Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Mutation Research 569 (2005) 101–110

Review

Role of cytochromes P450 in chemical toxicity and oxidative


stress: studies with CYP2E1
Frank J. Gonzalez∗
Laboratory of Metabolism, National Cancer Institute, Bethesda, MD 20892, USA

Received 15 March 2004; received in revised form 14 April 2004; accepted 18 April 2004
Available online 18 November 2004

Abstract

Cytochromes P450 are responsible for metabolism of most xenobiotics and are required for the efficient elimination of foreign
chemicals from the body. Paradoxically, these enzymes also metabolically activate biologically inert compounds to electrophilic
derivatives that can cause toxicity, cell death and sometimes cellular transformation resulting in cancer. To establish the role
of these enzymes in toxicity and carcinogenicity in vivo, gene knockout mice have been developed. To illustrate the role of
P450s in toxicity, CYP2E1-null mice were employed with the commonly used analgesic drug acetaminophen. CYP2E1 is the
rate-limiting enzyme that initiates the cascade of events leading to acetaminophen hepatotoxicity; in the absence of this P450,
toxicity will only be apparent at high concentrations. Other enzymes and nuclear receptors are also involved in activation or
inactivating chemicals. CYP2E1 is induced by alcohol and the primary P450 that carries out ethanol oxidation that can lead to the
production of activated oxygen species and oxidative stress that elevate ERK1/2 phosphorylation through EGRF/c-Raf signaling.
Paradoxically, activation of this pathway inhibits apoptotic cell death stimulated by reactive oxygen generating chemicals but
accelerates necrotic cell death produced by polyunsaturated fatty acids. CYP2E1 is thought to contribute to liver pathologies
that result from alcoholic liver disease and non-alcoholic steatohepatitis.
© 2004 Elsevier B.V. All rights reserved.

Keywords: Cytochromes P450; CYP2E1; Gene knockout mice; Acetaminophen; Metabolic activation; Carcinogens; Reactive oxygen species;
Oxidative stress ERK; MAPK

Abbreviations: ASH, alcoholic steatohepatitis; P450, cytochromes P450; ERK1/2, extracellular signal-regulated kinase 1/2; NAPQI, N-
acetyl-p-benzoquinone imine; ROS, reactive oxygen species; GST, glutathione S-transferase; PPAR␣, peroxisome proliferators-activated receptor
␣; PUFA, polyunsaturated fatty acid; EGFR, epidermal growth factor receptor
∗ Tel.: +1 301 496 9067; fax: +1 301 496 8419.

E-mail address: fjgonz@helix.nih.gov.

0027-5107/$ – see front matter © 2004 Elsevier B.V. All rights reserved.
doi:10.1016/j.mrfmmm.2004.04.021
102 F.J. Gonzalez / Mutation Research 569 (2005) 101–110

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102
1.1. Cytochromes P450. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 102
1.2. Methods for analysis of xenobiotic metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103
2. Xenobiotic metabolism null mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104
2.1. Acetaminophen hepatotoxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104
3. Role of CYP2E1 in oxidative stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
3.1. Mechanism of CYP2E1 induced ROS and toxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108

1. Introduction ing in cell toxicity and, in some cases, cell transfor-


mation leading to cancer. The O-acetylation and O-
Cytochromes P450 are a superfamily of heme- sulfation of arylamines, such as 4-aminobiphenyl and
containing monoxygenases that metabolize a large 2-acetylaminoflourene, and the heterocyclic amines
number of compounds. While a limited number of food mutagens can also result in unstable reactive
P450s are involved in the biosynthetic pathways of metabolites that can mutate DNA. Thus, xenobiotic
steroid and bile acid production, most P450s metabo- metabolism, while essential for catabolism and elim-
lize foreign compounds or xenobiotics including drugs, ination of drugs and other foreign compounds, can
toxicants and chemical carcinogens. P450s carry out produce activated toxicants and carcinogens. It has
the oxidation of carbon and nitrogen groups usually recently been recognized that under certain circum-
resulting in the addition of an alcohol. In some cases, stances, P450s can produce reactive oxygen species
an epoxide is added to an aromatic ring that is rapidly (ROS) that result in oxidative stress and cell death.
hydrolyzed to an alcohol by non-enzymatic addition of CYP2E1 is among the most active P450 producing
water, or converted to a trans dihydrodiol by epoxide ROS. This will be discussed in detail later.
hydrolases. Many metabolites generated by P450s are
subjected to conjugation with either glucuronic acid, 1.1. Cytochromes P450
sulfate, glutathione, or acetyl groups by phase 2 en-
zymes (see below). This can result in transformation P450s are complex and diverse in their regulation
of a hydrophobic molecule to a more water soluble and catalytic activities. Cloning and sequencing of
derivative that is easier to eliminate through renal and P450 cDNAs and, more recently, total genome se-
biliary excretion. quencing have revealed the existence of 102 putatively
Oxidation and conjugation of xenobiotics have his- functional genes and 88 pseudogenes in the mouse,
torically been referred to as phase 1 and phase 2 reac- and 57 putatively functional genes and 58 pseudo-
tions. The phase 1 enzymes include not only the P450s genes in humans [1]. These genes are grouped, based
but also the flavin-containing monoxygenases (FMO) on amino acid sequence similarity, into a large num-
and epoxide hydrolases (EH). The phase 2 enzymes ber of families. While several P450s are involved in
include the glutathione S-transferases (GST), UDP- the synthesis of steroid hormones and bile acids, and
glucuronosyltransferases (UGT), N-acetyltransferases the metabolism of retinoic acid and fatty acids, includ-
(NAT) and sulfotransferases (SULT). All of these en- ing prostaglandins and eicosanoids, a limited number
zymes consist of multiple forms or superfamilies sim- of P450s (15 in humans) are primarily involved in
ilar to the P450s (see below). While oxidation and xenobiotic metabolism. The xenobiotic-metabolizing
conjugation of xenobiotics usually lead to inert and P450s are found in families 1–4. Since a single P450
soluble derivatives, in some cases oxidation can lead can metabolize a large number of structurally diverse
to highly reactive electrophilic metabolites than can compounds these enzymes can collectively metabolize
covalently react with protein, RNA and DNA result- scores of chemicals found in the diet, environment and
F.J. Gonzalez / Mutation Research 569 (2005) 101–110 103

administered as drugs. While metabolism of xenobi- there is still a high degree of interindividual difference
otics such as drugs is required to efficiently eliminate in their expression as revealed by study of human liver
them from the body, as noted earlier, certain chem- specimens. For example, CYP2A6 exhibits at least a
icals are metabolically activated to reactive deriva- 100-fold difference in expression among human liver
tives that cause cell toxicity and cancer [2]. Among samples [5]. The molecular basis for these differences
these, the P450s that metabolically activate toxicants is not known but may be due to induction of the P450
and carcinogens are limited to a few forms including by endogenous hormonal and dietary compounds that
CYP1A1, CYP1A2, CYP1B1, CYP2A6, CYP2E1, and activate gene expression.
to a limited extent the CYP3A subfamily. CYP1A1 Drug interactions occur when two drugs are co-
and CYP1B1 metabolically activate polycyclic aro- administered and both are metabolized by the same
matic hydrocarbons such as benzo[a]pyrene and 7,12- P450. One drug can then inhibit the metabolism of an-
dimethylbenz[a]anthracene. CYP1A2 carries out the other drug leading to high serum levels and extended
N-oxidation of arylamines and heterocyclic amine food biological activity and sometimes toxicity. Drug inter-
mutagens and CYP2E1 metabolizes numerous low Mr actions are a major problem in human therapy espe-
toxicants and cancer suspect agents (see below). cially with CYP3A4, a P450 involved in metabolism
Xenobiotic-metabolizing P450s are found at highest of over 60% of all therapeutically used drugs [6]. In
levels in the liver, the mammalian metabolic clearing- addition, CYP3A4 activity is influenced by food and
house for both exogenous and endogenous chemicals. natural products such as grapefruit juice and is a target
However, some P450s are also expressed at lower levels for induction by ligands for the nuclear receptor PXR.
in extrahepatic tissues, most notably gut, lung, kidney
and hematopoetic tissue. Drugs administered orally or 1.2. Methods for analysis of xenobiotic
intravenously, are subject to “first pass metabolism” in metabolism
the liver that is mediated in large part by P450s. This
usually results in their biological inactivation while a Metabolism of xenobiotics can be assessed by use
drug that escapes first pass metabolism maintains its of cell extracts including membrane fractions derived
biological activity. In the absence of P450s and hepatic from liver. However, it is not certain whether data ob-
metabolic inactivation, drugs would stay in the body tained using primary cultures of hepatocytes reflect the
for long periods of time, a situation that would lead to intact liver. Indeed, in rat hepatocyte cultures it is well
an exaggerated and prolonged response to a drug. With established that the composition of P450s can change
orally administered drugs, metabolism can also be ini- [7]. A major limitation of this method to study human
tiated by P450s in the small intestine. Since metabolism xenobiotic metabolism is the general lack of availabil-
is the principle determinant of plasma concentrations ity of human liver. However, there are some limited
of a chemical, the composition and extent of expression commercial sources of human hepatocytes and liver
of P450s in gut and liver can have a major impact on slices that have been used to determine whether a chem-
the efficacy and toxicity of a drug. ical is metabolized by P450s and how it is metabolized
Drug toxicity can be affected by polymorphisms including the potential for production of an activated
in P450s and drug interactions. Humans polymor- metabolite [8]. Recombinant P450s can then be used
phisms occur with several of the xenobiotic metabo- to determine which particular P450 enzyme carries out
lizing P450s, including CYP2A6, CYP2C9, CYP2C19 metabolism of a chemical of interest.
and CYP2D6; with the exception of CYP2A6 that can A number of systems have been developed to pro-
activate certain nitrosamines, these enzymes are pri- duce catalytically active P450s. These enzymes are
marily involved in drug metabolism. No known func- particularly difficult to express because they are mem-
tional polymorphisms (that affects catalytic activity) brane bound and contain a non-covalently bound heme.
occur with P450s that metabolize toxins and carcino- In any case, bacteria, vaccinia virus, baculovirus, Cos
gens albeit there are rare mutant alleles for CYP1A2 cells, and lymphoblastoid cells have been used to ex-
[3] and CYP1B1. Mutants in CYP1B1 cause congenital press human P450s [9]. Today, recombinant human
glaucoma [4]. In spite of the lack of genetic polymor- P450s are available from commercial sources with the
phisms in toxin- and carcinogen-metabolizing P450s, most widely used produced using baculovirus. To de-
104 F.J. Gonzalez / Mutation Research 569 (2005) 101–110

termine the potential effects of xenobiotics in humans, not essential for reproduction, embryonic development
these systems are especially important since marked and physiological homeostasis. However, mice lacking
species differences in P450s are known to occur. Since cEH, NQO1 and GSTP1/2 are resistant to toxicants and
rat, mouse and human liver enzymes can metabolize a carcinogens [23].
chemical quite differently, rodent model systems can- Mice lacking receptors that regulate the expression
not be used to reliably predict human safety upon expo- of xenobiotic-metabolizing enzymes have been made
sure to xenobiotics. Genetically modified mice could including aryl hydrocarbon receptor (AHR) [24–26],
be a solution to this problem. constitutive androstane receptor (CAR) [27], and preg-
nane X receptor (PXR) [28,29]. With the exception of
AHR, these null mice also do not display any severe
2. Xenobiotic metabolism null mice phenotype suggesting that they are primarily involved
in regulating xenobiotic metabolism. However, PXR
To determine the role of P450s in xenobiotic [30–32] and CAR [33] are involved in the regulation
metabolism and toxicity, gene knockout mice can be of bile acid metabolism and bilirubin degradation, re-
used. Mice lacking several of the important toxin and spectively [34]. While, mice lacking AHR are viable,
carcinogen metabolizing P450s have been made. These they have a severe liver phenotype characterized by
include CYP1A1 [10], CYP1A2 [11], CYP1B1 [12] fibrosis [24,25,35,36]. All xenobiotic-metabolism and
and CYP2E1 [13]. Mice deficient in these enzymes or receptor-null mice exhibit differences from wild-type
even double null mice lacking two P450s [14] have no mice in their response to toxins and carcinogens [23].
deleterious phenotypes; they breed, develop normally
and exhibit no ill effects as a result of loss of P450 ex- 2.1. Acetaminophen hepatotoxicity
pression. A lack of phenotype was quite surprising in
the CYP2E1-null mice. The fact that there is no known CYP2E1 is responsible for the metabolism and
polymorphism in CYP2E1 and no report of any human metabolic activation of a large number of low Mr chem-
or human liver sample that completely lacks expres- icals, such as aliphatic, aromatic and halogenated hy-
sion of the enzyme, would argue that this P450 has an drocarbons, many of which are solvents and indus-
important role in mammalian development or physio- trial monomers, and some of which are cancer sus-
logical homeostasis. In addition, CYP2E1 metabolizes pect agents [37]. Among the CYP2E1 substrates are
ketone bodies that are liberated during starvation indi- acetaminophen (AP), azoxymethane, benzene, chlor-
cating a potentially important physiological function in zoxazone, carbon tetrachloride, chloroform, methacry-
gluconoeogenesis [15]. However, CYP2E1-null mice lonitrile, N-nitrosodimethylamine and vinyl chloride.
develop normally and show no overt deleterious phe- Thus, CYP2E1 may be an important determinant of
notype indicating that this P450 does not have a critical human susceptibility to toxicity and carcinogenicity of
role in physiological homeostasis [13]. Taken together, industrial and environmental chemicals. Indeed, cat-
analysis of P450-null mice indicate that these enzymes alytic activity of CYP2E1 has been associated with
have no role in mammalian development or physiolog- susceptibility of toxicity under industrial exposure
ical homeostasis but these observation do not exclude to benzene [38]. However, while there are a num-
potential subtle functions of P450s in the metabolism ber of SNPs in and around the gene, a polymor-
of endogenous compounds [16]. However, P450-null phism in CYP2E1 that leads to loss of expression
mice are resistant to toxicants and carcinogens [17]. has not been found (http://www.imm.ki.se/CYPalleles/
Other xenobiotic-metabolizing enzymes have been cyp2e1.htm). Among the most well-studied CYP2E1
subjected to targeted gene disruption including the sol- substrates is acetaminophen (AP), also called parac-
uble epoxide hydrolase (sEH) [18], microsomal epox- etamol.
ide hydrolase (mEH) [19], NAD(P)H:quinone oxidore- AP is a widely used over-the-counter analgesic
ductase (NQO) 1 [20] and 2 [21], and glutathione S- that can cause hepatotoxicity, sometimes resulting in
transferase (GST) P1 and P2 [22]. These mice also death. This is usually caused by deliberate and acci-
did not display any phenotype again indicating that dental overdosing the drug [39]. Over 30 years ago,
other non-P450 xenobiotic-metabolizing enzymes are the hepatotoxicity of AP was found to be due to its
F.J. Gonzalez / Mutation Research 569 (2005) 101–110 105

Fig. 2. The role of P450s in acetaminophen toxicity determined using


P450-null mice. AP was administered by intraperitoneal injection
to wild-type mice (䊉) and mice lacking CYP1A2 (), CYP2E1
() and both CYP1A2 and CYP2E1 (). The percent survival was
determined as described [14].

The CYP2E1-null mouse model has been used to


Fig. 1. Metabolism of acetaminophen by CYP2E1 and GST.
study a number of chemicals that are of concern for hu-
CYP2E1 produces the active metabolite from acetaminophen. The
active metabolite is a substrate for GST. Under conditions of high lev- man risk assessment including susceptibility to AP hep-
els of CYP2E1 and/or low levels of glutathione, the active metabolite atoxicity. Testing CYP2E1-null mice for sensitivity to
can bind to cellular macromolecules resulting in cell death. AP hepatotoxicity provided evidence that CYP2E1 was
responsible for initiating the cascade of toxic metabo-
metabolism and covalent binding to cellular macro- lites from AP. Mice lacking CYP2E1 were highly resis-
molecules [40,41]. P450s and GSTs are largely respon- tant to liver toxicity as compared with wild-type mice
sible for metabolism of the drug (Fig. 1). AP is con- [13,14]. Mice lacking both CYP2E1 and CYP1A2 were
verted by P450s to a highly reactive quinone metabo- almost totally resistant to liver toxicity at the highest
lite, N-acetyl-p-benzoquinone imine (NAPQI) that can doses of AP tested (Fig. 2). The toxicity of AP is also
produce reactive oxygen species and covalently bind influenced by the genetic status of GSTP1/2 [43] and
to cellular nucleophiles such as DNA, RNA and pro- the nuclear receptor CAR [44], an inducer of GSTPi.
teins, resulting in cell death. Typically, this metabo-
lite is inactivated by glutathione conjugation to the
electrophilic P450-derived quinone metabolite. Toxi- 3. Role of CYP2E1 in oxidative stress
city of AP is enhanced by depletion of hepatic levels of
glutathione. Inhibition of glutathione synthesis exac- Oxidative stress is the result of an increase in in-
erbates toxicity and administration of the glutathione tracellular prooxidant species such as H2 O2 , hydroxy
precursor N-acetylcysteine can be used for therapeutic radicals (• OH) and superoxide anion radical (O2 − ).
intervention by elevating the level of glutathione. Sul- High intracellular levels of these molecules, collec-
fotransferases can also neutralize the activated metabo- tively called reactive oxygen species (ROS), can lead
lites formed by P450s. CYP2E1 is the principle P450 to damaged mitochondria, DNA modification, lipid
responsible for metabolism of AP [13]. This offered peroxidation, elevated cytokine production and even
a possible explanation why ethanol, an inducer of cell death. In mammals, ROS can result in a num-
CYP2E1, increased the LD50 for the drug in rats ber of pathological conditions including Alzheimer’s
[42] and why AP-induced hepatotoxicity is frequently disease, atherosclerosis, cancer, diabetes, chronic ob-
associated with alcohol consumption in humans structive pulmonary disease, rheumatoid arthritis and
[42]. various neurological disorders [45]. Oxidative stress
106 F.J. Gonzalez / Mutation Research 569 (2005) 101–110

is also thought to be the principle cause of aging bodies that are inducers of CYP2E1. Thus, increase in
[46]. hepatic fatty acids leads to an increase in ketone bod-
Chronic consumption of alcohol can lead to hepato- ies as a result of fatty acid ␤-oxidation in peroxisomes
cyte cell death and cirrhosis. Indeed, alcohol-induced and mitochondria. The ketone bodies in turn induce
liver damage is thought to be due to oxidative stress CYP2E1 expression [56]. The elevated CYP2E1 can
that results from alcohol consumption [47]. Experi- then produce ROS that produces lipid peroxides. In hu-
mentally, ethanol-induced oxidative stress can be in- mans CYP2E1 protein is spontaneously induced in the
hibited by the administration of antioxidants including livers of NASH patients [57]. To measure CYP2E1 ex-
precursors to glutathione, as described above for AP. pression in vivo, chlorzoxazone pharmacokinetics has
Metabolism of ethanol by alcohol dehydrogenase can been used an in vivo metabolic probe. Obese humans
increase oxidative stress. CYP2E1, which can metab- had elevated CYP2E1 activity and protein indicating
olize ethanol to acetaldehyde and 1-hydroxyethyl rad- that CYP2E1 is related to or caused by hepatic pathol-
ical, produces ROS. On this basis, CYP2E1 is thought ogy resulting from morbid obesity; the level of P450
to have a role in alcoholic steatohepatitis (ASH) that was correlated with the degree of steatosis [58]. As in
leads to cirrhosis. As noted above, CYP2E1 also me- rodents a similar high level of CYP2E1 activity was
tabolizes a large number of other low Mr chemicals to found in non-diabetic patients with NASH, indicating
reactive intermediates that themselves can bind to cel- that CYP2E1 is elevated in humans with this condi-
lular macromolecules such as DNA, RNA and protein tion [59]. Chlorzoxazone metabolism in vivo was also
causing cell damage, hepatitis and cirrhosis. During used to determine that Type II diabetics have elevated
the course of the P450 catalytic cycle, P450s use H+ CYP2E1 expression [60]. No significant difference was
from NADPH to reduce O2 leading to the production found in Type I diabetics. While no significant dif-
of H2 O2 and superoxide anion radical. The process of ference was found between diabetics and controls in
uncoupling of the catalytic cycle can lead to escape of CYP2E1 activity in vivo but an increase with obesity
O2 − . Thus, CYP2E1 metabolism of a number of its was found [61].
substrates is known to lead to increased ROS. Interest- It should be emphasized that the situation may dif-
ingly, even in the absence of substrate, expression of fer between humans and rodents. Mice can be induced
CYP2E1 can generate ROS [48,49]. This phenomenon, to develop NASH by feeding a methionine and choline
known as futile cycling, is due to P450-derived NADPH deficient (MCD) diet. However, CYP2E1-null mice as
oxidase activity that is independent of exogenous xeno- well as wild-type mice also develop the steatotic liver
biotic substrates [50]. phenotype and still produce significant lipid peroxi-
Similar to ASH, induction of non-alcoholic steato- dase activity due to expression of other P450s such as
hepatitis (NASH) is thought to be due to elevated ex- CYP4A10 and CYP4A14 [52]. The latter P450s are
pression of CYP2E1 and the resultant oxidative stress induced through activation of the nuclear receptor per-
[51–53]. ROS from CYP2E1 can produce lipid perox- oxisome proliferators-activated receptor ␣ (PPAR␣).
ides from polyunsaturated fatty acids (PUFA) and these Indeed, peroxisome proliferators-activated receptor ␣
can react with other cellular macromolecules causing (PPAR␣)-null mice are protected against diet induced
cell toxicity and death. It is likely that the fatty livers NASH probably due to the enhanced metabolism of
directly cause induction of CYP2E1 that contributes to fatty acids and the removal of PUFAs from the liver
lipid peroxide production. In a rat model of NASH, pro- [62]. It should be recognized that a major species dif-
duced by feeding a high fat diet, CYP2E1 is markedly ference in PPAR␣ inducibility exist between rodent
induced [54]. In a genetic model of methionine adeno- models and humans. Mice and rats, CYP4A P450s are
syltransferase 1A (MAT1A) deficiency, NASH spon- readily induced whereas humans are refractive to in-
taneously develops and is associated with elevated duction [63]. Thus, the results in animal models must
CYP2E1 expression and increased lipid peroxidation be interpreted with caution [64].
that is decreased by addition of the CYP2E1 inhibitor CYP2E1 also appears to be involved in kidney tox-
diallyl sulfide [55]. The increase in CYP2E1 in these icity due to oxidative stress. Treatment of mice with
models is likely due to ␤-oxidation of fatty acids that the anticancer drug cisplatin produces nephrotoxicity
accumulate in liver resulting in high levels of ketone indicated by elevated serum creatinine and urea, de-
F.J. Gonzalez / Mutation Research 569 (2005) 101–110 107

creased creatinine clearance, and increased catalytic sitizes cells to exogenous agents such as menadione
iron [65]. This toxicity is markedly attenuated in mice and PUFAs as a result of activation of the EGFR/c-Raf
lacking CYP2E1. In vitro, kidney slices from CYP2E1- ERK1/2 signal transduction pathway. This can either
null mice treated with the drug produced considerably lead to protection against menadione-induced apopto-
less H2 O2 than those from wild-type mice. Cisplatin- sis or lead to an increase in PUFA-stimulated cell necro-
induced kidney cell death was due to apoptosis [65]. sis. It should be noted that these studies are largely
These studies show the role of CYP2E1 in ROS- derived from experiments using cultured cells where
induced disease is not restricted to kidney. CYP2E1 is over-expressed. It remains to be determined
whether this enzyme plays a role in non-xenobiotic me-
3.1. Mechanism of CYP2E1 induced ROS and diated hepatotoxicity and cell death leading to cirrhosis
toxicity in intact liver. The role of CYP2E1 in ERK1/2 signaling
in intact liver could be determined in the CYP2E1-null
A role for CYP2E1 in production of ROS and its mouse model [13].
consequences in intact cells has been investigated in The role of CYP2E1 in hepatoxicity can be illus-
cell lines stably expressing recombinant enzymes [66]. trated in the diagram shown in Fig. 3. CYP2E1 can me-
This system has been of great value in determining the tabolize substrates (R), and depending on the nature of
specific role of CYP2E1 in producing oxidative stress the compound, can produce electrophilic metabolites
in intact cells and the molecular consequence of the that can cause cell toxicity by reacting with cellular
oxidative stress [49]. Using a similar system, the mech- macromolecules. In the presence or even the absence
anism by which ROS produced by CYP2E1 affects of substrate, CYP2E1, with O2 and NADPH, can pro-
cell toxicity has recently been investigated [67]. Over-
expression of CYP2E1 in a hepatocyte-derived cell
line, results in an increase in phosphorylated extracel-
lular signal-regulated kinase 1/2 (ERK1/2), a mitogen-
activated protein kinase (MAPK) that is activated by
ROS. Treatment with menadione, a quinone that gen-
erates reactive oxygen, resulted in a markedly en-
hanced phosphorylation of ERK1/2 in the CYP2E1-
expressing cells as compared to control cells. Surpris-
ingly this is associated with protection of the cells from
menadione-induced apoptotic cell death. ERK1/2 in-
hibitors were used to demonstrate that the resistance
of CYP2E1-expressing cells to menadione-stimulated
cell death was due to the epidermal growth factor re-
ceptor (EGFR)c-Raf signal transduction pathway [68].
In contrast to the results with menadione, treatment of
CYP2E1-expressing cells with a PUFA such as arachi-
donic acid results in enhanced cell death that is inhib-
ited by antioxidants. PUFA treatment also caused an in-
crease in phosphorylation of ERK/1/2 in the CYP2E1-
expressing cells, but in contrast to menadione treat- Fig. 3. Schematic of the influence of CYP2E1 in generation of ROS
ment, inhibition of the ERK1/2 enhances cell death that and cell toxicity. Ethanol can induce expression of CYP2E1 result-
is due largely to necrosis, not apoptosis. PUFA-induced ing in elevated ROS. Fatty acids, most importantly PUFAs can be
cell death is thought to be the result of peroxidation of subjected to peroxidation leading to reactive species that can dam-
the unsaturated bonds in the fatty acids and their sub- age cellular macromolecules. Fatty acids can also be metabolized to
ketone bodies that can induce expression of CYP2E1 and be me-
sequent reaction with cellular macromolecules. These tabolized by CYP2E1 leading to even more ROS production. The
studies with CYP2E1-expressing cells suggests a low ROS increases phosphorylated ERK1/2 leading to gene activation
level of ROS produced by CYP2E1 expression sen- and modulation of apoptosis and necrosis.
108 F.J. Gonzalez / Mutation Research 569 (2005) 101–110

duce ROS that can cause cell toxicity by poisoning [3] H. Zhou, P.D. Josephy, D. Kim, F.P. Guengerich, Functional
mitochondria or reacting with PUFAs. Alcohol and a characterization of four allelic variants of human cytochrome
fatty diet can affect the extent of toxicity by directly P4501A2, Arch. Biochem. Biophys. 422 (2004) 23–30.
[4] D. WuDunn, Genetic basis of glaucoma, Curr. Opin. Ophthal-
and indirectly inducing levels of CYP2E1 with the re- mol. 13 (2002) 55–60.
sultant increase in ROS. ROS stimulated cell toxicity [5] C.H. Yun, T. Shimada, F.P. Guengerich, Purification and char-
and cell death is influenced by a yet to be determined acterization of human liver microsomal cytochrome P-4502A6,
mechanism through activation of the EGFR/c-Raf path- Mol. Pharmacol. 40 (1991) 679–685.
way involving the MAPK ERK1/2. There remains the [6] S.A. Wrighton, E.G. Schuetz, K.E. Thummel, D.D. Shen, K.R.
Korzekwa, P.B. Watkins, The human CYP3A subfamily: prac-
prospect that inhibitors of CYP2E1 may be of value tical considerations, Drug Metab. Rev. 32 (2000) 339–361.
as therapeutic aids for treatment of ASH and NASH in [7] V. Rogiers, A. Vercruysse, Hepatocyte cultures in drug
intriguing [69]. metabolism and toxicological research and testing, Methods
Mol. Biol. 107 (1998) 279–294.
[8] F.P. Guengerich, T. Shimada, Activation of procarcinogens by
human cytochrome P450 enzymes, Mutat. Res. 400 (1998)
4. Conclusions 201–213.
[9] F.J. Gonzalez, K.R. Korzekwa, Cytochromes P450 expression
P450s are required for the metabolism and inacti- systems, Annu. Rev. Pharmacol. Toxicol. 35 (1995) 369–390.
vation of drugs and to facilitate the removal of xeno- [10] T.P. Dalton, M.Z. Dieter, R.S. Matlib, N.L. Childs, H.G.
Shertzer, M.B. Genter, D.W. Nebert, Targeted knockout of
biotics from the body. However, they are also required
Cyp1a1 gene does not alter hepatic constitutive expression of
for the metabolic activation of many toxicants and car- other genes in the mouse [Ah] battery, Biochem. Biophys. Res.
cinogens. CYP2E1 is able to activate a large number Commun. 267 (2000) 184–189.
of chemicals and is required for the heptotoxicity of [11] J.T. Buters, B.K. Tang, T. Pineau, H.V. Gelboin, S. Kimura, F.J.
toxins such as chloroform [70] and carbon tetrachlo- Gonzalez, Role of CYP1A2 in caffeine pharmacokinetics and
metabolism: studies using mice deficient in CYP1A2, Pharma-
ride [71]. CYP2E1 is also thought to play a role in
cogenetics 6 (1996) 291–296.
hepatic injury produced by ASH and NASH through [12] J.T. Buters, S. Sakai, T. Richter, T. Pineau, D.L. Alexander,
its ability of produced ROS. Therapeutic intervention U. Savas, J. Doehmer, J.M. Ward, C.R. Jefcoate, F.J. Gon-
through inhibition of CYP2E1 activity for treatment zalez, Cytochrome P450 CYP1B1 determines susceptibility
of liver toxicity could be possible since mice lacking to 7,12-dimethylbenz[a]anthracene-induced lymphomas, Proc.
Natl. Acad. Sci. U.S.A. 96 (1999) 1977–1982.
expression of the enzyme are normal.
[13] S.S. Lee, J.T. Buters, T. Pineau, P. Fernandez-Salguero, F.J.
Finally, CYP2E1 is able to produce oxidative stress Gonzalez, Role of CYP2E1 in the hepatotoxicity of ac-
directly through NADPH oxidase activity and directly etaminophen, J. Biol. Chem. 271 (1996) 12063–12067.
through metabolism of xenobiotics. Metabolic activa- [14] H. Zaher, J.T. Buters, J.M. Ward, M.K. Bruno, A.M. Lucas,
tion of many of its substrates can lead to activated S.T. Stern, S.D. Cohen, F.J. Gonzalez, Protection against ac-
etaminophen toxicity in CYP1A2 and CYP2E1 double-null
metabolites that damage cellular macromolecules.
mice, Toxicol. Appl. Pharmacol. 152 (1998) 193–199.
Both pathways can lead to DNA damage and, in same [15] D.R. Koop, J.P. Casazza, Identification of ethanol-inducible P-
cases, gene mutation and cell transformation. Thus, this 450 isozyme 3a as the acetone and acetol monooxygenase of
P450 is a critical mediator of toxicity such as that as- rabbit microsomes, J. Biol. Chem. 260 (1985) 13607–13612.
sociated with ASH and NASH and carcinogenicity of [16] A.M. Yu, J.R. Idle, L.G. Byrd, K.W. Krausz, A. Kupfer,
F.J. Gonzalez, Regeneration of serotonin from 5-
low Mr carcinogenic agents.
methoxytryptamine by polymorphic human CYP2D6,
Pharmacogenetics 13 (2003) 173–181.
[17] F.J. Gonzalez, S. Kimura, Study of P450 function using gene
References knockout and transgenic mice, Arch. Biochem. Biophys. 409
(2003) 153–158.
[18] C.J. Sinal, M. Miyata, M. Tohkin, K. Nagata, J.R. Bend, F.J.
[1] D.R. Nelson, D.C. Zeldin, S.M.G. Hoffman, L.J. Maltais,
Gonzalez, Targeted disruption of soluble epoxide hydrolase re-
H.M. Wain, D.W. Nebert, Comparison of cytochrome P450
veals a role in blood pressure regulation, J. Biol. Chem. 275
(CYP) genes from the mouse and human genomes, includ-
(2000) 40504–40510.
ing nomenclature recommendations for genes, pseudogenes and
[19] M. Miyata, G. Kudo, Y.H. Lee, T.J. Yang, H.V. Gelboin, P.
alternative-splice variants, Pharmacogenetics 14 (2004) 1–18.
Fernandez-Salguero, S. Kimura, F.J. Gonzalez, Targeted dis-
[2] F.P. Guengerich, Cytochromes p450, drugs, and diseases, Mol.
ruption of the microsomal epoxide hydrolase gene. Microso-
Intervent. 3 (2003) 194–204.
F.J. Gonzalez / Mutation Research 569 (2005) 101–110 109

mal epoxide hydrolase is required for the carcinogenic activity [33] W. Huang, J. Zhang, S.S. Chua, M. Qatanani, Y. Han, R.
of 7,12-dimethylbenz[a]anthracene, J. Biol. Chem. 274 (1999) Granata, D.D. Moore, Induction of bilirubin clearance by the
23963–23968. constitutive androstane receptor (CAR), Proc. Natl. Acad. Sci.
[20] V. Radjendirane, P. Joseph, Y.H. Lee, S. Kimura, A.J. Klein- U.S.A. 100 (2003) 4156–4161.
Szanto, F.J. Gonzalez, A.K. Jaiswal, Disruption of the DT di- [34] W. Xie, M.F. Yeuh, A. Radominska-Pandya, S.P. Saini, Y.
aphorase (NQO1) gene in mice leads to increased menadione Negishi, B.S. Bottroff, G.Y. Cabrera, R.H. Tukey, R.M. Evans,
toxicity, J. Biol. Chem. 273 (1998) 7382–7389. Control of steroid, heme, and carcinogen metabolism by nu-
[21] D.J. Long 2nd, K. Iskander, A. Gaikwad, M. Arin, D.R. Roop, clear pregnane X receptor and constitutive androstane receptor,
R. Knox, R. Barrios, A.K. Jaiswal, Disruption of dihydroni- Proc. Natl. Acad. Sci. U.S.A. 100 (2003) 4150–4155.
cotinamide riboside:quinone oxidoreductase 2 (NQO2) leads to [35] G.P. Lahvis, S.L. Lindell, R.S. Thomas, R.S. McCuskey, C.
myeloid hyperplasia of bone marrow and decreased sensitivity Murphy, E. Glover, M. Bentz, J. Southard, C.A. Bradfield, Por-
to menadione toxicity, J. Biol. Chem. 277 (2002) 46131–46139. tosystemic shunting and persistent fetal vascular structures in
[22] C.J. Henderson, A.G. Smith, J. Ure, K. Brown, E.J. Bacon, C.R. aryl hydrocarbon receptor-deficient mice, Proc. Natl. Acad. Sci.
Wolf, Increased skin tumorigenesis in mice lacking pi class glu- U.S.A. 97 (2000) 10442–10447.
tathione S-transferases, Proc. Natl. Acad. Sci. U.S.A. 95 (1998) [36] F. Andreola, G.P. Hayhurst, G. Luo, S.S. Ferguson, F.J. Gonza-
5275–5280. lez, J.A. Goldstein, DeLucaF L.M., Mouse liver CYP2C39 is a
[23] F.J. Gonzalez, Transgenic models in xenobiotic metabolism and novel retinoic acid 4-hydroxylase. Its down-regulation offers a
toxicology, Toxicology 181/182 (2002) 237–239. molecular basis for liver retinoid accumulation and fibrosis in
[24] P. Fernandez-Salguero, T. Pineau, D.M. Hilbert, T. McPhail, aryl hydrocarbon receptor-null mice, J. Biol. Chem. 279 (2004)
S.S. Lee, S. Kimura, D.W. Nebert, S. Rudikoff, J.M. Ward, 3434–3438.
F.J. Gonzalez, Immune system impairment and hepatic fibrosis [37] F.P. Guengerich, D.H. Kim, M. Iwasaki, Role of human cy-
in mice lacking the dioxin-binding Ah receptor, Science 268 tochrome P-450 IIE1 in the oxidation of many low molec-
(1995) 722–726. ular weight cancer suspects, Chem. Res. Toxicol. 4 (1991)
[25] J.V. Schmidt, G.H. Su, J.K. Reddy, M.C. Simon, C.A. Bradfield, 168–179.
Characterization of a murine Ahr null allele: involvement of [38] J. Wan, J. Shi, L. Hui, D. Wu, X. Jin, N. Zhao, W. Huang, Z.
the Ah receptor in hepatic growth and development, Proc. Natl. Xia, G. Hu, Association of genetic polymorphisms in CYP2E1,
Acad. Sci. U.S.A. 93 (1996) 6731–6736. MPO, NQO1, GSTM1, and GSTT1 genes with benzene poi-
[26] J. Mimura, K. Yamashita, K. Nakamura, M. Morita, T.N. Tak- soning, Environ. Health Perspect. 110 (2002) 1213–1218.
agi, K. Nakao, M. Ema, K. Sogawa, M. Yasuda, M. Katsuki, [39] M.Q. Bromer, M. Black, Acetaminophen hepatotoxicity, Clin.
Y. Fujii-Kuriyama, Loss of teratogenic response to 2,3,7,8- Liver Dis. 7 (2003) 351–367.
tetrachlorodibenzo-p-dioxin (TCDD) in mice lacking the Ah [40] D.J. Jollow, J.R. Mitchell, W.Z. Potter, D.C. Davis, J.R. Gillette,
(dioxin) receptor, Genes Cells 2 (1997) 645–654. B.B. Brodie, Acetaminophen-induced hepatic necrosis. II. Role
[27] P. Wei, J. Zhang, M. Egan-Hafley, S. Liang, D.D. Moore, The of covalent binding in vivo, J. Pharmacol. Exp. Ther. 187 (1973)
nuclear receptor CAR mediates specific xenobiotic induction of 195–202.
drug metabolism, Nature 407 (2000) 920–923. [41] J.R. Mitchell, D.J. Jollow, W.Z. Potter, D.C. Davis, J.R. Gillette,
[28] W. Xie, J.L. Barwick, M. Downes, B. Blumberg, C.M. Si- B.B. Brodie, Acetaminophen-induced hepatic necrosis. I. Role
mon, M.C. Nelson, B.A. Neuschwander-Tetri, E.M. Brunt, of drug metabolism, J. Pharmacol. Exp. Ther. 187 (1973)
P.S. Guzelian, R.M. Evans, Humanized xenobiotic response 185–194.
in mice expressing nuclear receptor SXR, Nature 406 (2000) [42] J.L. Holtzman, The effect of alcohol on acetaminophen hepa-
435–439. totoxicity, Arch. Intern. Med. 162 (2002) 1193.
[29] J.L. Staudinger, B. Goodwin, S.A. Jones, D. Hawkins-Brown, [43] C.J. Henderson, C.R. Wolf, N. Kitteringham, H. Powell, D.
K.I. MacKenzie, A. LaTour, Y. Liu, C.D. Klaassen, K.K. Brown, Otto, B.K. Park, Increased resistance to acetaminophen hep-
J. Reinhard, T.M. Willson, B.H. Koller, S.A. Kliewer, The nu- atotoxicity in mice lacking glutathione S-transferase Pi, Proc.
clear receptor PXR is a lithocholic acid sensor that protects Natl. Acad. Sci. U.S.A. 97 (2000) 12741–12745.
against liver toxicity, Proc. Natl. Acad. Sci. U.S.A. 98 (2001) [44] J. Zhang, W. Huang, S.S. Chua, P. Wei, D.D. Moore, Modula-
3369–3374. tion of acetaminophen-induced hepatotoxicity by the xenobiotic
[30] J. Staudinger, Y. Liu, A. Madan, S. Habeebu, C.D. Klaassen, receptor CAR, Science 298 (2002) 422–424.
Coordinate regulation of xenobiotic and bile acid homeosta- [45] J.M. Mates, C. Perez-Gomez, I. Nunez de, Castro, Antioxi-
sis by pregnane X receptor, Drug. Metab. Dispos. 29 (2001) dant enzymes and human diseases, Clin. Biochem. 32 (1999)
1467–1472. 595–603.
[31] W. Xie, A. Radominska-Pandya, Y. Shi, C.M. Simon, M.C. Nel- [46] K. Hensley, R.A. Floyd, Reactive oxygen species and protein
son, E.S. Ong, D.J. Waxman, R.M. Evans, An essential role for oxidation in aging: a look back, a look ahead, Arch. Biochem.
nuclear receptors SXR/PXR in detoxification of cholestatic bile Biophys. 397 (2002) 377–383.
acids, Proc. Natl. Acad. Sci. U.S.A. 98 (2001) 3375–3380. [47] A. Cahill, C.C. Cunningham, M. Adachi, H. Ishii, S.M. Bailey,
[32] S.A. Kliewer, T.M. Willson, Regulation of xenobiotic and bile B. Fromenty, A. Davies, Effects of alcohol and oxidative stress
acid metabolism by the nuclear pregnane X receptor, J. Lipid on liver pathology: the role of the mitochondrion, Alcohol Clin.
Res. 43 (2002) 359–364. Exp. Res. 26 (2002) 907–915.
110 F.J. Gonzalez / Mutation Research 569 (2005) 101–110

[48] M. Mari, A.I. Cederbaum, CYP2E1 overexpression in HepG2 [60] Z. Wang, S.D. Hall, J.F. Maya, L. Li, A. Asghar, J.C.
cells induces glutathione synthesis by transcriptional activa- Gorski, Diabetes mellitus increases the in vivo activity of cy-
tion of gamma-glutamylcysteine synthetase, J. Biol. Chem. 275 tochrome P4502E1 in humans, Br. J. Clin. Pharmacol. 55 (2003)
(2000) 15563–15571. 77–85.
[49] A.A. Caro, A.I. Cederbaum, Oxidative stress, toxicology, and [61] D. Lucas, C. Farez, L.G. Bardou, J. Vaisse, J.R. Attali, P. Valensi,
pharmacology of cyp2e1* , Annu. Rev. Pharmacol. Toxicol. 44 Cytochrome P4502E1 activity in diabetic and obese patients as
(2004) 27–42. assessed by chlorzoxazone hydroxylation, Fund. Clin. Pharma-
[50] G. Ekstrom, M. Ingelman-Sundberg, Rat liver microsomal col. 12 (1998) 553–558.
NADPH-supported oxidase activity and lipid peroxidation de- [62] E. Ip, G.C. Farrell, G. Robertson, P. Hall, R. Kirsch,
pendent on ethanol-inducible cytochrome P-450 (P-450IIE1), I. Leclercq, Central role of PPARalpha-dependent hepatic
Biochem. Pharmacol. 38 (1989) 1313–1319. lipid turnover in dietary steatohepatitis in mice, Hepatology
[51] M.D. Weltman, G.C. Farrell, C. Liddle, Increased hepato- 38 (2003) 123–132.
cyte CYP2E1 expression in a rat nutritional model of hep- [63] A.R. Bell, R. Savory, N.J. Horley, A.I. Choudhury, M. Dick-
atic steatosis with inflammation, Gastroenterology 111 (1996) ins, T.J. Gray, A.M. Salter, D.R. Bell, Molecular basis of
1645–1653. non-responsiveness to peroxisome proliferators: the guinea-pig
[52] I.A. Leclercq, G.C. Farrell, J. Field, D.R. Bell, F.J. Gonzalez, PPARalpha is functional and mediates peroxisome proliferator-
G.R. Robertson, CYP2E1 and CYP4A as microsomal catalysts induced hypolipidaemia, Biochem. J. 332 (Pt. 3) (1998)
of lipid peroxides in murine nonalcoholic steatohepatitis, J. 689–693.
Clin. Invest. 105 (2000) 1067–1075. [64] F.J. Gonzalez, J.M. Peters, R.C. Cattley, Mechanism of action
[53] G. Robertson, I. Leclercq, G.C. Farrell, Nonalcoholic steato- of the nongenotoxic peroxisome proliferators: role of the per-
sis and steatohepatitis. II. Cytochrome P-450 enzymes and ox- oxisome proliferator-activator receptor alpha, J. Natl. Cancer
idative stress, Am. J. Physiol. Gastrointest. Liver Physiol. 281 Inst. 90 (1998) 1702–1709.
(2001) 1135–1139. [65] H. Liu, M. Baliga, R. Baliga, Effect of cytochrome P4502E1
[54] C.S. Lieber, M.A. Leo, K.M. Mak, Y. Xu, Q. Cao, C. Ren, A. inhibitors on cisplatin-induced cytotoxicity to renal proximal
Ponomarenko, L.M. DeCarli, Model of nonalcoholic steatohep- tubular epithelial cells, Anticancer Res. 22 (2002) 863–868.
atitis, Am. J. Clin. Nutr. 79 (2004) 502–509. [66] Y. Dai, J. Rashba-Step, A.I. Cederbaum, Stable expression of
[55] M.L. Martinez-Chantar, F.J. Corrales, L.A. Martinez-Cruz, E.R. human cytochrome P4502E1 in HepG2 cells: characterization
Garcia-Trevijano, Z.Z. Huang, L. Chen, G. Kanel, M.A. Avila, of catalytic activities and production of reactive oxygen inter-
J.M. Mato, S.C. Lu, Spontaneous oxidative stress and liver mediates, Biochemistry 32 (1993) 6928–6937.
tumors in mice lacking methionine adenosyltransferase 1A, [67] J.M. Schattenberg, Y. Wang, R.M. Rigoli, D.R. Koop, M.J.
Faseb. J. 16 (2002) 1292–1294. Czaja, CYP2E1 overexpression alters hepatocyte death from
[56] B.J. Song, R.L. Veech, S.S. Park, H.V. Gelboin, F.J. Gonzalez, menadione and fatty acids by activation of ERK1/2 signaling,
Induction of rat hepatic N-nitrosodimethylamine demethylase Hepatology 39 (2004) 444–455.
by acetone is due to protein stabilization, J. Biol. Chem. 264 [68] M. Torres, H.J. Forman, Redox signaling and the MAP kinase
(1989) 3568–3572. pathways, Biofactors 17 (2003) 287–296.
[57] M.D. Weltman, G.C. Farrell, P. Hall, M. Ingelman-Sundberg, [69] C.S. Lieber, CYP2E1: from ASH to NASH, Hepatol. Res. 28
C. Liddle, Hepatic cytochrome P4502E1 is increased in pa- (2004) 1–11.
tients with nonalcoholic steatohepatitis, Hepatology 27 (1998) [70] A.A. Constan, C.S. Sprankle, J.M. Peters, G.L. Kedderis,
128–133. J.I. Everitt, B.A. Wong, F.L. Gonzalez, B.E. Butterworth,
[58] M.G. Emery, J.M. Fisher, J.Y. Chien, E.D. Kharasch, E.P. Metabolism of chloroform by cytochrome P4502E1 is re-
Dellinger, K.V. Kowdley, K.E. Thummel, CYP2E1 activity be- quired for induction of toxicity in the liver, kidney, and
fore and after weight loss in morbidly obese subjects with non- nose of male mice, Toxicol. Appl. Pharmacol. 160 (1999)
alcoholic fatty liver disease, Hepatology 38 (2003) 428–435. 120–126.
[59] N. Chalasani, J.C. Gorski, M.S. Asghar, A. Asghar, B. Fores- [71] F.W. Wong, W.Y. Chan, S.S. Lee, Resistance to car-
man, S.D. Hall, D.W. Crabb, Hepatic cytochrome P4502E1 ac- bon tetrachloride-induced hepatotoxicity in mice which lack
tivity in nondiabetic patients with nonalcoholic steatohepatitis, CYP2E1 expression, Toxicol. Appl. Pharmacol. 153 (1998)
Hepatology 37 (2003) 544–550. 109–118.

You might also like