNA damage repair marker) at 12 weeks
Oxidative Stress (enhanced in GclmKO relative to WT mice)
 • Transient induction of some NRF2-target antioxidant genes at 1 week
 • Reduced GSH/GSSG ratio (index of oxidative stress) at 12 weeks
 • Progressive induction of CYP2E1 and lipid peroxidation
Chen et al. (2022) [13]
Male B6 Cyp2e1KO mice 5000 ppm in drinking water for 1 week or 12 weeks Liver cytotoxicity
 • No treatment-related histopathology at 12 weeks
 • No change in plasma ALT and AST activities at 12 weeks
Liver genotoxicity
 • Trend of increase in 8-OHdG levels (by ELISA assay) at 12 weeks
 • Decreased γH2AX/H2AX ratio (DNA damage repair marker) at 12 weeks
Oxidative Stress
 • No induction of NRF2-targeted antioxidant genes
 • No change in GSH/GSSG ratio (index of oxidative stress)
 • No induction of lipid peroxidation
Wang et al.
(Manuscript in preparation)

Abbreviations: 8-OHdG, 8-hydroxy-2′-deoxyguanosine; ALP, alkaline phosphatase; ALT, alanine transaminase; AST, aspartate aminotransferase; BrdU, bromodeoxyuridine; CYP, cytochrome P450; CYP2E1, cytochrome P450 2E1; DAMs, differentially abundant metabolites; DEGs, differentially expressed genes; DX, 1,4-dioxane; ELISA, enzyme-linked immunosorbent assay; Gclm, glutamate-cysteine ligase modifier subunit; GSH, glutathione; GSSG, glutathione disulfide; GST-P, glutathione S-transferase placental-form; H2AX, H2A histone family member X; γH2AX, phosphorylation of the Ser-139 residue of H2AX; HPLC-ECD, high performance liquid chromatography with electrochemical detection; HPLC-QTOF-MS, high-performance liquid chromatography-quadrupole time-of-flight mass spectrometer; MDA, malondialdehyde; MGMT, O6-methylguanine-DNA methyltransferase; NRF2, nuclear factor erythroid 2-related factor 2; PCNA, proliferating cell nuclear antigen; WT, wild type.

We performed a short-term exposure study in female BDF-1 mice using a range of DX doses (50, 500 or 5000 ppm) in drinking water for one or four weeks (Table 1) [12]. This study aimed to capture early molecular changes by DX in the liver by transcriptomic and metabolomic profiling along with histopathological examination. We chose the BDF-1 mouse strain because it has shown consistent evidence of liver tumors in the Kano et al. chronic study [25]. Livers from DX-exposed BDF-1 mice showed no gross pathological changes at any exposure dose or duration. However, the percentage of hepatocytes stained positively for phosphorylation of the Ser-139 residue of the histone variant H2AX (γH2AX), a marker of DNA damage, were elevated following one or four weeks of exposure to high dose DX (5000 ppm). Liver RNASeq analysis identified 65 differentially expressed genes (DEGs) by high dose DX for four weeks; top canonical pathways enriched by DEGs included xenobiotic metabolism, GSH-mediated detoxification, and NRF2-mediated oxidative stress response. Liver metabolomics in these same mice identified 81 differentially abundant metabolites (DAMs). These DAMs were involved in metabolic pathways of nucleotide salvage synthesis, oxidative stress response and detoxification, DNA damage, lipid metabolism, bile acid biosynthesis, and farnesoid X receptor (FXR)/ retinoid X receptor (RXR) activation. Collectively, we demonstrated that, in the absence of liver cytotoxicity, short-term exposure (up to four weeks) to high dose DX led to a mild increase in liver DNA damage and a hepatocellular oxidative stress response.

Deficiency in antioxidant GSH sensitizes mice to oxidative DNA damage following subchronic oral exposure to high dose DX

GSH is the most abundant cellular non-protein thiol, attaining millimolar concentrations in the liver [26]. GSH serves to protect cells against toxicity arising from exposure to excessive amounts of endogenous and exogenous electrophiles [27]. GSH scavenges hydroxyl radicals and superoxide directly, and serves as a cofactor for the enzyme glutathione peroxidase (GPX) in metabolizing hydrogen peroxide and lipid peroxides [28]. Through the action GST family of enzymes, GSH conjugates to a variety of electrophilic endogenous compounds and xenobiotics resulting in their efficient elimination from the body [29]. GSH and its oxidized form, glutathione disulfide (GSSG), form an important redox buffer that maintains cellular redox homeostasis [14]. As such, a decrease in the cellular GSH/GSSG ratio is often used as a marker of oxidative stress [30].

GSH is a ubiquitous tripeptide composed of glutamate, cysteine, and glycine. The rate-limiting enzymatic reaction in GSH biosynthesis is catalyzed by glutamate-cysteine ligase (GCL), a heterodimer comprising a catalytic (GCLC) and a modifier (GCLM) subunit. GCLC possesses all the catalytic activity of GCL, whereas GCLM serves to optimize the catalytic properties of GCL holoenzyme [31]. Our laboratories and others have developed several mouse models of GSH deficiency through genetic manipulation of the Gclc or Gclm genes [32]. The global Gclm-knockout (GclmKO) mice show no overt phenotype despite having 10–40% of normal tissue GSH levels [33]. The generally good health of GclmKO mice makes them a valuable model for studying the impact of chronic GSH deficiency. In the context of liver toxicity, GclmKO mice show high susceptibility to acute liver injuries caused by the drug acetaminophen (APAP) [34] and the environmental toxicant 2,3,7,8-tetrachlorodibenzodioxin (TCDD) [35]. To date, no liver cancer studies have been performed in GclmKO mice.

As an important component of hepatic antioxidant defense systems, the role of GSH in DX liver cytotoxicity and genotoxicity was investigated using the B6 GclmKO mouse model (Table 1) [13]. Male GclmKO mice and their wild-type (WT) littermates were exposed to a high dose DX orally for one or twelve weeks. Subchronic (12 weeks) DX exposure at 5000 ppm in drinking water caused only mild and yet comparable liver cytotoxicity in WT and GclmKO mice. A multitude of molecular changes in the liver were induced by high dose DX, including a transient compensatory NRF2 antioxidant response, persistent induction of cytochrome P450 2E1 (CYP2E1) enzyme and oxidative stress, elevated 8-OHdG levels and an increase in the γH2AX/H2AX ratio (a marker of DNA damage repair). Most importantly, GclmKO mice manifested increased sensitivity to DX-induced oxidative stress and genotoxicity in the liver. Collectively, these results provide molecular evidence linking redox dysregulation to DX liver genotoxicity, implying oxidative stress may act as a candidate MOA of DX liver carcinogenicity.

CYP2E1 plays a dominant role in DX metabolism and liver cytotoxicity, but contributes partially to DX liver genotoxicity

Two important observations from our GclmKO mice study pertain to the liver CYP2E1 enzyme. First, the hepatic oxidative stress resulting from subchronic DX exposure appeared to be associated with the progressive induction of CYP2E1 protein and activity [13]. This is consistent with the notion that, through one-electron reduction of oxygen, CYP2E1 activation leads to nonspecific formation of ROS, such as superoxide anion, hydrogen peroxide, hydroxyl radicals and lipid hydroperoxides [36]. These free radicals are potential inducers of cytotoxicity and mitogenic responses [37,38]; some of them, such as lipid peroxidation by-products malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE), are highly reactive and can form DNA-adducts leading to genotoxicity [3941]. Second, there is a good correlation between hepatic CYP2E1 activity and plasma concentrations of β-hydroxyethoxyacetic acid (HEAA), the end-product of DX metabolism [13]. This observation suggests that liver CYP2E1 may be an important contributor to DX metabolism in vivo. To date, the metabolic pathways and the pathophysiological significance of DX metabolism in the liver are poorly understood. Therefore, our findings prompted the question whether CYP2E1 induction is essential to DX liver cytotoxicity and genotoxicity.

To address this research question, we replicated the high dose DX exposure study in male B6 global Cyp2e1-knockout (Cyp2e1KO) mice (manuscript in preparation) (Table 1). The Cyp2e1KO mouse model has been used in numerous toxicity studies and these mice display overall protection against liver injuries induced by drugs, xenobiotics or alcohol [36,4244]. Such protection is believed to be attributable to abolished production of CYP2E1-derived toxic free radicals [42,45,46]. In our study, exposure to 5000 ppm DX in the drinking water for twelve weeks failed to cause liver cytotoxicity in Cyp2e1KO mice; this was accompanied by blunted induction of hepatic oxidative stress and NRF2 antioxidant response. The plasma level of HEAA in DX-exposed Cyp2e1KO mice was about 15% of that detected in DX-exposed WT mice. Unexpectedly, subchronic exposure to high dose DX induced a trend towards an increase in oxidative DNA damage (8-OHdG levels) and suppression of DNA damage repair (γH2AX/H2AX ratio) in the liver of Cyp2e1KO mice. Collectively, these findings are indicative of a dominant role of CYP2E1 in DX metabolism and that CYP2E1 is responsible for DX-induced liver cytotoxicity and oxidative stress. However, our data also suggest that high dose DX-induced liver genotoxicity may, at least in part, involve CYP2E1-independent metabolic and molecular pathways.

Public health implications

DX is classified as a Group 2B human carcinogen by the International Agency for Research on Cancer (IARC) based on sufficient evidence in animal studies and inadequate human data [47,48]. Further, it has been recently classified as a substance of very high concern by the European Chemical Agency [49]. Drinking water is the dominant route of human exposure to DX affecting large numbers of individuals worldwide; such exposure is potentially extensive for people living near industrial release sites [4,6]. The monitoring data by US Environmental Protection Agency (EPA) under the Third Unregulated Contaminant Monitoring Rule (UCMR3) showed that 21% of 4864 tested US public water systems (PWS) contained detectable levels of DX (ranging from 0.07 to 33 μg/L) and 6.9% PWS exceeded the EPA health-based reference concentration of 0.35 μg/L [50]. This concerning widespread of DX contamination in the drinking water and the public health impact of such exposure have not been adequately addressed via regulatory standard setting, which is in large due to undetermined carcinogenic mechanisms of DX and the potential for low dose effects. In this context, of cautionary note, the genotoxic and cancer effective doses of DX used in ours and other animal studies (Table 1) are approximately 4,000- to 10,000-fold above the exposure levels reported in humans. However, these high dose studies are important and necessary to initially explore the adverse biological outcomes in a limited number of tested animals. It is therefore important, in future studies, to track the dose response for mechanistically relevant events identified in these studies to the lower dose range of environmental relevance. In addition, the plausible mechanistic roles of oxidative stress and CYP2E1 activation in DX genotoxicity are of high public health relevance, given that co-occurring contaminants (e.g., TCE and 1,1-dichloroethane (1,1-DCA)) and some dietary components (e.g., alcohol) may share a complimentary MOA to cause liver cancer [5153] and therefore may greatly enhance the carcinogenic potency of the mixture.

Concluding remarks

In this mini-review, we present a brief discussion on a series of hypothesis-generating and following-up mechanistic studies that investigated redox changes associated with DX cytotoxicity and genotoxicity. These studies provide experimental evidence indicating that, following high dose DX exposure, (i) in the absence of liver cytotoxicity, DNA damage and alterations of oxidative stress defense systems are consistent and early molecular changes, (ii) deficiency in antioxidant glutathione increases the susceptibility to DX-induced oxidative DNA damage and DNA damage repair, and (iii) CYP2E1 plays a dominant role in DX metabolism and DX-induced hepatic oxidative stress and liver cytotoxicity, whereas CYP2E1 contributes partially to DX liver genotoxicity. The overall conclusions from these animal model studies support a direct genotoxic effect by high dose DX and imply that redox dysregulation and oxidative stress involving CYP2E1 activation may be causally involved in DX liver genotoxicity and potentially carcinogenicity. These mechanistic data derived from high dose studies can serve as important references to refine the assessment of carcinogenic pathways that may be triggered at environmentally relevant low doses in future human and animal studies.

Acknowledgement

This work was supported in part by the National Institutes of Health grants P42ES033815 and R01AA028859.

Footnotes

Declaration of competing interest

The authors declare that they have no conflict of interest with the contents of the article.

References

  • 1.Carrera G, Vegué L, Ventura F, Hernández-Valencia A, Devesa R, Boleda MR: Dioxanes and dioxolanes in source waters: Occurrence, odor thresholds and behavior through upgraded conventional and advanced processes in a drinking water treatment plant. Water Res 2019, 156:404–413. [DOI] [PubMed] [Google Scholar]
  • 2.EPA: Toxicological Review of 1,4- Dioxane (With Inhalation Update) (CAS No. 123–91-1) in Support of Summary Information on the Integrated Risk Information System (IRIS) [EPA Report]. Edited by. Washington DC: U.S. Environmental Protection Agency; 2013. [Google Scholar]
  • 3.Adamson DT, Anderson RH, Mahendra S, Newell CJ: Evidence of 1,4-dioxane attenuation at groundwater sites contaminated with chlorinated solvents and 1,4-dioxane. Environ Sci Technol 2015, 49:6510–6518. [DOI] [PubMed] [Google Scholar]
  • 4.ATSDR: Support Document to the 2017 Substance Priority List (Candidates for Toxicological Profiles). Edited by. Atlanta, GA: Agency for Toxic Substances and Disease Registry, Division of Toxicology and Human Health Sciences; 2017. [Google Scholar]
  • 5.Sun M, Lopez-Velandia C, Knappe DR: Determination of 1,4-Dioxane in the Cape Fear River Watershed by Heated Purge-and-Trap Preconcentration and Gas Chromatography-Mass Spectrometry. Environ Sci Technol 2016, 50:2246–2254. [DOI] [PubMed] [Google Scholar]
  • 6.Godri Pollitt KJ, Kim JH, Peccia J, Elimelech M, Zhang Y, Charkoftaki G, Hodges B, Zucker I, Huang H, Deziel NC, et al. : 1,4-Dioxane as an emerging water contaminant: State of the science and evaluation of research needs. Sci Total Environ 2019, 690:853–866. [DOI] [PubMed] [Google Scholar]
  • 7.Karges U, Ott D, De Boer S, Puttmann W: 1,4-Dioxane contamination of German drinking water obtained by managed aquifer recharge systems: Distribution and main influencing factors. Sci Total Environ 2020, 711:134783. [DOI] [PubMed] [Google Scholar]
  • 8.Kociba RJ, McCollister SB, Park CN, Torkelson TR, and Gehring PJ: 1,4-Dioxane. I. Results of a 2-year ingestion study in rats. Toxicology and Applied Pharmacology 1974, 30:275–286. [Google Scholar]
  • 9.Kano H, Umeda Y, Saito M, Senoh H, Ohbayashi H, Aiso S, Yamazaki K, Nagano K, Fukushima S: Thirteen-week oral toxicity of 1,4-dioxane in rats and mice. J Toxicol Sci 2008, 33:141–153. [DOI] [PubMed] [Google Scholar]
  • 10. EPA: Final Risk Evaluation for 1,4-Dioxane. Edited by. Washington DC: U.S. Environmental Protection Agency; 2020. *This is the most recent EPA report on the risk evaluation of 1,4-dioxane. It provides an update and comprehensive review on the current knowledge of the physiochemical properties, exposure risks and health hazards of 1,4-dioxane. It highlights the urgent need of mechanistic studies to elucidate the mode of action of 1,4-dioxane carcinogenicity.
  • 11. Institute NJDWQ: Health-Based Maximum Contaminant Level Support Document, 1,4-dioxane. Edited by. http://www.state.nj.us/dep/watersupply/pdf/14-dioxane-pub-rev-health-sub.pdf; 2020. *This report from the New Jersey Drinking Water Quality Institute Health Effects Subcommittee presents the Health Effects Subcommittee’s recommendation for a Health-based MCL for 1,4-dioxane. The Subcommittee’s review focused primarily on the carcinogenic effects and recognized that the mode of action has not been established.
  • 12. Charkoftaki G, Golla JP, Santos-Neto A, Orlicky DJ, Garcia-Milian R, Chen Y, Rattray NJW, Cai Y, Wang Y, Shern CT, et al. : Identification of dose-dependent DNA damage and repair responses from subchronic exposure to 1,4-dioxane in mice using a systems analysis approach. Toxicol Sci 2021. *The authors presented multicomics investigation on early molecular changes in the liver associated with exposures to a range of 1,4-dioxane doses in drinking water. This is the first report that identified 1,4-dioxane-induced DNA damage and changes in oxidative stress response at an early stage.
  • 13. Chen Y, Wang Y, Charkoftaki G, Orlicky DJ, Davidson E, Wan F, Ginsberg G, Thompson DC, Vasiliou V: Oxidative stress and genotoxicity in 1,4-dioxane liver toxicity as evidenced in a mouse model of glutathione deficiency. Sci Total Environ 2022, 806:150703. **Utilizing a mouse model of low glutathione, the authors showed for the first time that compromised antioxdiant defense renders mice more susceptible to 1,4-dioxane-induced genotoxicity.
  • 14.Dalton TP, Chen Y, Schneider SN, Nebert DW, Shertzer HG: Genetically altered mice to evaluate glutathione homeostasis in health and disease. Free Radic Biol Med 2004, 37:1511–1526. [DOI] [PubMed] [Google Scholar]
  • 15.Wu D, Cederbaum AI: Oxidative stress and alcoholic liver disease. Semin Liver Dis 2009, 29:141–154. [DOI] [PubMed] [Google Scholar]
  • 16.Gu X, Manautou JE: Molecular mechanisms underlying chemical liver injury. Expert Rev Mol Med 2012, 14:e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yuan L, Kaplowitz N: Mechanisms of drug-induced liver injury. Clin Liver Dis 2013, 17:507–518, vii. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Pessayre D, Fromenty B, Mansouri A: Mitochondrial injury in steatohepatitis. Eur J Gastroenterol Hepatol 2004, 16:1095–1105. [DOI] [PubMed] [Google Scholar]
  • 19.Fu Y, Chung FL: Oxidative stress and hepatocarcinogenesis. Hepatoma Res 2018, 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Checa J, Aran JM: Reactive Oxygen Species: Drivers of Physiological and Pathological Processes. J Inflamm Res 2020, 13:1057–1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Kirtonia A, Sethi G, Garg M: The multifaceted role of reactive oxygen species in tumorigenesis. Cell Mol Life Sci 2020, 77:4459–4483. *This review provides a comprehensive discussion on how dyregulated redox homeostasis promotes tumorigenesis, including by activating oncogenic signaling pathways, DNA mutations, immune escape, tumor microenvironment, metastasis, angiogenesis and extension of telomere.
  • 22.Mnaa S, Shaker ES, Mahmoud HI: Inhibitory Activity of Protected Edible Plants on Oxidative Stress Induced by Oral 1,4-Dioxane. J Egypt Soc Parasitol 2016, 46:135–143. [DOI] [PubMed] [Google Scholar]
  • 23.Gi M, Fujioka M, Kakehashi A, Okuno T, Masumura K, Nohmi T, Matsumoto M, Omori M, Wanibuchi H, Fukushima S: In vivo positive mutagenicity of 1,4-dioxane and quantitative analysis of its mutagenicity and carcinogenicity in rats. Arch Toxicol 2018, 92:3207–3221. [DOI] [PubMed] [Google Scholar]
  • 24. Totsuka Y, Maesako Y, Ono H, Nagai M, Kato M, Gi M, Wanibuchi H, Fukushima S, Shiizaki K, Nakagama H: Comprehensive analysis of DNA adducts (DNA adductome analysis) in the liver of rats treated with 1,4-dioxane. Proc Jpn Acad Ser B Phys Biol Sci 2020, 96:180–187. **This is the first report on 1,4-dioxane-induced DNA adducts formation in rat livers at the adductome level. This study demonstrates that oxidative DNA modification is characteristic of 1,4-dioxane exposure.
  • 25.Kano H, Umeda Y, Kasai T, Sasaki T, Matsumoto M, Yamazaki K, Nagano K, Arito H, Fukushima S: Carcinogenicity studies of 1,4-dioxane administered in drinking-water to rats and mice for 2 years. Food Chem Toxicol 2009, 47:2776–2784. [DOI] [PubMed] [Google Scholar]
  • 26.Kretzschmar M: Regulation of hepatic glutathione metabolism and its role in hepatotoxicity. Exp Toxicol Pathol 1996, 48:439–446. [DOI] [PubMed] [Google Scholar]
  • 27.Meister A: Glutathione deficiency produced by inhibition of its synthesis, and its reversal; applications in research and therapy. Pharmacol Ther 1991, 51:155–194. [DOI] [PubMed] [Google Scholar]
  • 28.Bannai S: Exchange of cystine and glutamate across plasma membrane of human fibroblasts. J Biol Chem 1986, 261:2256–2263. [PubMed] [Google Scholar]
  • 29.Rinaldi R, Eliasson E, Swedmark S, Morgenstern R: Reactive intermediates and the dynamics of glutathione transferases. Drug Metab Dispos 2002, 30:1053–1058. [DOI] [PubMed] [Google Scholar]
  • 30.Giustarini D, Colombo G, Garavaglia ML, Astori E, Portinaro NM, Reggiani F, Badalamenti S, Aloisi AM, Santucci A, Rossi R, et al. : Assessment of glutathione/glutathione disulphide ratio and S-glutathionylated proteins in human blood, solid tissues, and cultured cells. Free Radic Biol Med 2017, 112:360–375. [DOI] [PubMed] [Google Scholar]
  • 31.Chen Y, Shertzer HG, Schneider SN, Nebert DW, Dalton TP: Glutamate cysteine ligase catalysis: dependence on ATP and modifier subunit for regulation of tissue glutathione levels. The Journal of biological chemistry 2005, 280:33766–33774. [DOI] [PubMed] [Google Scholar]
  • 32.Chen Y, Dong H, Thompson DC, Shertzer HG, Nebert DW, Vasiliou V: Glutathione defense mechanism in liver injury: insights from animal models. Food Chem Toxicol 2013, 60:38–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yang Y, Dieter MZ, Chen Y, Shertzer HG, Nebert DW, Dalton TP: Initial characterization of the glutamate-cysteine ligase modifier subunit Gclm(−/−) knockout mouse. Novel model system for a severely compromised oxidative stress response. Journal of Biological Chemistry 2002, 277:49446–49452. [DOI] [PubMed] [Google Scholar]
  • 34.McConnachie LA, Mohar I, Hudson FN, Ware CB, Ladiges WC, Fernandez C, Chatterton-Kirchmeier S, White CC, Pierce RH, Kavanagh TJ: Glutamate cysteine ligase modifier subunit deficiency and gender as determinants of acetaminophen-induced hepatotoxicity in mice. Toxicol Sci 2007, 99:628–636. [DOI] [PubMed] [Google Scholar]
  • 35.Chen Y, Krishan M, Nebert DW, Shertzer HG: Glutathione-deficient mice are susceptible to TCDD-Induced hepatocellular toxicity but resistant to steatosis. Chem Res Toxicol 2012, 25:94–100. [DOI] [PubMed] [Google Scholar]
  • 36.Harjumäki R, Pridgeon CS, Ingelman-Sundberg M: CYP2E1 in Alcoholic and Non-Alcoholic Liver Injury. Roles of ROS, Reactive Intermediates and Lipid Overload. Int J Mol Sci 2021, 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Cederbaum AI, Yang L, Wang X, Wu D: CYP2E1 Sensitizes the Liver to LPS- and TNF alpha-Induced Toxicity via Elevated Oxidative and Nitrosative Stress and Activation of ASK-1 and JNK Mitogen-Activated Kinases. Int J Hepatol 2012, 2012:582790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Schattenberg JM, Czaja MJ: Regulation of the effects of CYP2E1-induced oxidative stress by JNK signaling. Redox Biol 2014, 3:7–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Guengerich FP: Cytochrome P450 2E1 and its roles in disease. Chem Biol Interact 2020, 322:109056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Linhart K, Bartsch H, Seitz HK: The role of reactive oxygen species (ROS) and cytochrome P-450 2E1 in the generation of carcinogenic etheno-DNA adducts. Redox Biol 2014, 3:56–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Peter Guengerich F, Avadhani NG: Roles of Cytochrome P450 in Metabolism of Ethanol and Carcinogens. Adv Exp Med Biol 2018, 1032:15–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lee SS, Buters JT, Pineau T, Fernandez-Salguero P, Gonzalez FJ: Role of CYP2E1 in the hepatotoxicity of acetaminophen. J Biol Chem 1996, 271:12063–12067. [DOI] [PubMed] [Google Scholar]
  • 43.Andrade RJ, Chalasani N, Björnsson ES, Suzuki A, Kullak-Ublick GA, Watkins PB, Devarbhavi H, Merz M, Lucena MI, Kaplowitz N, et al. : Drug-induced liver injury. Nat Rev Dis Primers 2019, 5:58. [DOI] [PubMed] [Google Scholar]
  • 44.Zong H, Armoni M, Harel C, Karnieli E, Pessin JE: CytochromeP-450 CYP2E1 knockout mice are protected against high-fat diet-induced obesity and insulin resistance. American Journal of Physiology-Endocrinology and Metabolism 2012, 302:E532–E539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kang JS, Wanibuchi H, Morimura K, Gonzalez FJ, Fukushima S: Role of CYP2E1 in diethylnitrosamine-induced hepatocarcinogenesis in vivo. Cancer Res 2007, 67:11141–11146. [DOI] [PubMed] [Google Scholar]
  • 46.Gonzalez FJ: Role of cytochromes P450 in chemical toxicity and oxidative stress: studies with CYP2E1. Mutat Res 2005, 569:101–110. [DOI] [PubMed] [Google Scholar]
  • 47.Roe FJC: International Agency for Research on Cancer (IARC) 1976 Annual Report. J Clin Pathol 1978, 31:200. [Google Scholar]
  • 48.Monographs I: 1,4-Dioxane. Edited by; 1987.
  • 49.ECHA: Annex XV Report: Proposal for Identification of a Substance of Very High Concern on the Basis of the Criteria Set Out in REACH Article 57. IDENTIFICATION OF 1,4-DIOXANE AS SVHC. 2021. [Google Scholar]
  • 50.Adamson DT, Pina EA, Cartwright AE, Rauch SR, Hunter Anderson R, Mohr T, Connor JA: 1,4-Dioxane drinking water occurrence data from the third unregulated contaminant monitoring rule. Sci Total Environ 2017, 596–597:236–245. [DOI] [PubMed] [Google Scholar]
  • 51.Seitz HK: The role of cytochrome P4502E1 in the pathogenesis of alcoholic liver disease and carcinogenesis. Chem Biol Interact 2020, 316:108918. [DOI] [PubMed] [Google Scholar]
  • 52.Toyooka T, Yanagiba Y, Ibuki Y, Wang RS: Trichloroethylene exposure results in the phosphorylation of histone H2AX in a human hepatic cell line through cytochrome P450 2E1-mediated oxidative stress. J Appl Toxicol 2018, 38:1224–1232. [DOI] [PubMed] [Google Scholar]
  • 53.Lash LH, Chiu WA, Guyton KZ, Rusyn I: Trichloroethylene biotransformation and its role in mutagenicity, carcinogenicity and target organ toxicity. Mutat Res Rev Mutat Res 2014, 762:22–36. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES