Next Article in Journal
NET Formation in Systemic Lupus Erythematosus: Changes during the COVID-19 Pandemic
Previous Article in Journal
Central Roles of STAT3-Mediated Signals in Onset and Development of Cancers: Tumorigenesis and Immunosurveillance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hepatic, Extrahepatic and Extracellular Vesicle Cytochrome P450 2E1 in Alcohol and Acetaminophen-Mediated Adverse Interactions and Potential Treatment Options

1
Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
2
Plough Center for Sterile Drug Delivery Solutions, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
*
Authors to whom correspondence should be addressed.
Cells 2022, 11(17), 2620; https://doi.org/10.3390/cells11172620
Submission received: 7 July 2022 / Revised: 3 August 2022 / Accepted: 19 August 2022 / Published: 23 August 2022
(This article belongs to the Section Mitochondria)

Abstract

:
Alcohol and several therapeutic drugs, including acetaminophen, are metabolized by cytochrome P450 2E1 (CYP2E1) into toxic compounds. At low levels, these compounds are not detrimental, but higher sustained levels of these compounds can lead to life-long problems such as cytotoxicity, organ damage, and cancer. Furthermore, CYP2E1 can facilitate or enhance the effects of alcohol-drug and drug-drug interactions. In this review, we discuss the role of CYP2E1 in the metabolism of alcohol and drugs (with emphasis on acetaminophen), mediating injury/toxicities, and drug-drug/alcohol-drug interactions. Next, we discuss various compounds and various nutraceuticals that can reduce or prevent alcohol/drug-induced toxicity. Additionally, we highlight experimental outcomes of alcohol/drug-induced toxicity and potential treatment strategies. Finally, we cover the role and implications of extracellular vesicles (EVs) containing CYP2E1 in hepatic and extrahepatic cells and provide perspectives on the clinical relevance of EVs containing CYP2E1 in intracellular and intercellular communications leading to drug-drug and alcohol-drug interactions. Furthermore, we provide our perspectives on CYP2E1 as a druggable target using nutraceuticals and the use of EVs for targeted drug delivery in extrahepatic and hepatic cells, especially to treat cellular toxicity.

1. Introduction

A recent report from the Centers for Disease Control and Prevention (CDC) showed approximately 100,306 drug overdose-related deaths in one year (2020–2021), which indicates an increase of 28.5% from the previous year [1]. Many drug overdoses, such as acetaminophen (APAP) cases, are accidental and due to inappropriate use, lack of education or false perception of safety, or easy accessibility of the product [2]. Millions of people self-medicate with APAP routinely and are uninformed that many combination products also contain APAP, which increases the risk of an overdose. Other APAP-associated risks (e.g., hepatotoxicity) are further exacerbated with the concomitant use of alcohol, especially in those who have existing liver damage [3]. Liver damage is caused by chronic abuse (≥6 months), heavy drinking (>14 drinks for men and >7 drinks for women per week), binge drinking (>6 drinks in one sitting), or APAP overdose (>4 g/day) [3,4]. Both APAP and alcohol are metabolized by cytochrome P450 2E1 (CYP2E1), which plays an important role in inducing hepatotoxicity caused by both alcohol and APAP [5].
CYP2E1, one of the major metabolizing enzymes in humans, is mainly expressed in the liver [6]. To a lesser extent, CYP2E1 has been found in extrahepatic systems such as lung microsomes (10 pmol of P450 per mg protein), neuronal cells, and kidney microsomes (expression varies with age) [7,8,9]. In other studies, CYP2E1 mRNA and/or protein expression has shown to be induced two to three-fold in lymphocytes, monocytes, macrophages, microglia and astrocytes, with alcohol exposure [10,11,12,13,14]. The involvement of CYP2E1 in the metabolism of alcohol, xenobiotic drugs, such as APAP, and organic compounds (halothane, enflurane, and isoflurane) can increase the risk of carcinogenic or toxic effects with repeated, improper, or abused use [15,16].
Although alcohol is primarily metabolized by alcohol dehydrogenase (ADH) to toxic acetaldehyde, it is the activation of the microsomal ethanol oxidizing system (MEOS), a secondary alcohol-metabolic pathway, that further causes cytotoxicity, organ damage, and cancer when CYP2E1 converts alcohol into acetaldehyde when ADH is saturated [17,18,19]. Additionally, like ADH-mediated alcohol metabolism, CYP2E1-mediated alcohol metabolism produces reactive oxygen species (ROS) and acetaldehyde in the liver, which subsequently cause DNA oxidation, lipid peroxidation, and protein oxidation, ultimately leading to liver damage, cirrhosis, and liver cancer [19,20].
In the era of the COVID-19 pandemic, there have been increasing reports of alcohol consumption and self-medication [21,22]. Since APAP is a substrate of CYP2E1, drug-drug and alcohol-drug interactions are common among individuals who routinely use APAP and/or are classified as chronic, heavy, or binge drinkers. Chronic use of APAP at a high dose (>4 g/day) is known to cause liver toxicity, and even death, in children due to the acute accumulation of its toxic metabolite, N-acetyl-p-benzoquinone imine (NAPQI) [3,4,23]. Although NAPQI is cleared quickly by glutathione conjugation, its acute accumulation even at 5% is toxic and causes damage. Furthermore, concurrent use of APAP and alcohol increases the risk of hepatic- and extrahepatic cytotoxicity [24]. Additionally, various disease conditions (diabetes, fasting, and obesity) alter the mRNA expression of CYP2E1 [25,26], which could potentially increase the chances of drug-drug and drug-alcohol interactions. However, subsequent studies have shown an increase in mRNA and protein expression of CYP2E1, but not its activity, in obese and/or diabetic animal models [26,27,28]. Interestingly, these studies showed decreased expression of most CYP enzymes. These findings suggest that further studies are needed to evaluate the role of CYP2E1 in drug-drug or drug-alcohol interactions in an obese/diabetic condition. Currently, N-acetylcysteine is the only therapeutic antidote for APAP overdose with added benefits (i.e., improved hemodynamics, oxygen use, and low side effect profile). Although the relative expression of CYP2E1 in extrahepatic organs is much lower than liver, various studies have shown the effects of xenobiotics, especially alcohol, on CYP2E1-induced toxicity in extrahepatic cells [12,13,14,29]. However, being a microsomal/membrane-bound enzyme, CYP2E1 cannot circulate freely in plasma. Interestingly, the discovery of extracellular vesicles (EVs) secreted from various cell types, which are found in biofluids (i.e., plasma, urine, milk, and saliva) that carry biomolecules [30,31,32], suggests that EVs may have a role in packaging and circulating CYP2E1 in plasma. To further corroborate this idea, Kumar and other groups have recently shown substantial expression of functional CYPs, including CYP2E1, in human plasma [33,34,35,36]. However, it is yet to be determined whether packaging of CYP2E1 in EVs and its secretion in plasma has a physiological role, or it is released into plasma as a result of hepatocyte death. Additionally, the ubiquitous expression of CYP2E1 makes it an important target for APAP metabolism and drug interactions, not only in the liver but also in extrahepatic locations, especially in the brain since antidotal therapy for alcohol- and drug-drug interactions do not readily cross the blood-brain barrier (BBB).
In this review article we first discuss the roles of CYP2E1 in the metabolism of alcohol and drugs, facilitating cellular injury/toxicities, and interactions involving drugs/alcohol metabolism. Next, we review nutraceuticals that have shown potential in decreasing or averting alcohol-/drug-induced toxicity, then we highlight provisional outcomes of alcohol/drug-induced toxicity and potential treatment strategies. Finally, we cover the role and implications of EVs containing CYP2E1 in hepatic and extrahepatic cells during drug/alcohol interactions.

2. The Role of Hepatic CYP2E1 in Causal Toxicity and Potential Treatment Options

In this section, we review in vitro and in vivo animal studies that have investigated the causal effects of hepatic CYP2E1 mRNA, protein, and/or activity on alcohol- and drug-induced hepatoxicities and liver damage. The expression of CYP2E1 mRNA, protein, and/or enzyme activity are not always correlated with each other. Subsequently, we discuss various nutraceutical compounds tested to prevent or treat hepatotoxicity and liver damage. The studies with nutraceuticals are important because disulfiram, and DAS, 4-methylpyrazole are non-specific inhibitors of CYP2E1 inhibitors, and have other targets/effects.

2.1. Acetaminophen (APAP)-Induced Liver Toxicity/Injury

APAP is a commonly used analgesic found in mono and combination formulations and thus is easily overdosed, especially with improper use or duplicate therapy. This explains why it is a common cause of acute liver injury [37,38,39]. Further, the COVID-19 pandemic has amplified reports of APAP drug-induced liver injury, as it is used for managing symptoms [40]. Although the majority of APAP is quickly detoxified, a small amount of APAP metabolite, NAPQI, binds to GSH or proteins. While binding of NAPQI to GSH leads to phase II metabolic clearance, its binding to protein causes loss of protein functions leading to necrosis or apoptosis [41,42]. APAP-induced liver injury significantly increases ROS, hepatic malondialdehyde (MDA), serum aspartate transaminase, alanine transaminase, and lactate dehydrogenase levels [43,44]. Approximately 90% of APAP is excreted by sulfation and glucuronidation pathways, and 5–10% is removed in the form of NAPQI [37,45]. In obese COVID-19 patients with metabolic dysfunction-associated fatty liver disease (MAFLD), Ferron et. al., reported increased activity of CYP2E1, thus creating another complexity in the treatment regimen of these patients leading to an increased risk of drug-induced liver injury [46].
A study conducted by Licong et al. found that fisetin (FST), a flavonoid, reverses APAP-induced toxicity and reduces ROS in human fetal hepatocytes (L-02 cells) and male C57 mice [47]. The Jiang group studied APAP-induced liver injury in murine models that were treated intragastrically with Artemisia (Eucalyptol), an essential oil. Eucalyptol, an inhibitor of Kelch-like ECH-associated protein 1 (Keap1), activates nuclear factor erythroid 2-related factor 2 (Nrf2) and inhibits CYP2E1 activity leading to the suppression of NAPQI formation [38,43]. Another natural compound, shikonin (nutraceutical from the roots of Lithospermum erythrorhizon plants) in the study by Guo et al., was injected intraperitoneally to investigate its effects on APAP-induced toxicity in alpha mouse liver 12 cells and Balb/c mice [44]. Moreover, the anti-inflammatory and antioxidant properties of shikonin decreased interleukin (IL-6 and IL-1β), tumor necrosis factor-α (TNF-α), and CYP2E1 mRNA expression [44]. When APAP-challenged human hepatocellular carcinoma cells (HepG2) were treated with red betel leaf extract (from Indonesia) at 25 and 100 μg/mL, Ginting et al., showed hepatoprotective effects with elevated CYP2E1 and glutathione peroxidase gene expression [48]. Choi et al. investigated the effects of the natural alkaloid, rutaecarpine, on APAP-induced liver injury in mice [49]. Toxicity in mice was induced via intraperitoneal injection of APAP followed by oral administration of rutaecarpine; it decreased CYP2E1 protein expression and inhibited the expression of inflammatory cytokines by reducing nuclear factor kappa B cell (NF-κB) expression. Furthermore, diallyl disulfide (DADS) was used to suppress APAP-induced chronic liver injury by inhibiting CYP2E1 protein expression, anti-inflammatory, and antiapoptotic effects by inhibition of NF-κB, thus showing it to be protective [50].

2.2. Alcohol-Induced Liver Toxicity/Injury

The metabolism of alcohol by hepatic CYP2E1 leads to ROS production, consequently generating damaging toxic effects in the liver through mitochondrial dysfunction, DNA damage, and lipid peroxidation in a chronic-binge mice model [51]. Enhanced superoxide and H2O2 production are mainly due to poor coupling of CYP2E1 with NADPH-cytochrome P450 reductase (CPR) [52]. ROS influence posttranscriptional modifications through small ubiquitin-like modifications that covalently attach to epsilon-amino groups of lysine residues and modulate protein stability, activity, and localization [53]. To prevent inflammatory and oxidant responses seen in alcohol-induced liver injury, Han et al. administered astaxanthin, a xanthophyll carotenoid compound, to mice, which blocked STAT3, reduced ROS and decreased CYP2E1 protein expression [51]. HepG2 cells were pretreated with gastrodin for 4 h prior to ethanol treatment in a study by Zhang et al. The authors reported nullification of ethanol-induced toxicity and apoptosis with gastrodin treatment. Furthermore, in mice with ethanol-induced acute liver injury, gastrodin decreased the enhanced alcohol-mediated induction of both protein and enzyme activity of ADH and CYP2E1 [54]. In an intragastric ethanol-fed mouse model, ubiquitin-conjugation enzyme 9 was expressed in the liver; silencing this enzyme by siRNA lowered CYP2E1 mRNA and protein expression and prevented ROS production involved in alcoholic liver disease (ALD) [53]. In the Nagappan et al., study with chronic ethanol-fed mice, the use of cryptotanshinone from Salivamiltiorrhiza Bunge plants protected hepatocytes from ethanol-induced acute liver injury by inhibiting oxidative stress and lipogenesis [55]. Cryptotanshinone conferred its hepatoprotective effects through the activation of the AMP-activated protein kinase (AMPK), sirtuin 1 (SIRT1) as well as Nrf2 and the inhibition of CYP2E1 pathways. Recently, Avila and coworkers discussed the benefits of inhibiting overexpressed CYP2E1 protein levels for the treatment of ALD [56]. In mouse models, pharmacological inhibition of CYP2E1 with clomethiazole and genetic deletion of CYP2E1 partly prevented ALD [57,58]. In contrast, CYP2E1 overexpression enhanced the severity of ALD, suggesting the detrimental role of CYP2E1 activity in alcohol-induced liver damage [59].
The N-terminal signal of CYP2E1 protein regulates its targeting to mitochondria [60]. CYP2E1 N-terminal signal variant W23R/W30R preferentially targets CYP2E1 to mitochondria and poorly targets it to the endoplasmic reticulum [61]. Mitochondrial CYP2E1 stimulates significantly higher alcohol-induced ROS generation and induces cell injury in comparison to microsomal CYP2E1 [61]. Further, previous investigations have shown that mitochondrial CYP2E1 promotes direct damage to mitochondrial cytochrome c oxidase, which can be rescued with mitochondrial antioxidant treatment [61,62], suggesting the role of mitochondrial CYP2E1 in alcohol-induced mitochondrial ROS production and hepatotoxicity.

2.3. Drug-Drug Interactions

Multi-drug combinations are given to increase the therapeutic efficacy and success of drugs in treatment regimens of different diseases and comorbidities. However, these same drug combinations can also lead to unfavorable outcomes such as toxicity, treatment failure, or adverse drug interactions [63,64]. Drug-drug interactions (DDI) occur when a drug inhibits or induces the level of a cytochrome CYP enzyme, especially the major drug-metabolic CYP3A4, which causes an alteration in the metabolism of another drug [65,66,67]. Inhibition of CYP enzymes can cause decreased metabolism of other substrates leading to increased drug plasma concentration and eventually drug-induced toxicity [68,69]. Induction of CYP enzymes by a drug causes increased metabolism of other substrates leading to suboptimal plasma drug concentration or elevated levels of drug metabolites, which can lead to metabolite-induced toxicity and reduced drug efficacy [68].
A study conducted by Liu et al. on rats, using APAP and roxithromycin in combination, showed the induction of CYP2E1 mRNA and protein, enhanced levels of NAD(P)H, quinone oxidoreductase 1 (NQO1), TNFα, malondialdehyde, and lower mRNA and protein expression of glutathione peroxidase and superoxide dismutase (SOD) [63]. They also showed an increased mRNA and protein expression of CYP2E1 resulting in oxidative stress-mediated liver damage. In another study with 10-week-old C57BL/6J mice, the co-administration of valproic acid (VPA) and APAP showed an increase in severity of liver damage consistent with high expression of CYP2E1 mRNA, the presence of NAPQI-protein, and depletion of glutathione in the liver [70]. This study also revealed upregulated expression of steroidogenic acute regulatory protein 1 (STARD1) with endoplasmic reticulum (ER) stress-causing hepatotoxicity mediated by phosphorylation of c-Jun N-terminal kinase (JNK1/2) and SH3BP5 (SH3 domain-binding protein 5 also called SAB). Furthermore, administration of N-acetylcysteine (NAC; 2.5 mmol/kg IP) demonstrated protection against VPA- and APAP-induced liver toxicity by preventing ER stress and STARD1 induction. Cepharanthine, a biscoclaurine alkaloid, has been shown to prevent DDI via CYP2E1 in vitro human liver microsomes [71]. Anemarsaponin BII (ABII) is an active saponin and competitive inhibitor of CYP2E1 activity with half-maximal inhibitory concentration (IC50) and dissociation constant (Ki) values of 19.72 μM and 9.82 μM, respectively, suggesting a DDI between ABII and drugs metabolized by CYP2E1 [72]. A study conducted on rats and hepatic cells revealed that anti-tuberculosis drugs, such as rifampicin and isoniazid, induce liver injury by regulating the nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3) inflammasome and enhancing CYP2E1 mRNA expression [73].

2.4. Alcohol-Drug Interactions

Heavy alcohol consumption with other drugs leads to liver damage stemming from the generation of superoxide anion radicals and hydrogen peroxide after CYP2E1 metabolism. [74]. In alcohol-drug interactions, CYP2E1 is regulated by NF-κB, which activates other factors involved in liver injuries, such as inducible nitric oxide synthase, TNF-α, and IL-1β [75]. Chlormethiazole, a sedative drug, and certain phytochemicals (phenethyl isothiocyanate and sulforaphane), are used as potent inhibitors of CYP2E1 for protection and treatment of liver damage in alcohol consumers [76]. A study by Jiang et al. in mice showed that compounds such as isoquercetin (50 mg/kg), hyperoxide (50 mg/kg), 3-hydroxyphenyl acetic (50 mg/kg), 4-hydroxyphenyl acetic acid (50 mg/kg), and 3,4-hydroxyphenyl acetic acid (50 mg/kg) inhibit the protein expression of CYP2E1 and protect the liver from alcohol-induced (50%, 15 mL/kg) and APAP-induced (300 mg/kg) liver damage [77]. In chronic alcohol-drinking rodent models, simulating liver injury, the administration of CYP2E1 inhibitors chlormethiazole, phenethyl isothiocyanate, and diallyl sulfide (DAS) reduced lipid peroxidation and alcohol-induced oxidative stress [74,78].

3. Extrahepatic CYP2E1 in Drug Interactions and Potential Treatments

In this section, we discuss the role of CYP2E1 in alcohol-induced and drug-induced toxicity in extrahepatic cells, and also organ damage, as well as drug-drug and alcohol-drug interactions in vitro and in vivo animal studies. Further, this section comprises a discussion on the nutraceuticals used in various studies to prevent or treat extra-hepatotoxicity and organ damage.

3.1. Kidney

In addition to liver damage, drug overdose and/or adverse drug interactions, such as with APAP, also cause kidney injury, especially nephrotoxicity [79]. Nephrotoxicity induced by APAP is characterized by renal tubular cell apoptosis via up-regulation of CYP2E1, NF-κB, TNF-α, and cyclooxygenase-2 [50]. Akakpo et al. showed that 4-methyl pyrazole inhibits APAP-induced nephrotoxicity (300 to 600 mg/kg) in C57BL/6 mice by suppressing NAPQI [80]. Data from this study also showed that 4-methylpyrazole prevents APAP-induced nephrotoxicity by reducing oxidative stress in renal tubular epithelial cells and decreasing mitochondrial dysfunction. Another study showed that the mitochondrial complex I inhibitor, rotenone, protects the kidney against APAP-induced injury through the inhibition of mitochondrial oxidative stress and inflammation [81].
Cheng et al., injected lipopolysaccharide (LPS) intraperitoneally into mice to induce acute kidney injury (AKI). AKI mice were then treated with romidepsin (FK228) or depsipeptide; both compounds downregulated the expression of CYP2E1 by blocking the binding of hepatocyte nuclear factor-1 alpha (HNF-1α) with CYP2E1 promoter, thus reducing nephrotoxicity [82]. In addition, Un et al., delivered a single dose of 10 mg/kg (IP) cisplatin, a chemotherapeutic drug, to rats to elicit nephrotoxicity. The cisplatin-induced hepatotoxicity and nephrotoxicity revealed an elevation in creatinine, blood urea nitrogen, tissue cytokines (NF-κB and TNF-α), and oxidative stress (via CYP2E1). Aprepitant was subsequently given orally to rats in different doses (5, 10, and 20 mg/kg) and all increments conferred nephroprotective effects in addition to innate anti-emetic effects [83].

3.2. Monocytes, Astrocytes and Neuronal Cells

Brain CYP enzymes cause an immediate impact on neurotoxicity and drug response through different metabolic mechanisms in the brain [84,85,86]. Each region and cell type of the brain has a different metabolic capacity due to functional heterogeneity [87]. Overall, the CYP content in the brain is around 0.5%–2% of hepatic CYP content [86]. Even with this seemingly low amount in the brain, due to the low level of ADH, the role of CYP2E1 was shown to be greater than ADH in alcohol metabolism [14]. Yu et al., in ischemia/reperfusion injury mouse model, revealed the role of CYP2E1 in the BBB integrity as measured by 20-hydroxyecosatetraenoic acid (20-HETE) activity and BBB transfer rate [88]. In this study, CYP2E1 knock-out mice showed lower production of ROS, reduced apoptosis, neurodegeneration, lesions, and fewer neurological issues compared to control mice. These findings were consistent with the increased 20-HETE level, decline in BBB damage, increased inflammatory cytokines and increased microglia activation.
U937 monocytic cell lines and primary cells derived from peripheral blood mononuclear cells (PBMCs) are model cells to study many diseases including HIV-related pathogenesis, especially in the presence of drug abuse including alcohol. Alcohol consumption is highly prevalent in the HIV population compared to the general population [89,90]. Furthermore, Rao and Kumar showed an interaction between alcohol and anti-HIV drugs leading to increased oxidative stress and cytotoxicity [29]. Though 10-fold to 20-fold lower than hepatic CYP2E1, the Kumar group showed substantial expression of CYP2E1 in monocytic U937 cells and primary macrophages, which was further induced (two to three-fold) by alcohol exposure [12,13]. The group further showed that CYP2E1 is induced by alcohol via ROS-mediated induction of protein kinase C/c-Jun N-terminal kinase/specificity protein1 (PKC/JNK/SP1) pathway, which ultimately causes apoptotic cell death [14]. Furthermore, SVGA astrocyte cell death was prevented by using DAS, antioxidants (vitamin C and E), and CYP2E1 siRNA. In a separate study, treatment with 2-chloroethanol induced the expression of CYP2E1 in rat brain and caused cytotoxicity in rat astrocytes with an enhanced level of ROS, MDA, and apoptotic cells. Increased levels of CYP2E1, ROS, MDA, apoptotic cells, and phosphorylated ERK1/2 (extracellular signal-regulated kinase1/2) were attenuated in rat astrocytes when pretreated with either diallyl sulfide (DAS) or vitamin C prior to exposure of 2-chloroethanol [91].

3.3. Tumor Cells

Heterocyclic amines such as 2-amino-1-methyl-6-phenylimidazo [4,5] pyridine (PhIP) and alcohol-induced breast cancer, showed elevation in CYP2E1 expression and enhanced ROS generation through the estrogen-receptor-α (ER-α)-STAT-3 pathway in human breast adenocarcinoma cell lines (MCF-7; ER-α+) [92]. Further, CYP2E1-mediated oxidation of APAP drives higher proliferation and migration of breast cancer cells with high estrogen receptor expression [93,94]. Moreover, ICI 182780 (faslodex), 4’-hydroxytamoxifen, and antiestrogens were shown to prevent APA-induced proliferation in estrogen receptor +/progesterone receptor + cells [82]. Additionally, Xu et al. revealed that ganoderic acid, derived from Ganoderma mushrooms, may have the potential to prevent CYP2E1-induced cancers and other diseases in alcohol drinkers [95]. In this study the authors showed that ganoderic acid inhibits several CYP isoforms, including CYP2E1, in human liver microsomes, resulting in reduced alcohol-mediate oxidative stress and potential prevention of certain cancers.

4. CYP2E1 in Clinical Drug-Drug and Alcohol-Drug Interactions

4.1. Genotype of CYP2E1 and Related Drug Interactions

Genotype sequencing is a commonly used and helpful tool to predict potential drug interactions in clinical research and practice [96]. Popular sequencing methods include polymerase chain reaction (PCR) [97], TaqMan assays, and the SNaPshot analysis method [98]. Vuilleumier et al. used the genotype sequencing method to investigate the role of CYP2E1 in isoniazid-induced hepatoxicity in patients with dormant tuberculosis. They found a positive correlation in patients with the CYP2E1*1A/*1A genotype with the rate of developing isoniazid-induced hepatotoxicity without hepatitis [96]. Further, Costa et al. expanded the research of Vuilleumier et al. to target CYP2E1-induced adverse effects. These studies confirmed that AT-variants of CYP2E1 correlates with a higher capacity to metabolize drugs, which results in adverse effects [97]. It should be noted that post-transcriptional changes have been shown to regulate CYP2E1 protein expression in human liver. Liao et al. demonstrated an association between SNP 1561A > G present in CYP2E1 3′-UTR and reduced CYP2E1 mRNA levels in human PBMCs [99]. Various single nucleotide variants of CYP2E1 and their association with different pathological disorders have been recently reviewed [100]. Further, the CYP2E1 genotype has been shown to affect APAP and alcohol metabolism. Ueshima et al. examined APAP metabolism in subjects carrying different CYP2E1 genotypes and found that in healthy individuals, type A (c1/c1) and type C (c2/c2) correlate with longest and shortest APAP half-life in blood, respectively [101]. Further, in alcoholic noncirrhotic patients with type B, APAP half-life reduces within one week after abstinence compared to that in alcoholic patients with type A and normal subjects with type B. Additionally, a study has reported that 84% ALD patients harbor the c2 genotype, indicating an association between CYP2E1 polymorphisms and development of ALD [102].

4.2. Probe Substrates and Drug Interactions

The metabolic process of chlorzoxazone through CYP2E1 and the formation of 6-hydroxychlorzoxazone, (detectable in human plasma or urine) is used to assess the enzymatic activity of CYP2E1 [103]. Bedada et al. used piperine, resveratrol, and quercetin to test their effects on CYP2E1-mediated hydroxylation of the probe substrate chlorzoxazone in healthy volunteers. They showed that all three compounds alter the pharmacokinetics of chlorzoxazone through CYP2E1 inhibition [104]. Furthermore, the authors’ goal was to seek out more potential drugs to prevent hepatoxicity for severe alcohol drinkers. Leclercq et al. found that watercress, a cruciferous vegetable, inhibits chlorzoxazone metabolism, suggesting a potential solution for alcohol drinkers to prevent further liver damage [103,104,105]. However, four years later, the same group determined that watercress-induced inhibition of chlorzoxazone is minimal and, therefore, watercress has no significant effect on the elimination of ethanol by CYP2E1 inhibition [106]. This discrepancy is mainly because in the later study watercress was ingested 11 h prior to chlorzoxazone to study its preventive effect, while in the former studies they were given simultaneously. Thus, although watercress has the ability to inhibit CYP2E1, it does not alter overall chlorzoxazone distribution and urinal clearance. In addition, while several drugs inhibit CYP2E1 activity in vitro, some compounds and various disease states show opposite effects in humans and upregulate enzymatic activity. Recently, Sun et al. utilized aniline as a probe to compare hepatic CYP2E1 activity and found that prenatal ethanol exposure (4 g/kg/day) in male rat offspring inhibits CYP2E1 and 2D1 activities [107]. Additionally, a cocktail probe method was employed to examine the effects of Aidi (ADI) injection (a traditional Chinese medicine used for chemotherapy for various tumors) on various CYP activities in normal and liver cancer rats [108]. The authors observed that ADI can suppress activities of several CYP enzymes, including CYP2E1, and may be used in combination with other therapeutic regimens. Moreover, an active constituent of Garcinia indica and Garcinia cambogia, Garcinol, causes herb-drug interaction by inhibiting CYPs involved in drug metabolism [109]. In a previous study by Trousil et al., the authors included healthy controls and ovarian cancer patients, and gave them caffeine, chlorzoxazone, dextromethorphan, and omeprazole as in vivo probes to analyze CYP activity. CYP2E1 activity was upregulated and reduced in groups that received hydroxychlorzoxazone/chlorzoxazone and omeprazole, respectively [110]. Table 1 depicts a summary of clinical research probing CYP2E1-related interactions.
Wang et al. observed increased CYP2E1 activity in obese type-II diabetic patients with increased expression of CYP2E1 mRNA [124]. Loizou et al. and Oneta et al. both reported quick induction of CYP2E1 activity after acute alcohol consumption in healthy volunteers [112]. In another experimental group, Loizou et al. also noticed inhibition of CYP2E1 from DAS, a garlic extract, and proposed that regular consumption of this compound could reduce CYP2E1 activity [113,114]. Mitra and colleagues tested disulfiram to inhibit CYP2E1 activity to prevent the formation of hydroxylamine, a toxic metabolite of dapsone [113]. Interestingly, Frye et al. and Manyike et al. found similar results by using disulfiram on the metabolism of APAP [122]. Frye et al. also did similar studies and found similar DDI between disulfiram and vesnarinone [120,121].

5. Extracellular Vesicles (EVs) and CYP2E1

EVs are small extracellular nanovesicles (30–200 nm) that are produced by many cell types and are secreted in biofluids such as plasma, cerebrospinal fluids, urine, saliva, and milk [123]. EVs play a vital role in intracellular and intercellular communication via semi-selective packaging and transport of biomolecules such as miRNAs, mRNAs, proteins, and other biological cargo to neighboring cells or distant tissues via biofluids [30,31,32]. EVs have a unique ability to transport biomolecules and influence the pathophysiology of recipient cells. EVs in plasma are secreted from a variety of tissues, especially from the liver, upon xenobiotic exposure [125,126,127,128]. Thus, EVs are likely to play a critical role in modulating xenobiotic-induced toxicity in various extrahepatic tissue systems via EV-based transport of biomolecules [127]. In this section, we he role of EVs in cell-cell communications, especially intercellular communication. Figure 1 depicts the mechanism of CYP2E1 induction by alcohol/APAP packaging of CYP2E1 in EVs and the secretion into plasma. The EVs containing CYP2E1 subsequently circulate via plasma into distant cells, including brain cells, and cause cellular toxicity when exposed to alcohol and APAP.

5.1. Extracellular Vesicular CYP2E1

Kumar’s group reported the presence of various relevant physiologically and pharmacologically active CYP enzymes in human plasma EVs, including CYP2E1, as well as in monocytic cell-derived EVs [33,129,130,131]. In these studies, EVs were isolated first by filtering plasma through a 0.22-micron filter to remove large vesicles (>200 nm), followed by precipitation or ultracentrifugation methods. There are many methods for EV isolation, e.g., precipitation, ultracentrifugation, gel filtration, and their combination. The International Society of Extracellular Vesicles recommends a suitable method for EV isolation based on availability of the samples and need for studies, which have no or minimal interference with the outcomes. They observed that plasma-EVs from healthy subjects packaged metabolically active CYP2E1, and the level of CYP2E1 mRNA was 1000-fold higher when compared to other plasma CYP enzymes. Interestingly, the authors reported higher levels of CYP2E1 level in plasma EVs than the amounts seen in hepatic and extrahepatic (monocyte) EVs [33]. However, elevated CYP2E1 levels may not correspond with a similar increase in CYP2E1 activity. Thus, future studies are required to simultaneously investigate CYP2E1 levels and their activity for a prudent conclusion. Furthermore, Cho et al. demonstrated an increase in the total number of EVs and CYP2E1 expression in alcoholics and alcohol-fed animal models [34]. In this study, EVs were isolated using filtration through a 0.22-micron filter followed by an ultracentrifugation method. The role of CYP2E1 was demonstrated using CYP2E1-null mice and the CYP2E1 inhibitor chlormethiazole, which stopped the EV-induced toxic effects in the presence of alcohol. Moreover, CYP2E1 activity increased oxidative and endoplasmic reticulum stress, contributing to further vesicular packaging of CYP2E1. This EVs-CYP2E1 complex could potentially act as a biomarker for liver damage from long-term alcohol exposure. Plasma EVs CYP2E1 levels, at least in part, associate with alcohol-induced toxicity [35]. Future studies are needed to investigate the role of plasma EV CYP2E1 in alcohol-induced or drug-induced toxicity by blocking synthesis and packaging of CYP2E1 in EVs or selectively inhibiting plasma EV CYP2E1.

5.2. CYP2E1 in Cell-Cell Interactions

Intracellular signaling of EVs induced by the metabolic processing of alcohol leads to hepatitis, alcoholic-associated liver disease, and liver fibrosis [132]. Additionally, EV-based intercellular communication induced by alcohol could lead to damage to the immune, heart, pancreas, epithelial, and endothelial cells. Cho et al., have reported that CYP2E1-containing EVs, obtained from mice with APAP-induced liver injury, can cause hepatotoxicity in the recipient’s naïve primary hepatocytes [32]. Furthermore, treatment of the EVs obtained from these mice caused elevation of plasma ROS in mice and increased expression of proteins associated with apoptotic signaling pathways such as phospho-JNK/JNK, Bax, and cleaved caspase-3 in the recipient hepatocytes. Similarly, several studies have demonstrated the role of liver-derived EVs containing CYP2E1 upon alcohol exposure on hepatic and extrahepatic toxicities via intra-cellular and inter-cellular communications, especially between hepatocytes and immune cells [133,134].
Rahman et al. demonstrated that both human and murine plasma EVs cargos, specifically CYP2E1, play a role in exacerbating xenobiotic-induced toxicity in hepatic and extrahepatic cells [35,36,135]. EVs isolated from the plasma of healthy individuals and alcohol-exposed mice further increased alcohol-and APAP-induced toxicities in hepatic and monocytic cells. Alcohol-exposed mice also showed greater levels of plasma EVs CYP2E1 and decreased levels of oxidative defense and antioxidant enzymes. Toxicity in this study was reduced by the use of CYP2E1 siRNA and a CYP2E1 selective inhibitor, DAS [135]. These recent findings suggest the role of EVs CYP2E1 in intracellular and intercellular interactions, and their effects on xenobiotic-induced toxicity. In separate studies using human subjects, Kodidela et al. demonstrated the circulation of several cell-cell communication factors (various proteins, neuronal factors, inflammatory cytokines, and chemokines) in the plasma of alcohol drinkers [136,137]. In this study, EVs were isolated using filtration with a 0.22-micron filter followed by use of a double isolation commercially available precipitation kit.

6. Authors’ Perspectives

CYP2E1 is a druggable target to reduce the incidence of alcohol- and APAP-induced toxicity and drug-drug and alcohol-drug interactions [35]. However, there are currently no commercially available small molecule drugs that can specifically inhibit CYP2E1. Therefore, there is a need to design a novel treatment strategy using selective CYP2E1 inhibitors for alcohol and APAP overdoses. There are several reports on the use of DAS, an active ingredient of garlic extracts, as a selective CYP2E1 inhibitor [138,139]. In addition to CYP2E1 inhibition, DAS has many therapeutic properties including anticancer, antioxidant, antivirals, and anti-inflammatory activities [139]. Further, DAS has been used as a supplement to prevent and treat these diseases and conditions. Despite having many therapeutic properties, the use of DAS is restricted due to its inherent toxicity [139]. DAS is also a substrate of CYP2E1, which is metabolized into toxic metabolites DADS and diallyl trisulfide (DATS) [140]. These metabolites cause oxidative stress and cellular toxicity. Therefore, there is a critical need for replacing DAS with a safer CYP2E1 inhibitor. Rahman et al. designed several DAS analogs and conducted a thorough analysis of CYP2E1 inhibition kinetics [140,141]. Out of six selected DAS analogs, thiophene, allyl methyl sulfide, and diallyl ether appeared to be less toxic than DAS in in vitro assays. These compounds were also better than DAS in rescuing CYP2E1-mediated alcohol-induced and APAP-induced toxicities [142]. Research by Rahman et al. revealed CYP2E1 inhibitors as superior alternatives to DAS, which have the potential to be used in combination/preventive therapy for alcohol-induced and APAP-induced toxicities. Comprehensive pharmacokinetic and pharmacodynamic evaluations of these analogs in animal models are needed before realizing their potential in human applications.
The role of plasma EVs in cell-cell communication is becoming extremely important to understand the various physiological effects in disease pathologies and drug-induced organ toxicities, especially upon alcohol and APAP exposures [136,137,143]. CYP2E1 encapsulated in EVs is likely to be secreted from the liver and then transported to extrahepatic cells where metabolism of xenobiotics and endogenous drugs can occur. It is, therefore, possible that EV CYP2E1 performs a critical role in drug-drug and alcohol-drug interactions, not only in hepatic cells but also in distant extrahepatic cells. These EVs can also infiltrate into the CNS and deliver CYP2E1 to brain cells such as macrophages, microglia, astrocytes, and neurons (Figure 1). Delivery of CYP2E1 to these cells has the potential to increase the local concentration of CYP2E1, which would further increase the metabolism of alcohol and APAP causing alcohol-induced and APAP-induced brain toxicity. Concurrent use of alcohol and APAP is also likely to cause adverse interactions with other drugs that are taken simultaneously. Kumar et al. did not observe detectable levels of cytochrome P450 reductase (CPR) in plasma EVs [33]. However, brain cells have abundant CPR levels, which may be sufficient to metabolize alcohol [144,145]. Further, Rowland and colleagues have recently reported the presence of peptides and mRNA originating from various CYP and CPR in exosomes isolated from human plasma. Future studies are required to determine the quantity of these enzymes in plasma-derived EVs. Further studies are also needed to address whether increased plasma EV CYP2E1 levels result from the increased expression of hepatic or non-hepatic CYP2E1, synthesis of EVs, packaging of CYP2E1 in EVs, and/or secretion of CYP2E1-containing EVs in plasma. To address the role of EV CYP2E1 in distant cells/organs, especially in the brain, it is important to study whether EV CYP2E1 can efficiently circulate via plasma, cross the BBB, and be delivered to brain cells. Currently, it is also difficult to distinguish the effects of locally expressed CYP2E1 vs. cargo from EVs. There is also a lack tools such as an inhibitor for CYP2E1 packaging in EVs and an inhibitor to block EV secretion from the cells. Thus, to fully examine the role of EV CYP2E1 in cell-cell interactions, especially with respect to the brain, these limitations need to be overcome through future studies.
EVs, being similar to that liposomes and niosomes, are actively being investigated to develop novel drug delivery systems, especially across the BBB [135,142]. Zhuang et al. used EVs to encapsulate curcumin, a popular anti-inflammatory compound, to target brain tissue in mice, which efficiently crossed the BBB and selectively delivered curcumin to brain cells [129,130]. From the perspective of xenobiotic-induced liver damage, Nojima et al. used EVs as a drug delivery system to encapsulate sphingosine-1-phosphate, which was successfully delivered to hepatocytes and showed increased cell proliferation and tissue recovery [146]. Furthermore, Wu et al. found that EVs from hepatocytes were able to promote hepatocyte proliferation and liver recovery [147]. Since EVs have the capability of encapsulating small molecular chemicals, it is reasonable to hypothesize that EVs present in plasma, or derived from liver cells, could be used to package DAS analogs, allyl methyl sulfide, diallyl ether, and 2- prop-2-enoxyacetamide. EV-encapsulated drugs would not only reduce alcohol- and APAP-induced toxicity in hepatic and extrahepatic cells, but also suppress HIV in the CNS reservoirs. Nonetheless, before further investigating the role of EV-encapsulated CYP2E1 inhibitors, the challenges (loading, drug compatibility, toxicity profile) with EVs as a drug delivery system will need to be addressed.
Although there is a potential for EVs in drug delivery, EVs-based therapy will not reach patients without regulatory approval. Since there have been unsatisfactory outcomes of therapeutics for cancer, liver and cardiovascular diseases, EV-based therapy might be eligible for facilitated regulatory pathways (FRPs) [148]. Researchers are actively testing techniques to produce large quantities of EV-based drug products in compliance with current good manufacturing practices (cGMP) [149,150]. To mimic stem cell therapy, the delivery formulation needs to be sterile, injectable, and endotoxin-free [130]. Pachler et al. described their cGMP protocol for manufacturing EVs from human mesenchymal stromal cells in Austria, where such production is regulated by the European Medicines Agency (EMA) [151,152,153]. Harting et al. used a tangential flow filtration system to extract EVs from cells based on size difference [149,150]. This method is highly scalable and compliant with cGMP [154].

Author Contributions

S.K. wrote the first draft, edited, and proofread the manuscript. B.S. helped with manuscript writing and editing. A.K.S., S.M.T.-G., K.Z. and U.P.S. assisted with writing the first draft, and reviewed and edited the article. All authors have read and agreed to the published version of the manuscript.

Funding

The authors acknowledge the financial support from the Plough Center of Drug Delivery Solutions and the Office of Sponsored Research, University of Tennessee Health Science Center.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ahmad, F.B.; Rossen, L.; Sutton, P. Provisional Drug Overdose Death Counts. Available online: https://www.cdc.gov/nchs/nvss/vsrr/drug-overdose-data.htm (accessed on 17 November 2021).
  2. Agrawal, S.; Khazaeni, B. Acetaminophen toxicity. In Statpearls [Internet]; Statpearls publishing: Tampa, FA, USA, 2021. [Google Scholar]
  3. Prescott, L.F. Paracetamol, alcohol and the liver. Br. J. Clin. Pharmacol. 2000, 49, 291–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Gómez-Moreno, G.; Guardia, J.; Cutando, A. Interaction of paracetamol in chronic alcoholic patients. Importance for odontologists. Med. Oral Patol. Oral. Cir. Bucal 2008, 13, E235–E238. [Google Scholar] [PubMed]
  5. Wolf, K.K.; Wood Sg Fau-Allard, J.L.; Allard Jl Fau-Hunt, J.A.; Hunt Ja Fau-Gorman, N.; Gorman N Fau-Walton-Strong, B.W.; Walton-Strong Bw Fau-Szakacs, J.G.; Szakacs Jg Fau-Duan, S.X.; Duan Sx Fau-Hao, Q.; Hao Q Fau-Court, M.H.; Court Mh Fau-von Moltke, L.L.; et al. Role of CYP3A and CYP2E1 in alcohol-mediated increases in acetaminophen hepatotoxicity: Comparison of wild-type and Cyp2e1(-/-) mice. Res. Artic. 2007, 35, 1223–1231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Gonzalez, F.J.; Ueno, T.; Umeno, M.; Song, B.J.; Veech, R.L.; Gelboin, H.V. Microsomal ethanol oxidizing system: Transcriptional and posttranscriptional regulation of cytochrome P450, CYP2E1. Alcohol Alcohol. Suppl. 1991, 1, 97–101. [Google Scholar]
  7. Yoon, M.; Madden, M.C.; Barton, H.A. Extrahepatic metabolism by CYP2E1 in PBPK modeling of lipophilic volatile organic chemicals: Impacts on metabolic parameter estimation and prediction of dose metrics. J. Toxicol. Environ. Health A 2007, 70, 1527–1541. [Google Scholar] [CrossRef]
  8. Arinç, E.; Arslan, S.; Bozcaarmutlu, A.; Adali, O. Effects of diabetes on rabbit kidney and lung CYP2E1 and CYP2B4 expression and drug metabolism and potentiation of carcinogenic activity of N-nitrosodimethylamine in kidney and lung. Food Chem. Toxicol. 2007, 45, 107–118. [Google Scholar] [CrossRef]
  9. Roberts, B.J.; Shoaf, S.E.; Jeong, K.S.; Song, B.J. Induction of CYP2E1 in liver, kidney, brain and intestine during chronic ethanol administration and withdrawal: Evidence that CYP2E1 possesses a rapid phase half-life of 6 hours or less. Biochem. Biophys. Res. Commun. 1994, 205, 1064–1071. [Google Scholar] [CrossRef]
  10. Hannon-Fletcher, M.P.; Barnett, Y.A. Lymphocyte cytochrome P450 expression: Inducibility studies in male Wistar rats. Br. J. Biomed. Sci. 2008, 65, 1–6. [Google Scholar] [CrossRef]
  11. Gong, Y.; Rao, P.S.S.; Sinha, N.; Ranjit, S.; Cory, T.J.; Kumar, S. The role of cytochrome P450 2E1 on ethanol-mediated oxidative stress and HIV replication in human monocyte-derived macrophages. Biochem. Biophys. Rep. 2019, 17, 65–70. [Google Scholar] [CrossRef]
  12. Jin, M.; Arya, P.; Patel, K.; Singh, B.; Silverstein, P.S.; Bhat, H.K.; Kumar, A.; Kumar, S. Effect of alcohol on drug efflux protein and drug metabolic enzymes in U937 macrophages. Alcohol. Clin. Exp. Res. 2011, 35, 132–139. [Google Scholar] [CrossRef] [Green Version]
  13. Kumar, S.; Jin, M.; Ande, A.; Sinha, N.; Silverstein, P.S.; Kumar, A. Alcohol consumption effect on antiretroviral therapy and HIV-1 pathogenesis: Role of cytochrome P450 isozymes. Expert Opin. Drug Metab. Toxicol. 2012, 8, 1363–1375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Jin, M.; Ande, A.; Kumar, A.; Kumar, S. Regulation of cytochrome P450 2e1 expression by ethanol: Role of oxidative stress-mediated pkc/jnk/sp1 pathway. Cell Death Dis. 2013, 4, e554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Gonzalez, F.J. Role of cytochromes P450 in chemical toxicity and oxidative stress: Studies with CYP2E1. Mutat. Res. 2005, 569, 101–110. [Google Scholar] [CrossRef] [PubMed]
  16. Pentiuk, O.O.; Kachula, S.O.; Herych, O. Cytochrome P4502E1. Polymorphism, physiological function, regulation, and role in pathology. Ukr. Biokhim. Zh. 2004, 76, 16–28. [Google Scholar]
  17. Haorah, J.; Ramirez, S.H.; Floreani, N.; Gorantla, S.; Morsey, B.; Persidsky, Y. Mechanism of alcohol-induced oxidative stress and neuronal injury. Free Radic. Biol. Med. 2008, 45, 1542–1550. [Google Scholar] [CrossRef] [Green Version]
  18. Conde de la Rosa, L.; Moshage, H.; Nieto, N. Hepatocyte oxidant stress and alcoholic liver disease. Rev. Esp. Enferm Dig. 2008, 100, 156–163. [Google Scholar] [CrossRef] [Green Version]
  19. Seitz, H.K.; Mueller, S. The role of Cytochrom P4502E1 in Alcoholic Liver Disease and alcohol mediated carcinogenesis. Z. Gastroenterol. 2019, 57, 37–45. [Google Scholar] [CrossRef]
  20. Linhart, K.; Bartsch, H.; Seitz, H.K. 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. [Google Scholar] [CrossRef] [Green Version]
  21. Bonsaksen, T.; Ekeberg, Ø.; Schou-Bredal, I.; Skogstad, L.; Heir, T.; Grimholt, T.K. Use of Alcohol and Addictive Drugs During the COVID-19 Outbreak in Norway: Associations With Mental Health and Pandemic-Related Problems. Front. Public Health 2021, 9, 667729. [Google Scholar] [CrossRef]
  22. Ramalho, R.A.-O. Alcohol consumption and alcohol-related problems during the COVID-19 pandemic: A narrative review. Australas. Psychiatry 2020, 28, 524–526. [Google Scholar] [CrossRef]
  23. Abdelmegeed, M.A.; Moon, K.H.; Chen, C.; Gonzalez, F.J.; Song, B.J. Role of cytochrome P450 2E1 in protein nitration and ubiquitin-mediated degradation during acetaminophen toxicity. Biochem. Pharm. 2010, 79, 57–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Hart, S.G.; Cartun, R.W.; Wyand, D.S.; Khairallah, E.A.; Cohen, S.D. Immunohistochemical localization of acetaminophen in target tissues of the CD-1 mouse: Correspondence of covalent binding with toxicity. Fundam. Appl. Toxicol. 1995, 24, 260–274. [Google Scholar] [CrossRef] [PubMed]
  25. Enriquez, A.; Leclercq, I.; Farrell, G.C.; Robertson, G. Altered expression of hepatic CYP2E1 and CYP4A in obese, diabetic ob/ob mice, and fa/fa Zucker rats. Biochem. Biophys. Res. Commun. 1999, 255, 300–306. [Google Scholar] [CrossRef] [PubMed]
  26. Lucas, D.; Farez, C.; Bardou, L.G.; Vaisse, J.; Attali, J.R.; Valensi, P. Cytochrome P450 2E1 activity in diabetic and obese patients as assessed by chlorzoxazone hydroxylation. Fundam. Clin. Pharm. 1998, 12, 553–558. [Google Scholar] [CrossRef]
  27. Tomankova, V.; Anzenbacher, P.; Anzenbacherova, E. Effects of obesity on liver cytochromes P450 in various animal models. Biomed. Pap. 2017, 161, 144–151. [Google Scholar] [CrossRef] [Green Version]
  28. Hanley, M.J.; Abernethy, D.R.; Greenblatt, D.J. Effect of obesity on the pharmacokinetics of drugs in humans. Clin. Pharm. 2010, 49, 71–87. [Google Scholar] [CrossRef]
  29. Rao, P.S.; Kumar, S. Chronic Effects of Ethanol and/or Darunavir/Ritonavir on U937 Monocytic Cells: Regulation of Cytochrome P450 and Antioxidant Enzymes, Oxidative Stress, and Cytotoxicity. Alcohol. Clin. Exp. Res. 2016, 40, 73–82. [Google Scholar] [CrossRef] [Green Version]
  30. Xu, L.; Wu, L.F.; Deng, F.Y. Exosome: An Emerging Source of Biomarkers for Human Diseases. Curr. Mol. Med. 2019, 19, 387–394. [Google Scholar] [CrossRef]
  31. Deng, F.; Miller, J. A review on protein markers of exosome from different bio-resources and the antibodies used for characterization. J. Histotechnol. 2019, 42, 226–239. [Google Scholar] [CrossRef]
  32. Sidhom, K.; Obi, P.O.; Saleem, A. A Review of Exosomal Isolation Methods: Is Size Exclusion Chromatography the Best Option? Int. J. Mol. Sci. 2020, 21, 6466. [Google Scholar] [CrossRef]
  33. Kumar, S.; Sinha, N.; Gerth, K.A.; Rahman, M.A.; Yallapu, M.M.; Midde, N.M. Specific packaging and circulation of cytochromes P450, especially 2E1 isozyme, in human plasma exosomes and their implications in cellular communications. Biochem. Biophys. Res. Commun. 2017, 491, 675–680. [Google Scholar] [CrossRef] [PubMed]
  34. Cho, Y.E.; Mezey, E.; Hardwick, J.P.; Salem, N., Jr.; Clemens, D.L.; Song, B.J. Increased ethanol-inducible cytochrome P450-2E1 and cytochrome P450 isoforms in exosomes of alcohol-exposed rodents and patients with alcoholism through oxidative and endoplasmic reticulum stress. Hepatol. Commun. 2017, 1, 675–690. [Google Scholar] [CrossRef] [PubMed]
  35. Rahman, M.A.; Kodidela, S.; Sinha, N.; Haque, S.; Shukla, P.K.; Rao, R.; Kumar, S. Plasma exosomes exacerbate alcohol- and acetaminophen-induced toxicity via CYP2E1 pathway. Sci. Rep. 2019, 9, 6571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Rahman, M.A.; Patters, B.J.; Kodidela, S.; Kumar, S. Extracellular Vesicles: Intercellular Mediators in Alcohol-Induced Pathologies. J. Neuroimmune Pharm. 2020, 15, 409–421. [Google Scholar] [CrossRef]
  37. Assis, J.B.; Cogliati, B.; Esteves, E.; Capurro, M.L.; Fonseca, D.M.; Sá-Nunes, A. Aedes aegypti mosquito saliva ameliorates acetaminophen-induced liver injury in mice. PLoS ONE 2021, 16, e0245788. [Google Scholar] [CrossRef] [PubMed]
  38. Zhao, H.; Jiang, Z.; Chang, X.; Xue, H.; Yahefu, W.; Zhang, X. 4-Hydroxyphenylacetic Acid Prevents Acute APAP-Induced Liver Injury by Increasing Phase II and Antioxidant Enzymes in Mice. Front. Pharm. 2018, 9, 653. [Google Scholar] [CrossRef]
  39. James, L.; Sullivan, J.E.; Roberts, D. The proper use of acetaminophen. Paediatr. Child. Health 2011, 16, 544–547. [Google Scholar] [CrossRef] [Green Version]
  40. Sodeifian, F.; Seyedalhosseini, Z.S.; Kian, N.; Eftekhari, M.; Najari, S.; Mirsaeidi, M.; Farsi, Y.; Nasiri, M.J. Drug-Induced Liver Injury in COVID-19 Patients: A Systematic Review. Front. Med. 2021, 8, 731436. [Google Scholar] [CrossRef]
  41. Jiang, S.; Vozmediano, V.; Abdel-Rahman, S.M.; Schmidt, S.; James, L.P. Acetaminophen Protein Adducts in Hospitalized Children Receiving Multiple Doses of Acetaminophen. J. Clin. Pharm. 2019, 59, 1291–1299. [Google Scholar] [CrossRef]
  42. Massart, J.; Begriche, K.; Fromenty, B. Cytochrome P450 2E1 should not be neglected for acetaminophen-induced liver injury in metabolic diseases with altered insulin levels or glucose homeostasis. Clin. Res. Hepatol. Gastroenterol. 2021, 45, 101470. [Google Scholar] [CrossRef]
  43. Jiang, Z.; Guo, X.; Zhang, K.; Sekaran, G.; Cao, B.; Zhao, Q.; Zhang, S.; Kirby, G.M.; Zhang, X. The Essential Oils and Eucalyptol From Artemisia vulgaris L. Prevent Acetaminophen-Induced Liver Injury by Activating Nrf2-Keap1 and Enhancing APAP Clearance Through Non-Toxic Metabolic Pathway. Front. Pharm. 2019, 10, 782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Guo, H.; Sun, J.; Li, D.; Hu, Y.; Yu, X.; Hua, H.; Jing, X.; Chen, F.; Jia, Z.; Xu, J. Shikonin attenuates acetaminophen-induced acute liver injury via inhibition of oxidative stress and inflammation. Biomed. Pharm. 2019, 112, 108704. [Google Scholar] [CrossRef] [PubMed]
  45. Alonso, E.M.; James, L.P.; Zhang, S.; Squires, R.H. Acetaminophen Adducts Detected in Serum of Pediatric Patients With Acute Liver Failure. J. Pediatr. Gastroenterol. Nutr. 2015, 61, 102–107. [Google Scholar] [CrossRef] [Green Version]
  46. Ferron, P.J.; Gicquel, T.; Mégarbane, B.; Clément, B.; Fromenty, B. Treatments in Covid-19 patients with pre-existing metabolic dysfunction-associated fatty liver disease: A potential threat for drug-induced liver injury? Biochimie 2020, 179, 266–274. [Google Scholar] [CrossRef] [PubMed]
  47. Zhao, L.; Zhang, J.; Pan, L.; Chen, L.; Wang, Y.; Liu, X.; You, L.; Jia, Y.; Hu, C. Protective effect of 7,3’,4’-flavon-3-ol (fisetin) on acetaminophen-induced hepatotoxicity in vitro and in vivo. Phytomedicine 2019, 58, 152865. [Google Scholar] [CrossRef] [PubMed]
  48. Ginting, C.N.; Lister, I.N.E.; Girsang, E.; Widowati, W.; Yusepany, D.T.; Azizah, A.M.; Kusuma, H.S.W. Hepatotoxicity prevention in Acetaminophen-induced HepG2 cells by red betel (Piper crocatum Ruiz and Pav) extract from Indonesia via antioxidant, anti-inflammatory, and anti-necrotic. Heliyon 2021, 7, e05620. [Google Scholar] [CrossRef] [PubMed]
  49. Choi, J.H.; Jin, S.W.; Lee, G.H.; Han, E.H.; Hwang, Y.P.; Jeong, H.G. Rutaecarpine Protects against Acetaminophen-Induced Acute Liver Injury in Mice by Activating Antioxidant Enzymes. Antioxidant 2021, 10, 86. [Google Scholar] [CrossRef]
  50. Ko, J.W.; Shin, J.Y.; Kim, J.W.; Park, S.H.; Shin, N.R.; Lee, I.C.; Shin, I.S.; Moon, C.; Kim, S.H.; Kim, S.H.; et al. Protective effects of diallyl disulfide against acetaminophen-induced nephrotoxicity: A possible role of CYP2E1 and NF-κB. Food. Chem. Toxicol. 2017, 102, 156–165. [Google Scholar] [CrossRef]
  51. Han, J.H.; Ju, J.H.; Lee, Y.S.; Park, J.H.; Yeo, I.J.; Park, M.H.; Roh, Y.S.; Han, S.B.; Hong, J.T. Astaxanthin alleviated ethanol-induced liver injury by inhibition of oxidative stress and inflammatory responses via blocking of STAT3 activity. Sci. Rep. 2018, 8, 14090. [Google Scholar] [CrossRef]
  52. Ekstrom, G.; Ingelman-Sundberg, M. Rat liver microsomal NADPH-supported oxidase activity and lipid peroxidation dependent on ethanol-inducible cytochrome P-450 (P-450IIE1). Biochem. Pharm. 1989, 38, 1313–1319. [Google Scholar] [CrossRef]
  53. Tomasi, M.L.; Ramani, K.; Ryoo, M.; Cossu, C.; Floris, A.; Murray, B.J.; Iglesias-Ara, A.; Spissu, Y.; Mavila, N. SUMOylation regulates cytochrome P450 2E1 expression and activity in alcoholic liver disease. FASEB J. 2018, 32, 3278–3288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Zhang, Y.; Wang, C.; Yu, B.; Jiang, J.D.; Kong, W.J. Gastrodin Protects against Ethanol-Induced Liver Injury and Apoptosis in HepG2 Cells and Animal Models of Alcoholic Liver Disease. Biol. Pharm. Bull. 2018, 41, 670–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Nagappan, A.; Kim, J.H.; Jung, D.Y.; Jung, M.H. Cryptotanshinone from the Salvia miltiorrhiza Bunge Attenuates Ethanol-Induced Liver Injury by Activation of AMPK/SIRT1 and Nrf2 Signaling Pathways. Int. J. Mol. Sci. 2019, 21, 265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Avila, M.A.; Dufour, J.F.; Gerbes, A.L.; Zoulim, F.; Bataller, R.; Burra, P.; Cortez-Pinto, H.; Gao, B.; Gilmore, I.; Mathurin, P.; et al. Recent advances in alcohol-related liver disease (ALD): Summary of a Gut round table meeting. Gut 2020, 69, 764–780. [Google Scholar] [CrossRef]
  57. Lu, Y.; Wu, D.; Wang, X.; Ward, S.C.; Cederbaum, A.I. Chronic alcohol-induced liver injury and oxidant stress are decreased in cytochrome P4502E1 knockout mice and restored in humanized cytochrome P4502E1 knock-in mice. Free Radic. Biol. Med. 2010, 49, 1406–1416. [Google Scholar] [CrossRef] [Green Version]
  58. Gouillon, Z.; Lucas, D.; Li, J.; Hagbjork, A.L.; French, B.A.; Fu, P.; Fang, C.; Ingelman-Sundberg, M.; Donohue, T.M., Jr.; French, S.W. Inhibition of ethanol-induced liver disease in the intragastric feeding rat model by chlormethiazole. Proc. Soc. Exp. Biol. Med. 2000, 224, 302–308. [Google Scholar] [CrossRef]
  59. Morgan, K.; French, S.W.; Morgan, T.R. Production of a cytochrome P450 2E1 transgenic mouse and initial evaluation of alcoholic liver damage. Hepatology 2002, 36, 122–134. [Google Scholar] [CrossRef]
  60. Bansal, S.; Anandatheerthavarada, H.K.; Prabu, G.K.; Milne, G.L.; Martin, M.V.; Guengerich, F.P.; Avadhani, N.G. Human cytochrome P450 2E1 mutations that alter mitochondrial targeting efficiency and susceptibility to ethanol-induced toxicity in cellular models. J. Biol. Chem. 2013, 288, 12627–12644. [Google Scholar] [CrossRef] [Green Version]
  61. Bansal, S.; Liu, C.P.; Sepuri, N.B.; Anandatheerthavarada, H.K.; Selvaraj, V.; Hoek, J.; Milne, G.L.; Guengerich, F.P.; Avadhani, N.G. Mitochondria-targeted cytochrome P450 2E1 induces oxidative damage and augments alcohol-mediated oxidative stress. J. Biol. Chem. 2010, 285, 24609–24619. [Google Scholar] [CrossRef] [Green Version]
  62. Chacko, B.K.; Srivastava, A.; Johnson, M.S.; Benavides, G.A.; Chang, M.J.; Ye, Y.; Jhala, N.; Murphy, M.P.; Kalyanaraman, B.; Darley-Usmar, V.M. Mitochondria-targeted ubiquinone (MitoQ) decreases ethanol-dependent micro and macro hepatosteatosis. Hepatology 2011, 54, 153–163. [Google Scholar] [CrossRef] [Green Version]
  63. Liu, X.; Chen, C.; Zhang, X. Drug-drug interaction of acetaminophen and roxithromycin with the cocktail of cytochrome P450 and hepatotoxicity in rats. Int. J. Med. Sci. 2020, 17, 414–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Liu, L.; Sun, S.; Rui, H.; Li, X. In vitro inhibitory effects of dihydromyricetin on human liver cytochrome P450 enzymes. Pharm. Biol. 2017, 55, 1868–1874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kumar, S.; Rao, P.S.; Earla, R.; Kumar, A. Drug-drug interactions between anti-retroviral therapies and drugs of abuse in HIV systems. Expert Opin. Drug Metab. Toxicol. 2015, 11, 343–355. [Google Scholar] [CrossRef] [Green Version]
  66. Abbott, K.L.; Flannery, P.C.; Gill, K.S.; Boothe, D.M.; Dhanasekaran, M.; Mani, S.; Pondugula, S.R. Adverse pharmacokinetic interactions between illicit substances and clinical drugs. Drug Metab. Rev. 2020, 52, 44–65. [Google Scholar] [CrossRef] [PubMed]
  67. Shakeel, F.; Fang, F.; Kidwell, K.M.; Marcath, L.A.; Hertz, D.L. Comparison of eight screening tools to detect interactions between herbal supplements and oncology agents. J. Oncol. Pharm. Pr. 2020, 26, 1843–1849. [Google Scholar] [CrossRef]
  68. Shah, R.R. Mechanistic basis of adverse drug reactions: The perils of inappropriate dose schedules. Expert Opin. Drug Saf. 2005, 4, 103–128. [Google Scholar] [CrossRef]
  69. Dresser, G.K.; Spence, J.D.; Bailey, D.G. Pharmacokinetic-pharmacodynamic consequences and clinical relevance of cytochrome P450 3A4 inhibition. Clin. Pharm. 2000, 38, 41–57. [Google Scholar] [CrossRef]
  70. Torres, S.; Baulies, A.; Insausti-Urkia, N.; Alarcón-Vila, C.; Fucho, R.; Solsona-Vilarrasa, E.; Núñez, S.; Robles, D.; Ribas, V.; Wakefield, L.; et al. Endoplasmic Reticulum Stress-Induced Upregulation of STARD1 Promotes Acetaminophen-Induced Acute Liver Failure. Gastroenterology 2019, 157, 552–568. [Google Scholar] [CrossRef] [Green Version]
  71. Zhang, X.; Feng, P.; Gao, X.; Wang, B.; Gou, C.; Bian, R. In vitro inhibitory effects of cepharanthine on human liver cytochrome P450 enzymes. Pharm. Biol. 2020, 58, 247–252. [Google Scholar] [CrossRef] [Green Version]
  72. Wang, M.; Jiang, W.; Zhou, J.; Xue, X.; Yin, C. Anemarsaponin BII inhibits the activity of CYP3A4, 2D6, and 2E1 with human liver microsomes. Pharm. Biol. 2020, 58, 1064–1069. [Google Scholar] [CrossRef]
  73. Su, Q.; Kuang, W.; Hao, W.; Liang, J.; Wu, L.; Tang, C.; Wang, Y.; Liu, T. Antituberculosis Drugs (Rifampicin and Isoniazid) Induce Liver Injury by Regulating NLRP3 Inflammasomes. Mediat. Inflamm. 2021, 2021, 8086253. [Google Scholar] [CrossRef] [PubMed]
  74. Lu, Y.; Cederbaum, A.I. Alcohol Upregulation of CYP2A5: Role of Reactive Oxygen Species. React. Oxyg. Species 2016, 1, 117–130. [Google Scholar] [CrossRef]
  75. Lin, Q.; Kang, X.; Li, X.; Wang, T.; Liu, F.; Jia, J.; Jin, Z.; Xue, Y. NF-κB-mediated regulation of rat CYP2E1 by two independent signaling pathways. PLoS ONE 2019, 14, e0225531. [Google Scholar] [CrossRef] [PubMed]
  76. McCarty, M.F. Inhibition of CYP2E1 with natural agents may be a feasible strategy for minimizing the hepatotoxicity of ethanol. Med. Hypotheses 2001, 56, 8–11. [Google Scholar] [CrossRef]
  77. Jiang, Z.; Jiang, X.; Li, C.; Xue, H.; Zhang, X. Development of an IgY Antibody-Based Immunoassay for the Screening of the CYP2E1 Inhibitor/Enhancer from Herbal Medicines. Front. Pharm. 2016, 7, 502. [Google Scholar] [CrossRef] [Green Version]
  78. Leung, T.M.; Lu, Y. Alcoholic Liver Disease: From CYP2E1 to CYP2A5. Curr. Mol. Pharm. 2017, 10, 172–178. [Google Scholar] [CrossRef]
  79. Inoue, D.; Usui, R.; Nitta, K.; Koike, M. A case of acetaminophen-induced acute tubulointerstitial nephritis in adult. CEN Case Rep. 2017, 6, 185–188. [Google Scholar] [CrossRef] [Green Version]
  80. Akakpo, J.Y.; Ramachandran, A.; Orhan, H.; Curry, S.C.; Rumack, B.H.; Jaeschke, H. 4-methylpyrazole protects against acetaminophen-induced acute kidney injury. Toxicol. Appl. Pharm. 2020, 409, 115317. [Google Scholar] [CrossRef]
  81. Hua, H.; Ge, X.; Wu, M.; Zhu, C.; Chen, L.; Yang, G.; Zhang, Y.; Huang, S.; Zhang, A.; Jia, Z. Rotenone Protects Against Acetaminophen-Induced Kidney Injury by Attenuating Oxidative Stress and Inflammation. Kidney Blood Press. Res. 2018, 43, 1297–1309. [Google Scholar] [CrossRef]
  82. Cheng, S.; Wu, T.; Li, Y.; Huang, J.; Cai, T. Romidepsin (FK228) in a Mouse Model of Lipopolysaccharide-Induced Acute Kidney Injury is Associated with Down-Regulation of the CYP2E1 Gene. Med. Sci. Monit. 2020, 26, e918528. [Google Scholar] [CrossRef]
  83. Un, H.; Ugan, R.A.; Kose, D.; Bayir, Y.; Cadirci, E.; Selli, J.; Halici, Z. A novel effect of Aprepitant: Protection for cisplatin-induced nephrotoxicity and hepatotoxicity. Eur. J. Pharm. 2020, 880, 173168. [Google Scholar] [CrossRef] [PubMed]
  84. Toselli, F.; Dodd, P.R.; Gillam, E.M. Emerging roles for brain drug-metabolizing cytochrome P450 enzymes in neuropsychiatric conditions and responses to drugs. Drug Metab. Rev. 2016, 48, 379–404. [Google Scholar] [CrossRef] [PubMed]
  85. McMillan, D.M.; Tyndale, R.F. CYP-mediated drug metabolism in the brain impacts drug response. Pharmacol. Ther. 2018, 184, 189–200. [Google Scholar] [CrossRef] [PubMed]
  86. Kuban, W.; Daniel, W.A. Cytochrome P450 expression and regulation in the brain. Drug Metab. Rev. 2021, 53, 1–29. [Google Scholar] [CrossRef]
  87. García-Suástegui, W.A.; Ramos-Chávez, L.A.; Rubio-Osornio, M.; Calvillo-Velasco, M.; Atzin-Méndez, J.A.; Guevara, J.; Silva-Adaya, D. The Role of CYP2E1 in the Drug Metabolism or Bioactivation in the Brain. Oxid. Med. Cell. Longev. 2017, 2017, 4680732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Yu, J.; Zhu, H.; Kindy, M.S.; Taheri, S. Cytochrome P450 CYP2E1 Suppression Ameliorates Cerebral Ischemia Reperfusion Injury. Antioxidant 2021, 10, 52. [Google Scholar] [CrossRef]
  89. Bonacini, M. Alcohol use among patients with HIV infection. Ann. Hepatol. 2011, 10, 502–507. [Google Scholar] [CrossRef]
  90. NIAAA. Alcohol Alert No. 57 September 2002. Available online: https://pubs.niaaa.nih.gov/publications/aa57.htm (accessed on 5 April 2021).
  91. Tang, H.; Sun, Q.; Wang, T.; Liao, Y.; Wang, G.; Zhao, F.; Jin, Y. Upregulation of CYP2E1 expression causes oxidative damage induced by 2-chloroethanol in primary cultured rat astrocytes. Neurotoxicology 2019, 75, 233–244. [Google Scholar] [CrossRef]
  92. Malik, D.E.; David, R.M.; Gooderham, N.J. Ethanol potentiates the genotoxicity of the food-derived mammary carcinogen PhIP in human estrogen receptor-positive mammary cells: Mechanistic support for lifestyle factors (cooked red meat and ethanol) associated with mammary cancer. Arch. Toxicol. 2018, 92, 1639–1655. [Google Scholar] [CrossRef] [Green Version]
  93. Harnagea-Theophilus, E.; Gadd Sl Fau-Knight-Trent, A.H.; Knight-Trent Ah Fau-DeGeorge, G.L.; DeGeorge Gl Fau-Miller, M.R.; Miller, M.R. Acetaminophen-induced proliferation of breast cancer cells involves estrogen receptors. Toxicol. Appl. Pharm. 1999, 155, 273–279. [Google Scholar] [CrossRef]
  94. Leung, T.; Rajendran, R.; Singh, S.; Garva, R.; Krstic-Demonacos, M.; Demonacos, C. Cytochrome P450 2E1 (CYP2E1) regulates the response to oxidative stress and migration of breast cancer cells. Breast Cancer Res. 2013, 15, R107. [Google Scholar] [CrossRef] [PubMed]
  95. Xu, S.; Zhang, F.; Chen, D.; Su, K.; Zhang, L.; Jiang, R. In vitro inhibitory effects of ganoderic acid A on human liver cytochrome P450 enzymes. Pharm. Biol. 2020, 58, 308–313. [Google Scholar] [CrossRef] [Green Version]
  96. Vuilleumier, N.; Rossier, M.F.; Chiappe, A.; Degoumois, F.; Dayer, P.; Mermillod, B.; Nicod, L.; Desmeules, J.; Hochstrasser, D. CYP2E1 genotype and isoniazid-induced hepatotoxicity in patients treated for latent tuberculosis. Eur. J. Clin. Pharm. 2006, 62, 423–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Costa, G.N.; Magno, L.A.; Santana, C.V.; Konstantinovas, C.; Saito, S.T.; Machado, M.; Di Pietro, G.; Bastos-Rodrigues, L.; Miranda, D.M.; De Marco, L.A.; et al. Genetic interaction between NAT2, GSTM1, GSTT1, CYP2E1, and environmental factors is associated with adverse reactions to anti-tuberculosis drugs. Mol. Diagn. Ther. 2012, 16, 241–250. [Google Scholar] [CrossRef]
  98. Kang, T.S.; Woo, S.W.; Park, H.J.; Lee, Y.; Roh, J. Comparison of genetic polymorphisms of CYP2E1, ADH2, and ALDH2 genes involved in alcohol metabolism in Koreans and four other ethnic groups. J. Clin. Pharm. Ther. 2009, 34, 225–230. [Google Scholar] [CrossRef] [PubMed]
  99. Liao, L.H.; Zhang, H.; Lai, M.P.; Chen, S.L.; Wu, M.; Shen, N. Single-nucleotide polymorphisms and haplotype of CYP2E1 gene associated with systemic lupus erythematosus in Chinese population. Arthritis Res. Ther. 2011, 13, R11. [Google Scholar] [CrossRef] [Green Version]
  100. Guengerich, F.P. Cytochrome P450 2E1 and its roles in disease. Chem. Biol. Interact. 2020, 322, 109056. [Google Scholar] [CrossRef]
  101. Ueshima, Y.; Tsutsumi, M.; Takase, S.; Matsuda, Y.; Kawahara, H. Acetaminophen metabolism in patients with different cytochrome P-4502E1 genotypes. Alcohol. Clin. Exp. Res. 1996, 20, 25A–28A. [Google Scholar] [CrossRef]
  102. Tsutsumi, M.; Takada, A.; Wang, J.S. Genetic polymorphisms of cytochrome P4502E1 related to the development of alcoholic liver disease. Gastroenterology 1994, 107, 1430–1435. [Google Scholar] [CrossRef]
  103. Bedada, S.K.; Neerati, P. Resveratrol Pretreatment Affects CYP2E1 Activity of Chlorzoxazone in Healthy Human Volunteers. Phytother. Res. 2016, 30, 463–468. [Google Scholar] [CrossRef]
  104. Bedada, S.K.; Boga, P.K. Effect of piperine on CYP2E1 enzyme activity of chlorzoxazone in healthy volunteers. Xenobiotica 2017, 47, 1035–1041. [Google Scholar] [CrossRef] [PubMed]
  105. Bedada, S.K.; Neerati, P. The effect of quercetin on the pharmacokinetics of chlorzoxazone, a CYP2E1 substrate, in healthy subjects. Eur. J. Clin. Pharm. 2018, 74, 91–97. [Google Scholar] [CrossRef] [PubMed]
  106. Leclercq, I.; Desager, J.P.; Horsmans, Y. Inhibition of chlorzoxazone metabolism, a clinical probe for CYP2E1, by a single ingestion of watercress. Clin. Pharmacol. Ther. 1998, 64, 144–149. [Google Scholar] [CrossRef]
  107. Sun, X.; He, L.; Bi, H.; Huang, M.; Xiang, E.; Li, X.; Wang, H.; Guo, Y. Prenatal ethanol exposure induces dynamic changes of expression and activity of hepatic cytochrome P450 isoforms in male rat offspring. Reprod. Toxicol. 2022, 109, 101–108. [Google Scholar] [CrossRef] [PubMed]
  108. Lu, Y.; Wang, Y.; He, Y.; Pan, J.; Jin, Y.; Zheng, L.; Huang, Y.; Li, Y.; Liu, W. Aidi injection altered the activity of CYP2D4, CYP1A2, CYP2C19, CYP3A2, CYP2E1 and CYP2C11 in normal and diethylnitrosamine-induced hepatocellular carcinoma in rats. J. Ethnopharmacol. 2022, 286, 114930. [Google Scholar] [CrossRef]
  109. Bolla, L.; Srivastava, P.; Ravichandiran, V.; Nanjappan, S.K. Cytochrome P450 and P-gp Mediated Herb-Drug Interactions and Molecular Docking Studies of Garcinol. Membranes 2021, 11, 992. [Google Scholar] [CrossRef]
  110. Trousil, S.; Lee, P.; Edwards, R.J.; Maslen, L.; Lozan-Kuehne, J.P.; Ramaswami, R.; Aboagye, E.O.; Clarke, S.; Liddle, C.; Sharma, R. Altered cytochrome 2E1 and 3A P450-dependent drug metabolism in advanced ovarian cancer correlates to tumour-associated inflammation. Br. J. Pharm. 2019, 176, 3712–3722. [Google Scholar] [CrossRef]
  111. Escudier, B.; Dorval, T.; Chaput, N.; André, F.; Caby, M.P.; Novault, S.; Flament, C.; Leboulaire, C.; Borg, C.; Amigorena, S.; et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: Results of thefirst phase I clinical trial. J. Transl. Med. 2005, 3, 10. [Google Scholar] [CrossRef] [Green Version]
  112. Wang, Z.; Hall, S.D.; Maya, J.F.; Li, L.; Asghar, A.; Gorski, J.C. Diabetes mellitus increases the in vivo activity of cytochrome P450 2E1 in humans. Br. J. Clin. Pharm. 2003, 55, 77–85. [Google Scholar] [CrossRef] [Green Version]
  113. Loizou, G.D.; Cocker, J. The effects of alcohol and diallyl sulphide on CYP2E1 activity in humans: A phenotyping study using chlorzoxazone. Hum. Exp. Toxicol. 2001, 20, 321–327. [Google Scholar] [CrossRef]
  114. Oneta, C.M.; Lieber, C.S.; Li, J.; Rüttimann, S.; Schmid, B.; Lattmann, J.; Rosman, A.S.; Seitz, H.K. Dynamics of cytochrome P4502E1 activity in man: Induction by ethanol and disappearance during withdrawal phase. J. Hepatol. 2002, 36, 47–52. [Google Scholar] [CrossRef]
  115. Polonikov, A.V.; Ivanov, V.P.; Solodilova, M.A. CYP2E1 gene promoter polymorphism -1293G>C increases the risk of essential hypertension in men with alcohol abuse. Bull. Exp. Biol. Med. 2013, 155, 734–737. [Google Scholar] [CrossRef] [PubMed]
  116. Huang, Y.S.; Chern, H.D.; Su, W.J.; Wu, J.C.; Chang, S.C.; Chiang, C.H.; Chang, F.Y.; Lee, S.D. Cytochrome P450 2E1 genotype and the susceptibility to antituberculosis drug-induced hepatitis. Hepatology 2003, 37, 924–930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Eap, C.B.; Schnyder, C.; Besson, J.; Savary, L.; Buclin, T. Inhibition of CYP2E1 by chlormethiazole as measured by chlorzoxazone pharmacokinetics in patients with alcoholism and in healthy volunteers. Clin. Pharmacol. Ther. 1998, 64, 52–57. [Google Scholar] [CrossRef]
  118. Hohmann, N.; Blank, A.; Burhenne, J.; Suzuki, Y.; Mikus, G.; Haefeli, W.E. Simultaneous phenotyping of CYP2E1 and CYP3A using oral chlorzoxazone and midazolam microdoses. Br. J. Clin. Pharm. 2019, 85, 2310–2320. [Google Scholar] [CrossRef]
  119. Palmer, J.L.; Scott, R.J.; Gibson, A.; Dickins, M.; Pleasance, S. An interaction between the cytochrome P450 probe substrates chlorzoxazone (CYP2E1) and midazolam (CYP3A). Br. J. Clin. Pharm. 2001, 52, 555–561. [Google Scholar] [CrossRef] [Green Version]
  120. Frye, R.F.; Branch, R.A. Effect of chronic disulfiram administration on the activities of CYP1A2, CYP2C19, CYP2D6, CYP2E1, and N-acetyltransferase in healthy human subjects. Br. J. Clin. Pharm. 2002, 53, 155–162. [Google Scholar] [CrossRef] [Green Version]
  121. Manyike, P.T.; Kharasch, E.D.; Kalhorn, T.F.; Slattery, J.T. Contribution of CYP2E1 and CYP3A to acetaminophen reactive metabolite formation. Clin. Pharmacol. Ther. 2000, 67, 275–282. [Google Scholar] [CrossRef]
  122. Mitra, A.K.; Thummel, K.E.; Kalhorn, T.F.; Kharasch, E.D.; Unadkat, J.D.; Slattery, J.T. Metabolism of dapsone to its hydroxylamine by CYP2E1 in vitro and in vivo. Clin. Pharmacol. Ther. 1995, 58, 556–566. [Google Scholar] [CrossRef]
  123. Frye, R.F.; Tammara, B.; Cowart, T.D.; Bramer, S.L. Effect of disulfiram-mediated CYP2E1 inhibition on the disposition of vesnarinone. J. Clin. Pharm. 1999, 39, 1177–1183. [Google Scholar]
  124. Desager, J.P.; Golnez, J.L.; De Buck, C.; Horsmans, Y. Watercress has no Importance for the elimination of ethanol by CYP2E1 inhibition. Pharm. Toxicol. 2002, 91, 103–105. [Google Scholar] [CrossRef] [PubMed]
  125. Makarova, J.; Turchinovich, A.; Shkurnikov, M.; Tonevitsky, A. Extracellular miRNAs and Cell-Cell Communication: Problems and Prospects. Trends Biochem. Sci. 2021, 46, 640–651. [Google Scholar] [CrossRef] [PubMed]
  126. Caruso Bavisotto, C.; Scalia, F.; Marino Gammazza, A.; Carlisi, D.; Bucchieri, F.; Conway de Macario, E.; Macario, A.J.L.; Cappello, F.; Campanella, C. Extracellular Vesicle-Mediated Cell⁻Cell Communication in the Nervous System: Focus on Neurological Diseases. Int. J. Mol. Sci. 2019, 20, 434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Gerth, K.; Kodidela, S.; Mahon, M.; Haque, S.; Verma, N.; Kumar, S. Circulating Extracellular Vesicles Containing Xenobiotic Metabolizing CYP Enzymes and Their Potential Roles in Extrahepatic Cells Via Cell-Cell Interactions. Int. J. Mol. Sci. 2019, 20, 6178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Akbar, N.; Azzimato, V.; Choudhury, R.P.; Aouadi, M. Extracellular vesicles in metabolic disease. Diabetologia 2019, 62, 2179–2187. [Google Scholar] [CrossRef] [Green Version]
  129. Kumar, A.; Zhou, L.; Zhi, K.; Raji, B.; Pernell, S.; Tadrous, E.; Kodidela, S.; Nookala, A.; Kochat, H.; Kumar, S. Challenges in Biomaterial-Based Drug Delivery Approach for the Treatment of Neurodegenerative Diseases: Opportunities for Extracellular Vesicles. Int. J. Mol. Sci. 2020, 22, 138. [Google Scholar] [CrossRef]
  130. Zhi, K.; Kumar, A.; Raji, B.; Kochat, H.; Kumar, S. Formulation, manufacturing and regulatory strategies for extracellular vesicles-based drug products for targeted therapy of central nervous system diseases. Expert Rev. Precis. Med. Drug Dev. 2020, 5, 469–481. [Google Scholar] [CrossRef]
  131. Haque, S.; Sinha, N.; Ranjit, S.; Midde, N.M.; Kashanchi, F.; Kumar, S. Monocyte-derived exosomes upon exposure to cigarette smoke condensate alter their characteristics and show protective effect against cytotoxicity and HIV-1 replication. Sci. Rep. 2017, 7, 16120. [Google Scholar] [CrossRef] [Green Version]
  132. Cho, Y.E.; Song, B.J.; Akbar, M.; Baek, M.C. Extracellular vesicles as potential biomarkers for alcohol- and drug-induced liver injury and their therapeutic applications. Pharmacol. Ther. 2018, 187, 180–194. [Google Scholar] [CrossRef]
  133. Cho, Y.E.; Seo, W.; Kim, D.K.; Moon, P.G.; Kim, S.H.; Lee, B.H.; Song, B.J.; Baek, M.C. Exogenous exosomes from mice with acetaminophen-induced liver injury promote toxicity in the recipient hepatocytes and mice. Sci. Rep. 2018, 8, 16070. [Google Scholar] [CrossRef] [Green Version]
  134. Thietart, S.; Rautou, P.E. Extracellular vesicles as biomarkers in liver diseases: A clinician’s point of view. J. Hepatol. 2020, 73, 1507–1525. [Google Scholar] [CrossRef] [PubMed]
  135. Rahman, M.A.; Gong, Y.; Kumar, S. In vitro evaluation of structural analogs of diallyl sulfide as novel CYP2E1 inhibitors for their protective effect against xenobiotic-induced toxicity and HIV replication. Toxicol. Lett. 2018, 292, 31–38. [Google Scholar] [CrossRef] [PubMed]
  136. Kodidela, S.; Ranjit, S.; Sinha, N.; McArthur, C.; Kumar, A.; Kumar, S. Cytokine profiling of exosomes derived from the plasma of HIV-infected alcohol drinkers and cigarette smokers. PLoS ONE 2018, 13, e0201144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Kodidela, S.; Wang, Y.; Patters, B.J.; Gong, Y.; Sinha, N.; Ranjit, S.; Gerth, K.; Haque, S.; Cory, T.; McArthur, C.; et al. Proteomic Profiling of Exosomes Derived from Plasma of HIV-Infected Alcohol Drinkers and Cigarette Smokers. J. Neuroimmune Pharm. 2020, 15, 501–519. [Google Scholar] [CrossRef]
  138. Diesinger, T.; Buko, V.; Lautwein, A.; Dvorsky, R.; Belonovskaya, E.; Lukivskaya, O.; Naruta, E.; Kirko, S.; Andreev, V.; Buckert, D.; et al. Drug targeting CYP2E1 for the treatment of early-stage alcoholic steatohepatitis. PLoS ONE 2020, 15, e0235990. [Google Scholar] [CrossRef]
  139. Rao, P.S.; Midde, N.M.; Miller, D.D.; Chauhan, S.; Kumar, A.; Kumar, S. Diallyl Sulfide: Potential Use in Novel Therapeutic Interventions in Alcohol, Drugs, and Disease Mediated Cellular Toxicity by Targeting Cytochrome P450 2E1. Curr. Drug Metab. 2015, 16, 486–503. [Google Scholar] [CrossRef] [Green Version]
  140. Yang, C.S.; Chhabra, S.K.; Hong, J.Y.; Smith, T.J. Mechanisms of inhibition of chemical toxicity and carcinogenesis by diallyl sulfide (DAS) and related compounds from garlic. J. Nutr. 2001, 131, 1041s–1045s. [Google Scholar] [CrossRef] [Green Version]
  141. Brady, J.F.; Li, D.C.; Ishizaki, H.; Yang, C.S. Effect of diallyl sulfide on rat liver microsomal nitrosamine metabolism and other monooxygenase activities. Cancer Res. 1988, 48, 5937–5940. [Google Scholar]
  142. Rahman, M.A.; Midde, N.M.; Wu, X.; Li, W.; Kumar, S. Kinetic characterizations of diallyl sulfide analogs for their novel role as CYP2E1 enzyme inhibitors. Pharm. Res. Perspect. 2017, 5, e00362. [Google Scholar] [CrossRef] [Green Version]
  143. Kodidela, S.; Gerth, K.; Sinha, N.; Kumar, A.; Kumar, P.; Kumar, S. Circulatory Astrocyte and Neuronal EVs as Potential Biomarkers of Neurological Dysfunction in HIV-Infected Subjects and Alcohol/Tobacco Users. Diagnostic 2020, 10, 349. [Google Scholar] [CrossRef]
  144. Bergh, A.F.; Strobel, H.W. Reconstitution of the brain mixed function oxidase system: Purification of NADPH-cytochrome P450 reductase and partial purification of cytochrome P450 from whole rat brain. J. Neurochem. 1992, 59, 575–581. [Google Scholar] [CrossRef] [PubMed]
  145. Riedl, A.G.; Watts, P.M.; Edwards, R.J.; Boobis, A.R.; Jenner, P.; Marsden, C.D. Selective localisation of P450 enzymes and NADPH-P450 oxidoreductase in rat basal ganglia using anti-peptide antisera. Brain Res. 1996, 743, 324–328. [Google Scholar] [CrossRef]
  146. Zhuang, X.; Xiang, X.; Grizzle, W.; Sun, D.; Zhang, S.; Axtell, R.C.; Ju, S.; Mu, J.; Zhang, L.; Steinman, L.; et al. Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol. Ther. 2011, 19, 1769–1779. [Google Scholar] [CrossRef] [PubMed]
  147. Nojima, H.; Freeman, C.M.; Schuster, R.M.; Japtok, L.; Kleuser, B.; Edwards, M.J.; Gulbins, E.; Lentsch, A.B. Hepatocyte exosomes mediate liver repair and regeneration via sphingosine-1-phosphate. J. Hepatol. 2016, 64, 60–68. [Google Scholar] [CrossRef] [Green Version]
  148. Wu, Q.; Zhou, L.; Lv, D.; Zhu, X.; Tang, H. Exosome-mediated communication in the tumor microenvironment contributes to hepatocellular carcinoma development and progression. J. Hematol. Oncol. 2019, 12, 53. [Google Scholar] [CrossRef]
  149. Liberti, L.; Breckenridge, A.; Hoekman, J.; Leufkens, H.; Lumpkin, M.; McAuslane, N.; Stolk, P.; Zhi, K.; Rägo, L. Accelerating access to new medicines: Current status of facilitated regulatory pathways used by emerging regulatory authorities. J. Public Health Policy 2016, 37, 315–333. [Google Scholar] [CrossRef]
  150. Liberti, L.; Breckenridge, A.; Hoekman, J.; Leufkens, H.; Lumpkin, M.; McAuslane, N.; Stolk, P.; Zhi, K.; Rägo, L. Practical aspects of developing, implementing and using facilitated regulatory pathways in the emerging markets. In Proceedings of the Proceedings of the Poster Drug Information Association Annual Meeting, Philadelphia, PA, USA, 28 June 2016. [Google Scholar]
  151. Zhi, K.; Raji, B.; Nookala, A.R.; Khan, M.M.; Nguyen, X.H.; Sakshi, S.; Pourmotabbed, T.; Yallapu, M.M.; Kochat, H.; Tadrous, E.; et al. PLGA Nanoparticle-Based Formulations to Cross the Blood-Brain Barrier for Drug Delivery: From R&D to cGMP. Pharmaceutics 2021, 13, 500. [Google Scholar] [CrossRef]
  152. Zhi, K.; Lebo, D.B. A preformulation strategy for the selection of controlled-release components to simulate a subcutaneous implant. Boletín Latinoam. Y Del Caribe De Plantas Med. Y Aromáticas 2020, 19, 344–356. [Google Scholar] [CrossRef]
  153. Polarine, J.; Chai, R.; Kochat, H.; Pulliam, P.J.; Zhi, K.; Brooks, K. In-Situ Disinfectant Validation Case Study. Am. Pharm. Rev. Microbiol. 2021, 1–6. [Google Scholar] [CrossRef]
  154. Harting, M.T.; Srivastava, A.K.; Zhaorigetu, S.; Bair, H.; Prabhakara, K.S.; Toledano Furman, N.E.; Vykoukal, J.V.; Ruppert, K.A.; Cox, C.S., Jr.; Olson, S.D. Inflammation-Stimulated Mesenchymal Stromal Cell-Derived Extracellular Vesicles Attenuate Inflammation. Stem Cells 2018, 36, 79–90. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Schematic representation (A) and mechanisms (B) of CYP2E1 induction by alcohol/APAP. Encapsulated CYP2E1 (EVs-CYP2E1) is packaged in EVs from hepatic and extrahepatic cells, released into plasma, and circulated in periphery and CNS cells. Alcohol/APAP exposure to these cells causes oxidative stress followed by cytotoxicity, organ damage, and cancers. Attenuation of these outcomes is remedied by EVs-loaded drugs or nutraceutical agents circulating via plasma. Compounds loaded in EVs enter into hepatic/extrahepatic cells to reduce oxidative stress and protect or reverse alcohol/APAP-induced damage.
Figure 1. Schematic representation (A) and mechanisms (B) of CYP2E1 induction by alcohol/APAP. Encapsulated CYP2E1 (EVs-CYP2E1) is packaged in EVs from hepatic and extrahepatic cells, released into plasma, and circulated in periphery and CNS cells. Alcohol/APAP exposure to these cells causes oxidative stress followed by cytotoxicity, organ damage, and cancers. Attenuation of these outcomes is remedied by EVs-loaded drugs or nutraceutical agents circulating via plasma. Compounds loaded in EVs enter into hepatic/extrahepatic cells to reduce oxidative stress and protect or reverse alcohol/APAP-induced damage.
Cells 11 02620 g001
Table 1. Summary of clinical research investigating CYP2E1 and related drug-drug and alcohol-drug interactions.
Table 1. Summary of clinical research investigating CYP2E1 and related drug-drug and alcohol-drug interactions.
ReferenceCondition/Drug 1EffectDrug 2: Induced Toxicity
[111]DiabetesInductionChlorzoxazone
[112]EthanolInductionChlorzoxazone
[113]EthanolInductionChlorzoxazone
[114]EthanolInductionN/A
[115]IsoniazidInductionChlorzoxazone
[116]ChlormethiazoleInhibitionChlorzoxazone
[117]ChlorzoxazoneInhibitionMidazolam
[118]ChlorzoxazoneInhibitionMidazolam
[119]Diallyl SulphideInhibitionChlorzoxazone
[113]DisulfiramInhibitionAcetaminophen
[120]DisulfiramInhibitionAcetaminophen
[121]DisulfiramInhibitionDapson
[122]DisulfiramInhibitionVesnarinone
[123]PiperineInhibitionChlorzoxazone
[104]QuercetinInhibitionChlorzoxazone
[105]ResveratrolInhibitionChlorzoxazone
[103]WatercressInhibitionChlorzoxazone
[106]WatercressInhibitionEthanol
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kumar, S.; Singla, B.; Singh, A.K.; Thomas-Gooch, S.M.; Zhi, K.; Singh, U.P. Hepatic, Extrahepatic and Extracellular Vesicle Cytochrome P450 2E1 in Alcohol and Acetaminophen-Mediated Adverse Interactions and Potential Treatment Options. Cells 2022, 11, 2620. https://doi.org/10.3390/cells11172620

AMA Style

Kumar S, Singla B, Singh AK, Thomas-Gooch SM, Zhi K, Singh UP. Hepatic, Extrahepatic and Extracellular Vesicle Cytochrome P450 2E1 in Alcohol and Acetaminophen-Mediated Adverse Interactions and Potential Treatment Options. Cells. 2022; 11(17):2620. https://doi.org/10.3390/cells11172620

Chicago/Turabian Style

Kumar, Santosh, Bhupesh Singla, Ajay K. Singh, Stacey M. Thomas-Gooch, Kaining Zhi, and Udai P. Singh. 2022. "Hepatic, Extrahepatic and Extracellular Vesicle Cytochrome P450 2E1 in Alcohol and Acetaminophen-Mediated Adverse Interactions and Potential Treatment Options" Cells 11, no. 17: 2620. https://doi.org/10.3390/cells11172620

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop