Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

TOXICOLOGY AND APPLIED PHARMACOLOGY 153, 102–108 (1998)

ARTICLE NO. TO988543

Role of CYP1A2 in the Hepatotoxicity of Acetaminophen:


Investigations Using Cyp1a2 Null Mice
Robert P. Tonge,*,1 Edward J. Kelly,† Sam A. Bruschi,* Tom Kalhorn,‡ David L. Eaton,† Daniel W. Nebert,§ and
Sidney D. Nelson*,2
*Department of Medicinal Chemistry, †Department of Environmental Health, ‡Department of Pharmaceutics, University of Washington, Seattle, Washington
98195–7631; and §Department of Environmental Health, University of Cincinnati Medical Center, Cincinnati, Ohio 45267– 0056

Received April 30, 1998; accepted August 6, 1998

Key Words: acetaminophen; APAP; Cyp1a2; knockout mice;


Role of CYP1A2 in the Hepatotoxicity of Acetaminophen: In- protein-adducts; hepatotoxicity.
vestigations Using Cyp1a2 Null Mice. Tonge, R. P., Kelly, E. J.,
Bruschi, S. A., Kalhorn, T., Eaton, D. L., Nebert, D. W., and
Nelson, S. D. (1998). Toxicol. Appl. Pharmacol. 153, 102–108.
Acetaminophen (APAP; paracetamol) is a widely used an-
Acetaminophen (APAP) is known to cause centrilobular he-
algesic and antipyretic drug that is relatively safe at normal
patic necrosis under overdose conditions. This is thought to be
mediated via the P450-generated reactive intermediate therapeutic dose levels but can lead to severe centrilobular
N-acetyl-p-benzoquinone imine (NAPQI). Initially, NAPQI is hepatic necrosis in man (Davidson and Eastham, 1966) and
detoxified by conjugation with glutathione (GSH), but once experimental animals (Boyd and Bereczky, 1966) following
GSH is depleted, NAPQI reacts more extensively with hepatic overdose. This toxicity is thought to be mediated via the
proteins leading to hepatocellular damage. The P450 isoforms cytochrome P450-generated electrophilic intermediate
thought to be responsible for APAP hepatotoxicity in humans N-acetyl-p-benzoquinone imine (NAPQI) (Dahlin et al., 1984)
are CYP2E1, CYP1A2, and CYP3A4, and thus, we have inves-
which, under normal dose conditions, is detoxified by conju-
tigated the effect of murine Cyp1a2 on APAP hepatotoxicity
using Cyp1a2 knockout mice (Liang et al., Proc. Natl. Acad. gation with glutathione (GSH). However, in overdose situa-
Sci. USA 93, 1671–1676, 1996). Doses of 250 mg/kg were mark- tions cellular GSH is depleted to an extent where further
edly hepatotoxic in these mice, and surprisingly, deaths only NAPQI production leads to hepatocellular protein adduction
occurred in the knock-out and heterozygote mice over a 24-h and toxicity (Jollow et al., 1973; Mitchell et al., 1973).
period after dosing. Furthermore, there were no significant The main P450 isoforms currently thought to be responsible
differences among survivors of any genotype in serum ALT for APAP bioactivation and, thus, hepatotoxicity are CYP2E1,
concentrations, a well correlated indicator of APAP hepatotox-
CYP1A2, and CYP3A4 (Raucy et al., 1989; Patten et al., 1993;
icity in mice. Finally, no differences were observed in the
urinary metabolites excreted over the 24-h period, including
Thummel et al., 1993; Nelson et al., 1995). An exciting new
those derived from GSH conjugation of the major reactive approach is now possible to evaluate the relative roles played
metabolite NAPQI. Consistent with the effects on hepatotoxic- by each isoform further, i.e., using specific P450 knockout
ity and metabolism, 2 h after hepatotoxic doses (500 mg/kg, ip) mice. The effect of mouse Cyp2e1 genotype on APAP toxicity
of APAP no significant differences were observed in total whole has already been evaluated, and the results suggested that
liver homogenate nonprotein thiol concentrations among the Cyp2e1 is the major P450 in mice responsible for hepatotox-
three genotypes even though hepatic thiols were decreased icity caused by APAP, although other P450 isoforms may also
compared to control animals (>90%). In addition, when the
contribute (Lee et al., 1996).
liver cytosol and microsome samples were examined by immu-
noblotting for the presence of APAP-protein adducts using a Here we report our investigations using Cyp1a2 knockout
specific antiserum, there were no observable differences in ei- mice. These animals have already been shown to metabolize
ther the intensity of staining or in the spectrum of adducts the muscle relaxant zoxazolamine in a Cyp1a2-dependent fash-
formed between APAP-dosed mice of any genotype. The cumu- ion (Liang et al., 1996). In addition, these Cyp1a2 null mice
lative data suggest that Cyp1a2 does not play a significant role have been used to demonstrate the necessity of this isozyme in
in APAP hepatotoxicity in these mice. © 1998 Academic Press the induction of murine uroporphyria (Sinclair et al., 1998) and
1
the unimportance of Cyp1a2 in Cyp1a1 expression (Liang et
Present address: Proteomics Group, I4.24 CTL, Zeneca Pharmaceuticals,
al., 1997). Similar Cyp1a2 knockout mice (Pineau et al., 1995)
Alderley Park, Macclesfield, Cheshire, SK10 7TG, England, UK.
2
To whom correspondence should be addressed. Fax: (206) 685-9297; have been used to demonstrate the involvement of Cyp1a2 in
E-mail: sidnels@u.washington.edu. caffeine metabolism (Buters et al., 1996) and in the hepatic

0041-008X/98 $25.00 102


Copyright © 1998 by Academic Press
All rights of reproduction in any form reserved.
ACETAMINOPHEN HEPATOTOXICITY IN Cyp1a2 NULL MICE 103

sequestering of 2,3,7,8-tetrachlorodibenzo-p-dioxin and mg/L leupeptin, and 1 mM phenylmethanesulfonyl fluoride (PMSF)), and 0.6
2,3,4,7,8-pentachlorodibenzofuran (Diliberto et al., 1997). ml of homogenate was frozen in liquid N2 for nonprotein thiol measurements.
The remaining liver homogenate was used to prepare cytosolic and microsomal
Following APAP exposure, toxicity was evaluated in each fractions by standard procedures (Fleischer and Kervina, 1974). Briefly, the
Cyp1a2 genotype by estimating nonprotein thiol depletion and liver homogenate was centrifuged at 250g for 10 min. The pellet (unbroken
APAP-covalent binding in the liver along with serum alanine cells and connective tissue) was discarded and the supernatant was centrifuged
aminotransferase (ALT) activity. APAP-covalent binding was at 14,000g for 10 min. The resulting pellet (nuclei, plasma membrane, and
determined immunochemically using a specific affinity puri- mitochondria) was discarded and the supernatant was centrifuged at 105,000g
for 60 min. The remaining supernatant (cytosol) was snap frozen in liquid N2,
fied anti(APAP) antiserum raised in our laboratory (Tonge et and the pellet was resuspended in 10 ml homogenization buffer and centri-
al., 1997). Analysis of the urine for all major APAP metabo- fuged at 105,000g for 60 min. The supernatant was discarded, and the pellet
lites, including NAPQI-derived thioether metabolites as bio- (microsomes) was resuspended in 1 ml homogenization buffer and snap frozen
markers of NAPQI formation (Prescott et al., 1981; Sarich et in liquid N2. All centrifugation procedures were carried out at 4°C and all
al., 1997), was also performed. Results of these studies indicate samples were stored at 270°C prior to analysis.
that Cyp1a2 does not play a significant role in APAP hepato- Urine analysis. The urine samples, including cage washes, were lyophi-
toxicity in mice in contrast to other P450 isoforms known to lized to dryness and redissolved in 20 ml water. Any solid material was
removed from the sample by centrifugation (500g 10 min). Urine was hydro-
more efficiently oxidize APAP to its toxic metabolite. These lyzed with b-glucuronidase/arylsulfatase (Helix Pomatia crude preparation) by
data are consistent with studies in humans which have shown diluting 0.5 ml 1:1 with 200 mM sodium acetate buffer (pH 5) and incubating
that induction of CYP1A2 does not significantly increase the overnight at 37°C with 40 ml enzyme. Both hydrolyzed and unhydrolyzed
clearance of APAP through the formation of NAPQI (Miners et samples were filtered and deproteinated by passage through a 10-kDa cutoff
al., 1984; Sarich et al., 1997). membrane (Alltech, Deerfield, IL). Urinary metabolites were analyzed by
injection of 10 ml supernatant onto a HPLC [2 Waters 510 HPLC pumps,
Waters Automated Gradient Controller, Waters 484 Tunable Absorbance De-
MATERIALS AND METHODS tector (Waters Chromatography, Fairfax, VA), HP3393A Integrator (Hewlett
Packard, Wilmington, DE)] equipped with a C18 reversed-phase column (4.6 3
Chemicals. All chemicals were from Sigma Chemical Co. (St. Louis, MO) 100 mm, 3 mm, 100 Å, Microsorb-MV, Rainin, Woburn, MA) and UV
unless otherwise specified. Electrophoresis and Western blotting reagents were absorbance monitored at 254 nm. Elution was achieved using the following
from BioRad (Hercules, CA). program: 0 min, 93% A, 7% B; 11 min, 93% A, 7% B; 20 min, 70% A, 30%
Animal husbandry and genotyping. The Cyp1a2 knockout mice were B; 25 min, 0% A, 100% B; 35 min, 93% A, 7% B (Buffer A, 20 mM
propagated by interbreeding heterozygotes to allow littermate comparisons of ammonium phosphate/0.1% acetic acid, pH 3.2; Buffer B, acetonitrile). Me-
all genotypes. Animals were weaned at 3– 4 weeks of age and a small portion tabolite identification was achieved by comparison of retention times with
(;1 cm) of tail was excised for genotyping. The tissue was digested overnight those of authentic standards produced in our laboratory. Metabolite quantita-
at 37°C in 2 ml 10 mM Tris/HCl (pH 7.4) containing 1% SDS, 5 mM EDTA, tion was achieved by determining the relative UV response of each metabolite
150 mM NaCl, and 200 mg/ml Proteinase K (Boehringer-Mannheim, Mann- to that of APAP and by then determining standard curves for APAP in our
heim, Germany). Total nucleic acid (TNA) was precipitated by ethanol addi- HPLC method (Sarich et al., 1997). Typical chromatograms are presented in
tion, resuspended in 10 mM Tris/HCl (pH 7.4) containing 1 mM EDTA, and Fig. 1.
heated to 100°C to inactivate the Proteinase K. The TNA was then subjected Serum ALT estimation. ALT activity in 5 ml serum was determined using
to PCR amplification utilizing Cyp1a2-specific oligonucleotide primers de- standard commercial methods (Sigma Diagnostics, Procedure No. DG159-
scribed previously (Liang et al., 1996). The PCR reaction buffer contained 10 UV).
mM Tris/HCl (pH 8.4), 1.5 mM MgCl2, 50 mM KCl, 200 mM dG/A/T/C, 1
Hepatic nonprotein thiol estimation. Protein was precipitated from 0.6 ml
mM of each primer, and 1 unit of Taq polymerase (Boehringer Mannheim) in
whole liver homogenate by adding 0.6 ml 4% sulfosalicylic acid, vortexing,
a final volume of 30 ml. The samples were overlaid with a drop of paraffin oil,
and centrifuging for 10 min at 16,000g. Supernatant (0.2 ml) was mixed with
and PCR was run in a thermal cycler (M. J. Research, Watertown, MA) using
1.5 ml 0.1 mM Ellman’s reagent [5,59-dithio-bis(2-nitrobenzoic acid)] and
standard conditions (94°C, 30 s; 62°C, 45 s; 72°C, 60 s repeated for 35 cycles).
allowed to stand for 15 min. After this time, absorbance was read at 412 nm.
The products were then analyzed by agarose gel electrophoresis and visualized
Quantitation was possible using a GSH standard curve.
using ethidium bromide staining.
APAP toxicity. Male wild-type [Cyp1a2(1/1)], heterozygous Immunoblot analysis. SDS-PAGE was performed as described elsewhere
[Cyp1a2(1/2)], and knockout [Cyp1a2(2/2)] mice (2– 4 months old) were (Ausubel et al., 1993) using a discontinuous buffer system and minigels (5.5
housed in modified rat metabolism cages (Nalgene, Naperville, IL), 2 animals cm, 4% stacking gel and 10% resolving gel) (Mini-Protean II, BioRad).
of similar genotype per cage, and fasted overnight prior to dosing, APAP (25 Sample (50 mg) was loaded into each sample well. After electrophoresis, the
mg/ml in phosphate-buffered saline, pH 7.4) was given at 250 or 500 mg/kg proteins were transferred electrophoretically to nitrocellulose (2 h, 4°C, 100
(ip). Animals receiving 250 mg/kg (n 5 18) were killed by cervical dislocation V). APAP-modified proteins were detected by incubating the nitrocellulose at
24 h after dosing, and blood, liver, and urine samples were taken. Urine was 4°C for 3 h sequentially with 1:200 diluted affinity-purified anti(APAP)
collected over 50 mg ascorbic acid into an ice-cooled container to minimize antiserum (Tonge et al., 1997) and 1:1000 diluted alkaline phosphatase-
atmospheric oxidation of urinary metabolites. Residual urine remaining in the conjugated anti-rabbit IgG (Sigma) using previously described techniques
bladder of the mice was taken and added to that in the metabolism cage. After (Kenna et al., 1987). Bound antibodies were visualized colorimetrically using
the experiment the metabolism cages were washed down with deionized water NBT/BCIP (Promega Co., Madison, WI) (Ausubel et al., 1993). Immunoblots
to recover urine remaining on the walls or floor of the cage. Animals receiving were digitized using a flat-bed scanner (300 d.p.i.), and densitometric analysis
500 mg/kg (n 5 22) were killed 2 h after dosing, and liver samples were was performed on a Macintosh computer using the public domain NIH Image
collected. Serum was prepared from blood samples for ALT measurements. program (available at http://rsb.info.nih.gov/nih-image/).
Livers were weighed and homogenized into 5 ml ice cold homogenization Statistical analysis. Students t tests were carried out where appropriate on
buffer (10 mM Tris–HCl (pH 7.4) containing 0.25 M sucrose, 1 mM EDTA, a Macintosh computer with Statworks Version 1.2 (Cricket Software, Phila-
0.5 g/L bovine serum albumin, 2.5 mg/L aprotinin, 2.5 mg/L antipain, 2.5 delphia, PA).
104 TONGE ET AL.

FIG. 2. Effect of mouse Cyp1a2 genotype on the urinary excretion of


APAP. Mice were dosed with 250 mg/kg APAP (ip) (n 5 18), and their urine
was collected for 24 h. Three urine samples (n 5 3) from each genotype were
analyzed by HPLC. Each urine sample was pooled from 2 animals.

dosing of the animals), there were no statistically significant


differences in serum ALT levels among genotypes. The mean
FIG. 1. Typical HPLC chromatograms of urinary metabolites of APAP. ALT levels were 15,804 6 5345 U/L (mean 6 SD, n 5 5),
Urine was analyzed undiluted (unhydrolyzed) or diluted (1:1) before analysis 14,485 6 1851 U/L (mean 6 SD, n 5 5), and 13,712 6 5254
(hydrolyzed). A, APAP glucuronide (4 min); B, 3-hydroxy APAP (7.8 min); C, U/L (mean 6 SD, n 5 4) for (1/1), (1/2), and (2/2) mice,
APAP sulfate (9.5 min); D, 3-(cystein-S-yl) APAP (APAP-Cys) (11.3 min); E, respectively. Serum obtained from 2 mice at the point of death
APAP (12.1 min); F, 3-methoxy APAP (16.7 min); G, 3-(N-acetyl cystein-S-
contained ALT activities among the highest measured (Fig. 3).
yl) APAP (APAP-NAC) (22.6 min).
Serum obtained from control mice contained no measurable
ALT activity.
RESULTS Hepatic nonprotein thiols. Following exposure of each
genotype to 500 mg/kg APAP for 2 h, there was a massive
Urine analysis. Mice were exposed to 250 mg/kg APAP decrease in hepatic nonprotein thiols (.90.1%) compared to
for 24 h because this was the maximum tolerated dose for this
duration in the pilot study. Despite this, 4 animals [2 (1/2)
and 2 (2/2)] died during the experiment between 6 and 11 h.
There were no statistically significant differences in any
APAP-derived urinary metabolite assayed between genotypes
(Fig. 2). The major APAP-derived urinary metabolite in each
genotype was APAP-glucuronide (53.1–59.1% total metabo-
lites), and the APAP-GSH derived metabolites (APAP-Cys and
APAP-NAC) varied between 19.2% and 21.4%. Unmetabo-
lized urinary APAP was between 7.8% and 10.8% of total
administered dose. Metabolite recovery for (1/1) mice was
16.2 6 3.4 g (95.3 6 19.8%), for (1/2) mice was 16.4 6 2.2 g
(88.2 6 11.8%), and for (2/2) mice was 19.1 6 2.9 g (101 6
15.5%) (mean 6 SD, n 5 3).
Serum ALT. The majority of mice exposed to APAP at 250
mg/kg for 24 h showed elevated serum ALT activities (Fig. 3) FIG. 3. Effect of mouse Cyp1a2 genotype on serum ALT activity follow-
although this effect was variable. Activities ranged from 453 to ing APAP exposure. Mice were dosed with 250 mg/kg APAP ip (n 5 18), and
a blood sample was taken after 24 h for ALT estimation. Figure shows ALT
22,034 U/L, 12,218 to 17,364 U/L, and 0 to 18,656 U/L for levels for individual animals. Black crosses represent animals that died during
(1/1), (1/2), and (2/2) mice, respectively. If the outlying the dose period from which serum could be obtained (n 5 2). Control animals
points at 0 and 453 U/L were disregarded (possibly due to low showed zero serum ALT activities (data not shown).
ACETAMINOPHEN HEPATOTOXICITY IN Cyp1a2 NULL MICE 105

Cyp1a2 genotype had a negligible effect on the quantity of


either microsomal or cytosolic adducts formed (Fig. 4B). Fur-
ther, there were no observable differences in the spectrum of
proteins modified by APAP in any genotype (Fig. 5). The main
APAP-protein adducts had molecular weights of 108, 77, 50,
47, 39, and 34 kDa in cytosol and 142, 117, 105, 78, 73, 70,
and 38 kDa in microsomes. There were no antibody detectable
proteins in the control microsomal samples, but a minor, non-
specific, 34-kDa protein was detected in control cytosolic
samples (Fig. 5).

DISCUSSION

Previous investigations have suggested that the human P450


isoforms responsible for APAP bioactivation are CYP2E1,
CYP1A2, and CYP3A4 (Raucy et al., 1989; Patten et al., 1993;
Thummel et al., 1993; Nelson et al., 1995). However, this
investigation has demonstrated that Cyp1a2 plays a negligible
role in the hepatotoxicity of acetaminophen in the mouse under
the conditions used in this study.

FIG. 4. Effect of mouse Cyp1a2 genotype on hepatic nonprotein thiol


depletion (A) and on the formation of APAP-hepatic protein adducts (B)
following APAP exposure. Mice were dosed with 500 mg/kg APAP ip (n 5
22) and were killed 2 h later. Livers were removed, homogenized, and
analyzed for the presence of GSH. Cytosolic and microsomal fractions were
prepared from the liver homogenate and analyzed immunochemically for the
presence of APAP-protein adducts. The quantity of adducts was assessed by
the relative density of staining on immunoblots. Numbers in parentheses in A
represent numbers of animals in each group. *Significantly different than
(1/2) microsomes ( p , 0.05).

control mice (Fig. 4). Concentrations of nonprotein thiols per


gram wet wt liver in control tissues were 0.55 6 0.05 mM/g,
0.59 6 0.10 mM/g, and 0.54 6 0.15 mM/g for (1/1), (1/2),
and (2/2) mice, respectively. In comparison, those in APAP-
treated animals were 0.04 6 0.01 mM/g, 0.05 6 0.01 mM/g,
and 0.05 6 0.01 mM/g, respectively (mean 6 SD, see Fig. 4A
for number of replicates). There were no statistical differences
in nonprotein thiol levels between mice of any genotype either FIG. 5. Immunoblots showing APAP-protein adducts in cytosol and mi-
before or after APAP treatment (Fig. 4A). crosomes of control (C) and APAP-exposed mice of different Cyp1a2 geno-
type. The migration of prestained molecular weight standards are indicated on
Hepatic APAP-protein adducts. Immunochemical analy- the left of each panel, and the estimated molecular weight (kDa) of APAP-
ses for the presence of APAP-hepatic protein adducts in control modified immunoreactive proteins for all genotypes are shown on the right of
and APAP-treated mice (500 mg/kg 2 h) revealed that the each panel.
106 TONGE ET AL.

APAP hepatotoxicity has often been estimated by measuring synthetase (Bulera et al., 1995). It is possible that our major
the levels of hepatic enzymes such as transaminases, and it is 38-kDa adduct could be 44-kDa glutamine synthetase and that
generally accepted that such measurements are a reliable indi- the unidentified adducts that we observed at 70.5, 78, and 105
cator of the tissue damage (Placke et al., 1987; Pumford et al., kDa could be similar to those seen at 72, 76, and 105 kDa in
1989; Roberts et al., 1991). The high serum ALT levels in mice CD1 mice.
24 h following APAP exposure (250 mg/kg) clearly indicate The major APAP adducts seen in our cytosolic fractions
APAP-mediated hepatotoxicity in these animals, but this effect were at 39, 47, 50, 77, and 108 kDa. It is possible that the
was not dependent on Cyp1a2 genotype. This finding was observed cytosolic 39-kDa adduct is glyceraldehyde-3-phos-
further substantiated by the similarity in urinary excretion of phate dehydrogenase but the others remain unidentified at this
the NAPQI-derived metabolites APAP-Cys and APAP-NAC time. The cytosolic adduct pattern observed in these studies is,
between different genotypes. Generally, APAP is primarily however, very similar to that seen following the exposure of
excreted as the glucuronide and sulfate conjugates (Brodie and male B6C3/F1 mice to [14C] APAP (400 mg/kg ip 2 h) by
Axelrod, 1948), but a proportion is oxidized to NAPQI in the phosphorimaging in our laboratory (Dietze et al., 1997). This
liver by cytochromes P450 (Mitchell et al., 1973a; Potter et al., observation lends additional evidence in support of the speci-
1973; Dahlin et al., 1984). NAPQI is detoxified by conjugation ficity of our affinity-purified anti(APAP) antiserum (Tonge et
with GSH but can react extensively with cellular macromole- al., 1997).
cules when GSH is depleted. This leads to the characteristic Cyp2e1 knockout mice have been used to examine the role
centrilobular necrosis seen in APAP hepatotoxicity (Jollow et played by Cyp2e1 in APAP activation (Lee et al., 1996), and
al., 1973; Mitchell et al., 1973b). APAP-GSH is subsequently it was demonstrated that these animals were considerably less
excreted in the bile (Hinson et al., 1982) and forms APAP-Cys sensitive to APAP-induced hepatotoxicity than wild-type ani-
by the action of g-glutamyltranspeptidase and aminopeptidase. mals. Minimal lethality was observed after 48 h to a dose of
APAP-Cys can be acetylated in the kidney by N-acetyl trans- 400 mg/kg APAP in Cyp2e1(2/2) animals, whereas .50% of
ferase to give the mercapturate, APAP-NAC. APAP-Cys and the wild-type Cyp2e1 animals died at this dose level. When the
APAP-NAC are detected in the urine following APAP expo- APAP dose was increased to .600 mg/kg, toxicity was also
sure (Jollow et al., 1974; Wilson et al., 1982) and are good observed in the Cyp2e1(2/2) mice. This suggests that al-
indicators of NAPQI formation (Prescott et al., 1981, 1983; though Cyp2e1 primarily mediates APAP hepatotoxicity, other
Sarich et al., 1997). P450s such as Cyp1a2, with a higher Km for APAP than
When administered at 500 mg/kg, APAP is a well docu- Cyp2e1, may mediate APAP toxicity at very high doses. Since
mented hepatotoxin in mice (Jollow et al., 1973), and APAP- our animals exhibited marked hepatotoxicity and death at doses
covalent binding and GSH depletion are classical biomarkers well below 600 mg/kg, it was impossible to evaluate the effect
of APAP hepatotoxicity (Jollow et al., 1973; Mitchell et al., of Cyp1a2 genotype at such high doses. Such an evaluation
1973). For example, covalent binding and GSH depletion is would be relevant only in a situation where Cyp2e1 was either
highest in species most susceptible to APAP hepatotoxicity absent or inhibited.
(Davis et al., 1974). In these studies Cyp1a2 genotype had no The relevance of the information obtained at hepatotoxic
effect on either of these parameters following APAP adminis- doses of APAP in animal model systems to humans will have
tration, and thus, these findings also suggest that Cyp1a2 has to await studies with human hepatocytes. Studies with human
only a minor role in the bioactivation of APAP at concentra- liver microsomes (Raucy et al., 1989) and with transfected
tions likely to be hepatotoxic. HepG2 cells (Patten et al., 1993) suggest that both human
Several groups have investigated and identified the forma- CYP2E1 and CYP1A2 are important for APAP oxidation at
tion of APAP-protein adducts in mice by immunochemical, high concentrations. In addition, studies with purified
radiochemical, and fluorescent means. After doses of APAP in CYP3A4, CYP2E1, and CYP2A6 (Thummel et al., 1993; Chen
vivo, the adducted proteins so far identified include a 56-kDa et al., 1998) suggest that CYP3A4 is the major isoform cata-
selenium binding protein of unknown function (Bartolone et lyzing APAP oxidation to NAPQI at therapeutic concentrations
al., 1992; Pumford et al., 1992), a 74-kDa nuclear lamin A (;0.1 mM). This is in contrast to the relative isozyme contri-
(Hong et al., 1994), a 44-kDa glutamine synthetase (Bulera et butions at higher APAP concentrations (1–10 mM), i.e.,
al., 1995), a 50-kDa glutamate dehydrogenase (Halmes et al., CYP2E1. CYP2A6. CYP1A2. Interestingly, induction of
1996), a 54-kDa aldehyde dehydrogenase (Landin et al., 1996), CYP1A2 in humans by either cigarette smoking (Miners et al.,
a 100-kDa N10-formyl tetrahydrofolate dehydrogenase (Pum- 1984) or by the widely used drug omeprazole (Sarich et al.,
ford et al., 1997), a 38-kDa glyceraldehyde-3-phosphate dehy- 1997) did not increase the formation of NAPQI after therapeu-
drogenase (Dietze et al., 1997), and a 130-kDa carbamyl tic doses of APAP.
phosphate synthetase I (Gupta et al., 1997). Other workers In conclusion, it would appear that Cyp1a2 does not play a
have demonstrated liver microsomal APAP-protein adducts of significant role in APAP hepatotoxicity in mice as long as other
25, 44, 52, 72, 76, and 105 kDa in CD1 mice (Bulera et al., P450s are present and that this is likely to be the case in
1996) and went on to identify the 44-kDa adduct as glutamine humans.
ACETAMINOPHEN HEPATOTOXICITY IN Cyp1a2 NULL MICE 107

ACKNOWLEDGMENTS 3-(Glutathion-S-yl) acetaminophen: A biliary metabolite of acetaminophen.


Drug Metab. Dispos. 10, 47–50.
This work was supported by National Institutes of Health Grants GM25418 Hong, M., Cohen, S. D., and Khairallah, E. A. (1994). Translocation of the
and GM32165 (S. D. Nelson), ES07033 (S. D. Nelson and D. L. Eaton), and major cytosolic acetaminophen (APAP) protein adducts into the nucleus.
ESO6321 (D. W. Nebert). Toxicologist 14, 427 (1691).
Jollow, D. J., Thorgiersson, S. S., Potter, W. Z., Hashimoto, M., and Mitchell,
REFERENCES J. R. (1974). Acetaminophen induced hepatic necrosis. VI. Metabolic dis-
position of toxic and non-toxic doses of acetaminophen. Pharmacology 12,
Ausubel, F. M., Brent, R., Kingston, R. E., Moore, D. D., Seidman, J. G., 251–271.
Smith, J. A., and Struhl, K. (Eds.) (1993). Current Protocols in Molecular Jollow, D. J., Mitchell, J. R., Potter, W. Z., Davis, D. C., Gillette, J. R., and
Biology, Vol. 2. John Wiley, Canada. Brodie, B. B. (1973). Acetaminophen-induced hepatic necrosis. II. Role of
Bartolone, J. B., Birge, R. B., Bulera, S. J., Bruno, M. K., Nishanian, E. V., covalent binding in vivo. J. Pharmacol. Exp. Ther. 187, 195–202.
Cohen, S. D., and Khairallah, E. A. (1992). Purification, antibody production Kenna, J. G., Neuberger, J., and Williams, R. (1987). Identification by immu-
and partial amino acid sequence of the 58-kDa acetaminophen-binding liver noblotting of 3 halothane-induced microsomal liver polypeptide antigens
proteins. Toxicol. Appl. Pharmacol. 113, 19 –29. recognised in sera from patients with halothane-associated hepatitis.
Boyd, E. M., and Bereczky, G. M. (1966). Liver necrosis from paracetamol. J. Pharmacol. Exp. Ther. 242, 733–740.
Br. J. Pharmacol. 26, 606 – 614. Landin, J. S., Cohen, S. D., and Khairallah, E. A. (1996). Identification of a
Brodie, B. B., and Axelrod, J. (1948). The estimation of acetanilide and its 54-kDa mitochondrial acetaminophen-binding protein as aldehyde dehydro-
metabolic products, aniline, N-acetyl-p-aminophenol and p-aminophenol genase. Toxicol. Appl. Pharmacol. 141, 299 –307.
(free and total conjugated) in biological fluids and tissues. J. Pharmacol. Lee, S. S. T., Buters, J. T. M., Pineau, T., Fernandez-Salguero, P., and
Exp. Ther. 94, 22–28. Gonzalez, F. J. (1996). Role of CYP2E1 in the hepatotoxicity of acetamin-
Bulera, S. J., Cohen, S. D., and Khairallah, E. A. (1996). Acetaminophen ophen. J. Biol. Chem. 271, 12063–12067.
arylated proteins are detected in hepatic subcellular fractions and numerous Liang, H. C., McKinnon, R. A., and Nebert, D. W. (1997). Sensitivity of
extra-hepatic tissues in CD-1 and C57B1/6J mice. Toxicology 109, 85–99. CYP1A1 mRNA inducibility by dioxin is the same in (1/1) wild-type and
Bulera, S. J., Birge, R. B., Cohen, S. D., and Khairallah, E. A. (1995). Cyp1a2(2/2) null mutant mice. Biochem. Pharmacol. 54, 1127–1131.
Identification of the mouse liver 44-kDa acetaminophen-binding protein as Liang, H. C. L., Li, H., McKinnon, R. A., Duffy, J. J., Potter, S. S., Puga, A.,
a subunit of glutamine synthetase. Toxicol. Appl. Pharmacol. 134, 313–320. and Nebert, D. W. (1996) Cyp1a2(2/2) null mutant mice develop normally
Buters, J. T. M., Tang, B., Pineau, T., Gelboin, H. V., Kimura, S., and but show deficient drug metabolism. Proc. Natl. Acad. Sci. USA 93, 1671–
Gonzalez, F. J. (1996). Role of CYP1A2 in caffeine pharmacokinetics and 1676.
metabolism: Studies using mice deficient in CYP1A2. Pharmacogenetics 6, Miners, J. O., Attwood, J., and Birkett, D. J. (1984). Determinants of acet-
291–296. aminophen metabolism: Effect of inducers and inhibitors of drug metabo-
Chen, W., Koenigs, L. L., Thompson, S. J., Peter, R. M., Rettie, A. E., Trager, lism on acetaminophen’s metabolic pathways. Clin. Pharmacol. Ther. 35,
W. F., and Nelson, S. D. (1998). Oxidation of acetaminophen and its toxic 480 – 486.
quinone imine and nontoxic catechol metabolites by baculovirus-expressed Mitchell, J. R., Jollow, D. J., Potter, W. Z., Davis, D. C., Gillette, J. R., and
and purified human cytochromes P450 2E1 and 2A6. Chem. Res. Toxicol. Brodie, B. B. (1973a). Acetaminophen-induced hepatic necrosis. I. Role of
11, 295–301. drug metabolism. J. Pharmacol. Exp. Ther. 187, 185–194.
Dahlin, D. C., Miwa, G. T., Lu, A. Y. H., and Nelson, S. D. (1984). N-acetyl- Mitchell, J. R., Jollow, D. J., Potter, W. Z., Gillette, J. R., and Brodie, B. B.
p-benzoquinone imine: A cytochrome P450-mediated oxidation product of (1973b). Acetaminophen-induced hepatic necrosis. IV. Protective role of
acetaminophen. Proc. Natl. Acad. Sci. USA 81, 1327–1331. glutathione. J. Pharmacol. Exp. Ther. 187, 211–217.
Davidson, D. G. D., and Eastham, W. N. (1966). Acute liver necrosis following Nelson, S. D. (1995). Mechanism of the formation and disposition of reactive
overdose of paracetamol. Br. Med. J. 5512, 497– 499. metabolites that can cause acute liver injury. Drug Metab. Rev. 27, 147–177.
Davis, D. C., Potter, W. Z., Jollow, D. J., and Mitchell, J. R. (1974). Species Patten, C. J., Thomas, P. E., Guy, R. L., Lee, M., Gonzalez, F. J., Guengerich,
differences in hepatic glutathione depletion, covalent binding and hepatic F. P., and Yang, C. S. (1993). Cytochrome P450 enzymes involved in
necrosis after acetaminophen. Life Sci. 14, 2099 –2109. acetaminophen activation by rat and human liver microsomes and their
Dietze, E. C., Schäfer, A., Omichinski, J. G., and Nelson, S. D. (1997). kinetics. Chem. Res. Toxicol. 6, 511–518.
Inactivation of glyceraldehyde-3-phosphate dehydrogenase by a reactive Pineau, T., Fernandez-Salguero, P., Lee, S. S. T., McPhail, T., Ward, J. M., and
metabolite of acetaminophen and mass spectral characterization of an ary- Gonzalez, F. J. (1995). Neonatal lethality associated with respiratory distress
lated active site peptide. Chem. Res. Toxicol. 10, 1097–1103. in mice lacking cytochrome P450 1A2. Proc. Natl. Acad. Sci. USA 92,
Diliberto, J. J., Burgin, D., and Birnbaum, L. S. (1997). Role of CYP1A2 in the 5134 –5138.
sequestration of dioxin: studies using CYP1A2 knockout mice. Biochem. Placke, M. E., Ginsberg, G. L., Wyand, D. S., and Cohen, S. D. (1987).
Biophys. Res. Commun. 236, 431– 433. Ultrastructural changes during acute acetaminophen-induced hepatotoxicity
Fleischer, S., and Kervina, M. (1974). Subcellular fractionation of rat liver. In in the mouse: A time and dose study. Toxicol. Pathol. 15, 431– 438.
Methods in Enzymology, Volume 31, Biomembranes Part A (S. Fleischer Potter, W. Z., Davis, D. C., Mitchell, J. R., Jollow, D. J., Gillette, J. R., and
and L. Packer, Eds.), pp. 6 – 41. Academic Press, New York. Brodie, B. B. (1973). Acetaminophen-induced hepatic necrosis. III. Cyto-
Gupta, S., Rogers, L. K., Taylor, S. K., and Smith, C. V. (1997). Inhibition of chrome P-450-mediated covalent binding in vitro. J. Pharmacol. Exp. Ther.
carbamyl phosphate synthetase-I and glutamine synthetase by hepatotoxic 187, 203–210.
doses of acetaminophen in mice. Toxicol. Appl. Pharmacol. 146, 317–327. Prescott, L. F. (1983). Paracetamol overdosage. Pharmacological consider-
Halmes, N. C., Hinson, J. A., Martin, B. M., and Pumford, N. R. (1996). ations and clinical management. Drugs 25, 290 –314.
Glutamate dehydrogenase covalently binds to a reactive metabolite of acet- Prescott, L. F., Critchley, J. A. J. H., Balali-Mood, M., and Pentland, B. (1981).
aminophen. Chem. Res. Toxicol. 9, 541–546. Effects of microsomal induction on paracetamol metabolism in man. Br. J.
Hinson, J. A., Monks, T. J., Hong, M., Highet, R. J., and Pohl, L. R. (1982). Clin. Pharmac. 12, 149 –153.
108 TONGE ET AL.

Pumford, N. R., Halmes, N. C., Martin, B. M., Cook, R. J., Wagner, C., and tion and quantification of the 3-(cystein-S-yl)-acetaminophen protein ad-
Hinson, J. A. (1997). Covalent binding of acetaminophen to N-10-formyl- ducts in acetaminophen hepatotoxicity. Am. J. Pathol. 138, 359 –371.
tetrahydrofolate dehydrogenase in mice. J. Pharmacol. Exp. Ther. 280, Sarich, T., Kalhorn, T., Magee, S., Al-Sayegh, F., Adams, S., Slattery, J.,
501–505. Goldstein, J., Nelson, S., and Wright, J. (1997). The effect of omeprazole
Pumford, N. R., Martin, B. M., and Hinson, J. A. (1992). A metabolite of pretreatment on acetaminophen metabolism in rapid and slow metabolisers
acetaminophen covalently binds to the 56 kDa selenium binding protein. of S-mephenytoin. Clin. Pharmacol. Ther. 62, 21–28.
Biochem. Biophys. Res. Commun. 182, 1348 –1355. Sinclair, P. R., Gorman, N., Dalton, T., Walton, H. S., Bement, W. J., Sinclair,
Pumford, N. R., Hinson, J. A., Potter, D. W., Rowland, K. L., Benson, R. W., J. F., Smith, A. G., and Nebert, D. W. (1998). Uroporphyria produced in
and Roberts, D. W. (1989). Immunochemical quantitation of 3-(cystein-S- mice by iron and 5-aminolaevulinic acid does not occur in Cyp1a2(2/2)
yl) acetaminophen adducts in serum and liver proteins of acetaminophen null mutant mice. Biochem. J. 330, 149 –153.
treated mice. J. Pharmacol. Exp. Ther. 248, 190 –196. Tonge, R. P., Gartner, C., Chen, W., Bruschi, S., and Nelson, S. D. (1997).
Raucy, J. L., Lasker, J. M., Liebler, C. S., and Black, M. (1989). Acetamin- Immunochemical detection of acetaminophen-protein adducts in human
ophen activation by human liver cytochromes P4502E1 and P4501A2. Arch. liver slices in vitro. Toxicologist 36, 57 (Abstract No. 291).
Biochem. Biophys. 271, 270 –283. Wilson, J. M., Slattery, J. T., Forte, A. J., and Nelson, S. D. (1982). Analysis of
Roberts, D. W., Bucci, T. J., Benson, R. W., Warbritton, A. R., McRae, T. A., acetaminophen metabolites in urine by high performance liquid chromatogra-
Pumford, N. R., and Hinson, J. A. (1991). Immunohistochemical localiza- phy with UV and amperometric detection. J. Chromatogr. 227, 453– 462.

You might also like