ArticlePDF Available

Role of nitric oxide in hepatic microvascular injury elicited by acetaminophen in mice

Authors:

Abstract and Figures

Nitric oxide (NO) is suggested to play a role in liver injury elicited by acetaminophen (APAP). Hepatic microcirculatory dysfunction also is reported to contribute to the development of the injury. As a result, the role of NO in hepatic microcirculatory alterations in response to APAP was examined in mice by in vivo microscopy. A selective inducible NO synthase (iNOS) inhibitor,l-N6-(1-iminoethyl)-lysine (L-NIL), or a nonselective NOS inhibitor, NG-nitro-l-arginine methyl ester (L-NAME), was intraperitoneally administered to animals 10 min before APAP gavage. L-NIL suppressed raised alanine aminotransferase (ALT) values 6 h after APAP, whereas L-NAME increased those 1.7-fold. Increased ALT levels were associated with hepatic expression of iNOS. L-NIL, but not L-NAME, reduced the expression. APAP caused a reduction (20%) in the numbers of perfused sinusoids. L-NIL restored the sinusoidal perfusion, but L-NAME was ineffective. APAP increased the area occupied by infiltrated erythrocytes into the extrasinusoidal space. L-NIL tended to minimize this infiltration, whereas L-NAME further enhanced it. APAP caused an increase (1.5-fold) in Kupffer cell phagocytic activity. This activity in response to APAP was blunted by L-NIL, whereas L-NAME further elevated it. L-NIL suppressed APAP-induced decreases in hepatic glutathione levels. These results suggest that NO derived from iNOS contributes to APAP-induced parenchymal cell injury and hepatic microcirculatory disturbances. L-NIL exerts preventive effects on the liver injury partly by inhibiting APAP bioactivation. In contrast, NO derived from constitutive isoforms of NOS exerts a protective role in liver microcirculation against APAP intoxication and thereby minimizes liver injury.
Content may be subject to copyright.
Role of nitric oxide in hepatic microvascular injury
elicited by acetaminophen in mice
Yoshiya Ito, Edward R. Abril, Nancy W. Bethea, and Robert S. McCuskey
Department of Cell Biology and Anatomy, College of Medicine, University of Arizona, Tucson, Arizona 85724-5044
Submitted 12 May 2003; accepted in final form 9 September 2003
Ito, Yoshiya, Edward R. Abril, Nancy W. Bethea, and Robert S.
McCuskey. Role of nitric oxide in hepatic microvascular injury
elicited by acetaminophen in mice. Am J Physiol Gastrointest Liver
Physiol 286: G60–G67, 2004. First published September 11, 2003;
10.1152/ajpgi.00217.2003.—Nitric oxide (NO) is suggested to play a
role in liver injury elicited by acetaminophen (APAP). Hepatic mi-
crocirculatory dysfunction also is reported to contribute to the devel-
opment of the injury. As a result, the role of NO in hepatic microcir-
culatory alterations in response to APAP was examined in mice by in
vivo microscopy. A selective inducible NO synthase (iNOS) inhibitor,
L-N
6
-(1-iminoethyl)-lysine (L-NIL), or a nonselective NOS inhibitor,
N
G
-nitro-L-arginine methyl ester (L-NAME), was intraperitoneally
administered to animals 10 min before APAP gavage.
L-NIL sup-
pressed raised alanine aminotransferase (ALT) values 6 h after APAP,
whereas
L-NAME increased those 1.7-fold. Increased ALT levels
were associated with hepatic expression of iNOS.
L-NIL, but not
L-NAME, reduced the expression. APAP caused a reduction (20%) in
the numbers of perfused sinusoids.
L-NIL restored the sinusoidal
perfusion, but
L-NAME was ineffective. APAP increased the area
occupied by infiltrated erythrocytes into the extrasinusoidal space.
L-NIL tended to minimize this infiltration, whereas L-NAME further
enhanced it. APAP caused an increase (1.5-fold) in Kupffer cell
phagocytic activity. This activity in response to APAP was blunted by
L-NIL, whereas L-NAME further elevated it. L-NIL suppressed
APAP-induced decreases in hepatic glutathione levels. These results
suggest that NO derived from iNOS contributes to APAP-induced
parenchymal cell injury and hepatic microcirculatory disturbances.
L-NIL exerts preventive effects on the liver injury partly by inhibiting
APAP bioactivation. In contrast, NO derived from constitutive iso-
forms of NOS exerts a protective role in liver microcirculation against
APAP intoxication and thereby minimizes liver injury.
sinusoids; endothelial cell; Kupffer cell; hemorrhage
ACETAMINOPHEN, ALSO KNOWN AS paracetamol (N-acetyl-para-
aminophenol; APAP), is a commonly used over-the-counter
analgesic and antipyretic with few side effects when taken at
therapeutic doses. However, APAP intoxication from overdos-
ing can result in severe hepatic damage, which is characterized
by hemorrhagic centrilobular (CL) necrosis and by towering
the levels of transaminase in both humans and animals (34).
Recently, it has been shown that nitric oxide (NO) is involved
in the progression of APAP-induced liver injury. APAP hep-
atotoxicity is associated with increased expression of inducible
NO synthase (iNOS) (7, 33) and the formation of nitrotyrosine-
protein adducts (12, 19). Inhibition of iNOS with aminogua-
nidine is reported to minimize liver injury elicited by APAP (7,
8). However, others (11) have reported that NOS inhibitors
aggravated the injury.
The APAP-induced hepatic necrosis is preceded by CL
microvascular congestion due to collapse of the sinusoidal wall
and the infiltration of blood elements into the space of Disse
(25, 36). That APAP injures sinusoidal endothelial cells
(SECs) is reflected in the appearance of large gaps in their
cytoplasm (25, 36). These findings suggest that, in addition to
direct hepatocellular damage through metabolic activation of
APAP, hepatic microcirculatory disturbance participates in
liver injury elicited by APAP overdose. We have recently
shown (15) that hepatic microvascular derangement contrib-
utes to the development of APAP hepatotoxicity by using in
vivo microscopy. Initial events occurring in the hepatic micro-
vasculature following APAP included SEC injury exhibited by
the penetration of erythrocytes into the extrasinusoidal space.
NO plays a critical role in maintaining ample blood supply
into the hepatic microvasculature by affecting the expression of
adhesion molecules on leukocytes, platelets, and endothelial
cells (20). The NOS inhibitors exacerbated the hepatic micro-
vascular inflammatory responses including leukocyte-endothe-
lial interaction to endotoxin (30) and ischemia-reperfusion
(13), suggesting that NO plays a protective role in stabilizing
the hepatic microcirculation. However, little is known about
the involvement of NO in hepatic microvascular injury elicited
by APAP. As a result, the present study was conducted to
examine the effects of NOS inhibition on hepatic microvascu-
lar injury after APAP administration by using in vivo micro-
scopic methods (24).
MATERIALS AND METHODS
Experimental animals. Male C57BL/6 mice (7–8 wk of age),
weighing 23–25 g, were purchased from Charles River Laboratories
(Charles River, Windham, ME) and were used for these experiments.
All animals were allowed free access to food and water and were
fasted 24 h before the experiments. The present study was performed
in adherence to the National Institutes of Health guidelines for the use
of experimental animals and followed a protocol approved by the
Animal Care and Use Committee of the University of Arizona. APAP
(300 mg/kg liquid Tylenol; McNeil-PPC, Ft. Washington, PA) was
given to mice by oral gavage. N
G
-nitro-L-arginine methyl ester (L-
NAME; Sigma, St. Louis, MO; 10 mg/kg body wt in 0.1 ml PBS),
which has 30-fold selectivity for constitutive NO synthase (cNOS)
compared with that for iNOS (10), and
L-N
6
-(1-iminoethyl)-lysine
(L-NIL; Cayman Chemical, Ann Arbor, MI; 1, 3, and 10 mg/kg body
wt in 0.1 ml PBS), an inhibitor with 30- to 100-fold selectivity toward
iNOS compared with cNOS (29), were intraperitoneally administered
to animals 10 min before APAP gavage. Animals receiving the same
amount of vehicle (PBS) served as controls. Some of the animals were
treated with
L-NIL (10 mg/kg) 10 min before or simultaneously with
an intraperitoneal injection of APAP (300 mg/kg; Sigma). In some
Address for reprint requests and other correspondence: R. S. McCuskey,
Dept. of Cell Biology and Anatomy, College of Medicine, P.O. Box
245044, Univ. of Arizona, Tucson, AZ 85724-5044 (E-mail:
mccuskey@email.arizona.edu).
The costs of publication of this article were defrayed in part by the payment
of page charges. The article must therefore be hereby marked “advertisement
in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Am J Physiol Gastrointest Liver Physiol 286: G60–G67, 2004.
First published September 11, 2003; 10.1152/ajpgi.00217.2003.
0193-1857/04 $5.00 Copyright
©
2004 the American Physiological Society http://www.ajpgi.orgG60
experiments, the NO donor S-nitro-N-acetylpenicillamine (SNAP;
Cayman) (6) was administered (20 g/kg ip) to mice 20 min before
APAP gavage.
In vivo microscopy. Animals were anesthetized with an intraperi-
toneal injection of 50 mg/kg pentobarbital sodium (Nembutal; Abbott
Laboratories, Abbott Park, IL). The hepatic microvascular responses
were examined by using established high-resolution in vivo micro-
scopic methods (24). Briey, a compound binocular microscope
(Leitz, Wetzlar, Germany) adapted for in vivo microscopy was
equipped to provide either transillumination or epi-illumination as
well as video microscopy by using a charge-coupled device camera
(MTI, Michigan City, IN). The liver was exteriorized through a left
subcostal incision and positioned over a window of optical-grade mica
in a specially designed tray mounted on a microscopic stage. The tray
provided for the drainage of irrigating uids, and the window overlaid
a long-working-distance condenser. The liver was covered by a piece
of Saran Wrap (Dow Chemical, Midland, MI), which held it in
position and limited movement. Homeostasis was ensured by a con-
stant suffusion of the organ with Ringer solution maintained at body
temperature. With the 801.0 numerical aperture water immersion
objective (Leitz) employed for these studies, the methods permitted
visualization of the cells comprising the sinusoidal lining, the formed
elements of the blood, the nuclei, nucleoli, mitochondria and fat
droplets in hepatic cells, and bile canaliculi. Under optimal conditions,
the resolution was 0.30.5 m. Microvascular events were observed
and recorded for at least 30 s for subsequent ofine analysis by using
a Sony Betacam video tape recorder (Sony Medical Electronics, Park
Ridge, NJ).
Kupffer cell (KC) function was assessed by measuring the phago-
cytosis of uorescent 1.0-m latex particles (Fluoresbrite-uorescent
monodispersed polystyrene microspheres; Polysciences, Warrington,
PA) by individual cells. The latex was diluted 1:10 with sterile saline
and injected in 0.1-ml volume through a mesenteric vein by using a
30-gauge lymphangiography needle (Becton Dickinson, Franklin
Lakes, NJ). The distribution and relative number of phagocytic KCs
was measured by counting the number of cells that phagocytosed latex
particles in a standardized microscopic eld 15 min after each mouse
had received the injection of latex. To assess regional distribution, the
number of phagocytic KCs per microscopic eld was counted in 10
periportal (PP) and 10 CL regions in each animal. The relative
adequacy of blood perfusion through the sinusoids was evaluated by
counting the number of sinusoids containing blood ow (SCF) in the
same 10 microscopic elds in which the numbers of phagocytic KCs
were determined. Because reduced perfusion of individual sinusoids
can limit the delivery of the latex particles to KCs in these vessels, the
ratio of KCs that phagocytosed latex particles to SCF was used as an
overall index of KC phagocytic activity.
To examine the interaction of leukocytes with the sinusoidal wall,
quantication of leukocytes adhering to the endothelial lining of
sinusoids was calculated by counting the number of leukocytes per
unit length of sinusoid (adherent leukocytes/100 m) in the same
microscopic elds. A leukocyte was dened as adhering to the
sinusoidal wall if it remained stationary for at least 30 s. Endothelial
swelling, which is thought to be an indication of activation and/or
injury, was assessed by counting the numbers of swollen cells whose
nuclear regions protruded across one-third or more of the lumen in the
same microscopic elds. The results were averaged, and the data were
represented as the average number in each animal.
To quantify the extent of hemorrhage elicited by APAP gavage, the
area occupied by extrasinusoidal erythrocytes was measured in the
same microscopic elds by using a computer-assisted digital imaging
processor (Scion Image; Scion, Frederick, MD). The results were
expressed as extrasinusoidal area occupied by erythrocytes (m
2
/10
CL regions). In addition, the diameters of SCF were measured in the
same microscopic elds from video recordings.
Sampling and assays. In a separate set of experimental animals,
blood was collected from the inferior vena cava and the samples were
separated by centrifugation at 13,000 g for 5 min at 4°C. The samples
were stored at 70°C until assays were performed. The serum
activities of alanine aminotransferase (ALT) were measured by enzy-
matic procedures by using a diagnostic kit (Sigma). In addition, the
serum concentrations of nitrite/nitrate were determined with a kit
(Cayman) by means of Greiss reaction.
Histology and immunohistochemical studies. The hepatic tissues
were immediately xed with 4% paraformaldehyde in 0.1 M phos-
phate buffer solution (pH 7.4). After xation, the tissues were dehy-
drated with a graded series of ethanol solutions and embedded in
parafn. The sections (5 m) from the parafn-embedded tissues
were stained with hematoxylin and eosin. For immunostaining, the
sections were incubated rst with normal goat serum and then with
rabbit anti-murine iNOS antibody (1:100 dilution; Santa Cruz Bio-
technology, Santa Cruz, CA) or with anti-nitrotyrosine antibody
(1:100 dilution; Molecular Probes, Eugene, OR) for 1 h. After being
washed, the sections were incubated with biotinylated anti-rabbit IgG
followed by incubation with avidin-biotin-peroxidase complex (Dako
kit; DakoCytomation, Carpinteria, CA). Reaction products were vi-
sualized by treating sections with 0.02% 3,3-diaminobenzidine and
0.3% nickel ammonium sulfate in 50 mM Tris HCl buffer (pH 7.4).
The sensitivity of the reaction was veried by using isotype-matched
immunoglobulin (rabbit immunoglobulin; Sigma) in the same man-
ner. All steps were performed at room temperature.
RT-PCR analysis. Two and six hours after APAP gavage, 100 mg
of the liver tissue were excised and stored at 20°C until being used.
Total cellular RNA was extracted from the tissue by using a commer-
cially available kit (SV total RNA isolation system; Promega, Mad-
ison, WI) in accordance with the manufacturers instructions. Hepatic
iNOS mRNA was determined by means of semiquantitative RT-PCR
(PCR ELISA kit; Roche Diagnostics, Basel, Switzerland). Amplica-
tion was performed as follows: 94°C, 45 s; 54°C, 45 s; and 72°C, 45 s
for iNOS and -actin, followed by a nal extension for 10 min at 72°C
and by soak at 4°C with the addition of 62.5 M digoxigenin
(DIG)-dUTP for PCR labeling. To avoid tube-to-tube variation, the
primers for -actin and iNOS were added together to each reaction.
The two cDNA were coamplied. One microliter of the PCR products
was denatured and incubated with commercially biotinylated probes
(murine -actin: 5-GGGTGTTGAAGGTCTCAAACATGATCT-
GGG-3; murine iNOS: 5-TCTTGGGTCTCCGCTTCTCGTC-3)
for4hat37°C on a shaker in a streptavidin-coated 96-well plate.
Peroxidase-conjugated anti-DIG antibody was added and was mea-
sured by an ELISA reader. Quantitative determination of mRNA was
calculated based on the slope of the standard curve and was expressed
in attomoles per milliliter.
Measurement of hepatic total glutathione. Hepatic total glutathione
(GSH) was determined colorimetrically by using a commercially
available kit (Oxford Biomedical Research, Oxford, MI). Briey, a
portion of liver tissue was homogenized in 10 ml of cold 5%
metaphosphoric acid and was centrifuged at 3,000 g for 10 min. The
upper aqueous layer was collected and assayed according to the
manufacturers protocol.
Statistical analysis. All data were expressed as means SE.
Multiple comparisons were performed by using one-way ANOVA
with a post hoc Fishers test. Differences were considered to be
signicant at P 0.05.
RESULTS
Effect of NOS inhibitors on the levels of ALT after APAP
gavage. Figure 1 shows the levels of ALT activities after oral
gavage with APAP. The levels of ALT 2 h after APAP gavage
did not change signicantly. Neither L-NIL nor L-NAME
signicantly changed ALT levels in mice treated with APAP.
At 6 h after APAP, the levels of ALT were markedly increased.
L-NIL decreased the ALT levels by 90% in a dose-dependent
manner, whereas L-NAME further elevated them. Both L-NIL
G61NO AND MICROVASCULAR INJURY
AJP-Gastrointest Liver Physiol VOL 286 JANUARY 2004 www.ajpgi.org
and L-NAME by themselves resulted in no signicant changes
in ALT levels (data not shown). On the basis of these results,
we selected a dose of 10 mg/kg L-NIL for the remaining
studies. In addition, the intraperitoneal administration of APAP
caused a signicant increase in ALT levels 6 h after APAP
(1,480 193 IU/l; n 5). Pretreatment with L-NIL (10 mg/kg)
signicantly reduced those (824 107 IU/l; n 5), whereas
cotreatment with L-NIL and APAP did not signicantly change
them (977 109 IU/l; n 5).
Effects of NOS inhibitors on liver microcirculation in re-
sponse to APAP. Figure 2 illustrates the effects of L-NIL and
L-NAME on hepatic microvascular responses to APAP. The
numbers of SCF in CL regions were signicantly decreased 2 h
after APAP when compared with untreated controls. L-NIL, but
not L-NAME, restored sinusoidal perfusion (Fig. 2A). At 6 h
after, the numbers of SCF in PP and CL regions were reduced
by 9 and 21%, respectively. Also, L-NIL, but not L-NAME,
improved the SCF. The oral gavage with APAP caused a
signicant increase in KC phagocytic activity in both PP and
CL regions 2 and 6 h after APAP (Fig. 2B). L-NAME further
raised KC phagocytic activity after APAP, whereas L-NIL
inhibited this activity. APAP failed to induce any changes in
the numbers of leukocytes adhering to the sinusoids in both PP
and CL regions as well as to the central venules 2 and 6 h after
APAP (not shown). The numbers of swollen SECs in PP
regions were signicantly increased (2.8-fold) 2 and 6 h after
APAP. Neither L-NIL nor L-NAME changed the numbers of
SECs in APAP-treated mice. Although there was no evidence
of inltrated erythrocytes into the space of Disse (namely,
hemorrhage) 2 h after APAP gavage, inltration was observed
6 h after APAP (612 362 m
2
/10 CL regions). L-NAME
elicited hemorrhage as early as 2 h after APAP (108 89
m
2
/10 CL regions), although the hemorrhagic area was small.
At 6 h after APAP, the hemorrhagic regions in
L-NAME-
treated mice (2,396 1,173 m
2
/10 CL regions) exceeded
those in vehicle-treated mice. The diameters of PP sinusoids
did not change signicantly in each experimental group (not
shown). In contrast, those of CL sinusoids were signicantly
decreased 2 and 6 h after APAP (6.7 and 12.4%, respectively).
L-NAME did not change the diameters in APAP-treated mice,
whereas L-NIL inhibited the reduced diameters.
Effects of NOS inhibitors on iNOS and nitrotyrosine expres-
sion in the liver treated with APAP. To elucidate the role of NO
in APAP liver injury, we determined the hepatic expression of
iNOS and nitrotyrosine. The expression of hepatic iNOS
mRNA was upregulated 2 h (5.6-fold) and 6 h (9.3-fold) after
APAP, respectively, when compared with controls (Fig. 3).
L-NIL suppressed the increased expression of iNOS mRNA 6 h
after APAP by 56.2%, whereas L-NAME did not change
signicantly.
Fig. 1. Effect of L-N
6
-(1-iminoethyl)-lysine (L-NIL) or N
G
-
nitro-
L-arginine methyl ester (L-NAME) on serum activities of
alanine aminotransferase (ALT) in mice treated with acetamin-
ophen (APAP). The levels of ALT were determined in un-
treated controls and 2 and 6 h after oral gavage with 300 mg/kg
APAP. Mice were pretreated with L-NIL (1, 3, and 10 mg/kg
ip) or L-NAME (10 mg/kg ip) 10 min before APAP. Veh,
vehicle. Data are means SE. *P 0.05 vs. untreated controls
(time 0).
Fig. 2. Effects of L-NIL or L-NAME on the
numbers of sinusoids containing blood ow
(SCF; A) and on Kupffer cell (KC) phago-
cytic activity (B) in mice treated with APAP.
The numbers of SCF and phagocytic KCs
were measured in 10 periportal (closed bars)
and 10 centrilobular (open bars) regions in
each animal. Mice were pretreated with L-
NIL (10 mg/kg ip) or L-NAME (10 mg/kg
ip) 10 min before APAP. Cont, control. Data
are means SE from 6 mice per group.
*P 0.05 vs. untreated controls (time 0).
P 0.05 vs. vehicle-treated mice.
G62 NO AND MICROVASCULAR INJURY
AJP-Gastrointest Liver Physiol VOL 286 JANUARY 2004 www.ajpgi.org
The immunoreactivity with iNOS in the liver was negative
2 h after APAP (data not shown). At 6 h after APAP, the
staining for iNOS was expressed in CL hepatocytes in the same
area where necrosis was exhibited (Fig. 4A). L-NIL and L-
NAME inhibited the expression of iNOS in CL regions (Fig. 4,
B and C). Liver sections stained with hematoxylin and eosin
from mice pretreated with L-NIL showed reduction in hepatic
necrosis after APAP gavage (Fig. 4D).
The staining for nitrotyrosine, a convenient marker of per-
oxynitrite (2), in the liver from controls was negative (Fig. 5A).
Two hours after APAP, the immunoreactivity with nitroty-
rosine was intensely expressed in the CL sinusoids and was
also shown in some individual hepatocytes in midlobular to CL
regions (Fig. 5B). Six hours after APAP, a conuent staining of
PP hepatocytes was seen, although there was no staining of the
sinusoids or hepatocytes in the CL regions (Fig. 5C). Pretreat-
ment with
L-NIL exhibited the positive staining for nitroty-
rosine in individual hepatocytes in PP regions 2 and 6 h after
APAP (Fig. 5D). L-NAME induced strong positive staining for
nitrotyrosine in the CL sinusoids 2 and 6 h after APAP (Fig. 5,
E and F).
Table 1 summarizes the serum concentrations of nitrite/
nitrate at 2 and 6 h after APAP gavage. There were no
signicant differences in the levels of nitrite/nitrate among the
experimental groups.
Effect of L-NIL on total GSH levels in the liver treated with
APAP gavage. The bioactivation of APAP during the initiation
phase of the liver injury causes a signicant decrease in hepatic
GSH levels, which correlate with APAP hepatotoxicity (28).
To address the question of whether L-NIL may affect APAP
bioactivation, we measured hepatic GSH levels. As shown in
Fig. 6, the total GSH levels 1 and 2 h after APAP were
decreased by 59.3 and 70.3%, respectively. L-NIL restored
GSH levels to 75% of control levels at both time points after
APAP.
Effect of NO donors on liver microcirculation after APAP
gavage. To further investigate the role of NO in liver injury in
response to APAP, the NO donor SNAP was administered to
mice treated with APAP. Table 2 summarizes the effects of
pretreatment with SNAP on ALT activity and liver microcir-
culation 6 h after APAP administration. SNAP further in-
creased ALT levels when compared with vehicle-treated mice;
however, no signicant changes in liver microcirculation were
shown between SNAP-treated and vehicle-treated mice.
DISCUSSION
The results of the present study demonstrated that oral
gavage with APAP resulted in liver microcirculatory dysfunc-
tion including impaired sinusoidal perfusion, swelling of SECs,
inltrated erythrocytes in the extrasinusoidal space, and acti-
vated KC phagocytic function (15). The liver microcirculatory
dysfunction elicited by APAP preceded parenchymal cell in-
Fig. 3. Effect of L-NIL or L-NAME on hepatic inducible nitric oxide (NO)
synthase (iNOS) mRNA expression after APAP gavage. The levels of iNOS
mRNA in the liver were determined by RT-PCR analysis. Mice were pre-
treated with L-NIL (10 mg/kg ip) or L-NAME (10 mg/kg ip) 10 min before
APAP. Data are means SE from 3 animals per group. #P 0.05 vs.
untreated controls (time 0). *P 0.05 for vehicle vs. L-NIL at 6 h.
Fig. 4. Photographs of the liver sections stained with iNOS
(A, B, and C) or hematoxylin and eosin (D) 6 h after APAP
in combination with vehicle (A), L-NIL (B and D), or
L-NAME (C). A: immunoreactivity with iNOS was shown
in centrilobular hepatocytes treated with APAP. B: staining
for iNOS was negative in the centrilobular hepatocytes
pretreated with L-NIL. C: L-NAME reduced the expression
of iNOS in centrilobular hepatocytes. D: hematoxylin and
eosin-stained liver sections from mice pretreated with L-
NIL showed minimal changes. PV, portal vein; CV, central
vein. Original magnication, 200.
G63NO AND MICROVASCULAR INJURY
AJP-Gastrointest Liver Physiol VOL 286 JANUARY 2004 www.ajpgi.org
jury as indicated by a signicant increase in ALT values and
hepatic necrosis. L-NIL, an inhibitor of iNOS, attenuated
APAP-induced hepatocellular injury as well as hepatic micro-
circulatory dysfunction. In contrast, L-NAME, a nonselective
NOS inhibitor that favors cNOS over iNOS, aggravated the
injury. In particular, L-NAME exacerbated the extrasinusoidal
area occupied by erythrocytes and KC phagocytic function in
response to APAP.
NO has been implicated in the progression of APAP hepa-
totoxicity. APAP-induced liver injury is associated with in-
creased iNOS protein expression (7, 8, 33) as well as increased
iNOS mRNA expression (14), which is consistent with our
present results. The immunohistochemical studies showed that
iNOS was expressed in the damaged hepatocytes in the CL
regions, suggesting that these hepatocytes are a source of iNOS
(7). Increased NO production from isolated hepatocytes also
has been reported (7). Furthermore, mice decient in iNOS
moderately reduce release of hepatic enzyme after APAP
treatment (8). However, Michael et al. (26) reported that there
is no difference in the amount of hepatic necrosis between
iNOS knockout mice and wild-type mice, and Bourdi et al. (3)
reported that both iNOS knockout and wild-type mice exhib-
ited the same raised ALT levels after APAP (150 mg/kg). The
discrepancy within iNOS knockout mice still remains unclear.
Pretreatment with aminoguanidine, a selective iNOS inhibitor,
minimizes liver injury elicited by APAP (7, 8). However,
others (11) have shown that the simultaneous administration of
aminoguanidine with APAP increased the initial rate of devel-
opment of hepatotoxicity as indicated by ALT values (within
4 h after APAP), but there was no signicant difference in ALT
levels 6 and 8 h after APAP. The current study also showed
that another selective iNOS inhibitor, L-NIL, attenuated APAP-
induced liver injury, which was associated with reduced ex-
pression of iNOS in the liver treated with APAP. These
ndings suggest that NO derived from iNOS is involved in the
development of APAP hepatotoxicity. In contrast, Hinson et al.
(11) reported that the simultaneous administration of L-NIL (3
mg/kg ip) with APAP (300 mg/kg ip) approximately doubled
ALT levels 4 h after APAP treatment in B6C3F1 mice. There-
fore, we also treated mice with L-NIL (10 mg/kg ip) 10 min
Fig. 5. Photographs of the liver sections for nitrotyrosine protein adducts after APAP in combination with vehicle, L-NIL, or
L-NAME. A: untreated controls showed no staining for nitrotyrosine in the liver. B: strong positive staining was seen in the
centrilobular sinusoids and centrilobular to midlobular hepatocytes 2 h after APAP. C: 6 h after APAP, a conuent staining for
nitrotyrosine was observed in the periportal hepatocytes. D: immunoreactivity with nitrotyrosine was expressed in the periportal
hepatocytes pretreated with L-NIL at 6 h after APAP. E: L-NAME exhibited intense expression of nitrotyrosine in the centrilobular
sinusoids 2 h after APAP. F: at 6 h after APAP, pretreatment with L-NAME caused intense staining for nitrotyrosine in the
centrilobular sinusoids. Original magnication, 200 for A, D, and E and 400 for B, C, and F.
Table 1. Effect of L-NIL or L-NAME on the serum concentration of nitrite/nitrate in mice treated with APAP
Control
Time After APAP Gavage
2h 6h
Vehicle
L-NIL L-NAME Vehicle L-NIL L-NAME
73.17.2 86.77.6 63.45.4 75.424.1 104.95.8 79.74.8 89.321.5
Data are means SE (M) from 36 mice. There were no signicant differences in nitric oxide (NO) concentrations among the treatment groups.
L-N
6
-(1-iminoethyl)-lysine (L-NIL) (10 mg/kg, ip) or N
G
-nitro-L-arginine methy ester (L-NAME) (10 mg/kg, ip) was administered 10 min before acetaminophen
(APAP) gavage.
G64 NO AND MICROVASCULAR INJURY
AJP-Gastrointest Liver Physiol VOL 286 JANUARY 2004 www.ajpgi.org
before APAP (300 mg/kg ip) or with L-NIL in combination
with APAP. As shown in RESULTS, the levels of ALT 6 h after
APAP were increased, and pretreatment with L-NIL slightly
reduced those, whereas cotreatment with L-NIL and APAP did
not signicantly change them. Therefore, the discrepancy may
be due to the differences in the animal species, doses of
inhibitors, timing of their administration, or route of APAP
administration being used.
Since it has been suggested that L-NIL inhibits iNOS by
reacting with the protein at the enzyme active site, L-NIL does
not appear to suppress the expression of iNOS mRNA. On the
other hand, we demonstrated that L-NIL attenuated the expres-
sion of iNOS mRNA and iNOS protein in the liver. Although
the mechanisms by which L-NIL prevents iNOS expression in
this model remain unknown, Kang et al. (16) reported that, in
mouse peritoneal macrophages, L-NIL inhibited lipopolysac-
charide-induced activation of NF-B, which regulates iNOS
expression (37). Thus it may be plausible that L-NIL sup-
pressed iNOS mRNA expression in the liver treated with
APAP by inactivation of NF-B.
The bioactivation of APAP results in the formation of the
reactive metabolite, presumably N-acetyl-p-benzoquinone-
imine, which depletes intracellular sources of GSH (28). In the
current study, L-NIL suppressed the initial drop in hepatic total
GSH levels following APAP gavage, suggesting that the in-
hibitory effect of L-NIL on liver injury is partially mediated
through a decrease in bioactivation of APAP. The possibility
that L-NIL prevents APAP metabolic activation also suggests
that L-NIL could inhibit covalent binding to proteins of APAP.
Further studies will be required to elucidate the effect of L-NIL
on APAP covalent binding. The other possibility that L-NIL
maintained GSH levels is due to the reduced NO production in
the liver because reactive nitrogen species are scavenged by
GSH (18). Therefore, the mechanisms involved in the protec-
tive action of L-NIL against APAP-induced liver injury appear
to be multifactorial in this model.
The exogenous administration of NO donor (SNAP) exac-
erbated ALT release but did not affect the hepatic microcircu-
latory dysfunction following APAP gavage. These results sug-
gest that excess NO in the liver treated with APAP causes liver
injury and that NO may act directly on hepatocytes. Recently,
Liu et al. (22) reported that continuous administration of
O
2
-vinyl-1-(pyrrolidin-1-yl)diazen-1-ium-1,2-diolate (V-PYRRO/
NO), a liver-selective NO donor, protected the liver against
APAP hepatotoxicity. The differences may be caused by dif-
ferent delivery of NO donor to the liver; V-PYRRO is me-
tabolized to NO preferentially in hepatocytes. Moreover,
excessive extracellular NO elicited by a bolus injection of
SNAP could mediate hepatocellular mitochondrial dysfunction
through its actions on an enzyme such as cytochrome c oxi-
dase, an enzyme constituting the distal end of the mitochon-
drial respiratory chain (4). The mitochondrial dysfunction
caused by SNAP could be amplied in addition to that by
APAP (14). In a preliminary study, we administered 10 mg/kg
of V-PYRRO/NO (Cayman) subcutaneously to mice gavaged
with APAP according to the experimental protocols by Liu et
al. (23). Since V-PYRRO/NO was supplied as a solution in
absolute ethanol, some animals received ethanol as vehicle.
Our preliminary study revealed that not only V-PYRRO but
also absolute ethanol as vehicle reduced ALT levels 6 h after
APAP gavage. Since a serum level of 5 mM ethanol has been
found to protect animals from APAP hepatotoxicity (35), the
effect of vehicle also could contribute to protective effect of
V-PYRRO/NO on APAP-induced liver injury, though we are
uncertain whether Liu et al. have used ethanol as a vehicle for
V-PYRRO/NO.
The toxicological effect of NO seems to occur through
peroxynitrite, a potent oxidant formed by reaction of NO with
superoxide. The nding of nitrotyrosine staining shown in the
hepatic CL sinusoids 2 h after APAP gavage, which is consis-
tent with those by others (12, 19), suggested that peroxynitrite
was formed early by the sinusoidal lining cells including KCs
and SECs. Thus it may be plausible that peroxynitrite injures
SECs in an early phase of APAP hepatotoxicity, resulting in
hemorrhage through the formation of gaps in their cytoplasm
(25, 36).
L-NIL diminished the initial nitrotyrosine formation in
the sinusoids, indicating that NO derived from iNOS in the
sinusoidal lining cells is involved in the formation of peroxyni-
Table 2. Effects of SNAP on serum ALT activity and on liver
microcirculation 6 h after APAP gavage
APAP Gavage
Vehicle, n4 SNAP, n3
ALT, IU/l 2,405422 4,609632*
No. of perfused sinusoids/eld
Periportal 4.60.2 4.90.2
Centrilobular 3.80.3 3.90.3
No. of adherent leukocytes/100 m
Periportal 0.040.03 0.040.03
Centrilobular 0.040.04 0.000.00
No. of swollen SECs/eld
Periportal 0.20.1 0.30.0
Centrilobular 0.20.1 0.30.0
Kupffer cell phagocytic activity
Periportal 1.00.1 0.50.1
Centrilobular 1.10.1 0.80.2
Area occupied by inltrated erythrocytes,
m
2
/10 centrilobular regions
247144 00
Data are means SE. S-nitro-N-acetylpenicillamine (SNAP; 20 g/kg, ip)
or vehicle was administered 20 min before APAP (300 mg/kg) gavage. ALT,
alanine aminotransferase; SEC, sinusoidal endothelial cells. *P 0.05 vs.
vehicle-treated mice.
Fig. 6. Effect of L-NIL on hepatic total glutathione (GSH) levels after APAP
gavage. The levels of hepatic GSH were determined in untreated controls and
1 and 2 h after oral gavage with 300 mg/kg APAP. Mice were pretreated with
L-NIL (10 mg/kg ip) 10 min before APAP. Data are means SE. #P 0.05
vs. untreated controls (time 0). *P 0.05 for vehicle vs. L-NIL.
G65NO AND MICROVASCULAR INJURY
AJP-Gastrointest Liver Physiol VOL 286 JANUARY 2004 www.ajpgi.org
trite. This nding is in agreement with that of Hinson et al. (11)
showing that aminoguanidine attenuated APAP-induced nitra-
tion of tyrosine in liver tissue. In considering the results of
microvascular inammatory responses to APAP, KCs and
SECs appear to be sources of NO. However, iNOS protein in
the liver 2 h after APAP was not expressed in the sinusoidal
lining cells, suggesting that iNOS expression was not enough
to be detected by means of immunohistochemistry; even
mRNA for iNOS was marginally transcripted. Pretreatment
with L-NIL also minimized APAP-induced hepatic microcir-
culatory dysfunction, presumably by inhibiting KC function.
Because inactivation of KCs with gadolinium chloride restored
liver microcirculation in response to APAP (15), the inhibitory
effect of L-NIL on KC activity contributes to preserving he-
patic microcirculation, resulting in reduction of liver injury
(21, 27). However, the mechanisms by which L-NIL suppresses
KC function or whether KCs release NO in the early phase of
APAP hepatotoxicity remain unknown.
At later time point (6 h after APAP gavage), nitrotyrosine
staining was negative in the CL regions of the liver, whereas
iNOS staining was positive in the same regions. On the other
hand, nitrotyrosine staining was positive in the PP hepatocytes.
The results indicate that the location for nitrotyrosine staining
in the liver was shifted from the CL regions to the PP regions
with time after APAP treatment. The reduced intensity of
nitrotyrosine staining in CL hepatocytes 6 h after APAP may
have resulted from inability to nitrate protein because of
cellular lysis (31). On the other hand, the staining for nitroty-
rosine was observed in the PP hepatocytes with no evidence of
necrosis. These results are inconsistent with those reported by
others, who demonstrated nitrotyrosine expression in the CL
hepatocytes (19), but not in the PP ones (12), at 6 h after
APAP. The reasons for these differences remain unclear, al-
though it may be due to differences in animal species, response
to APAP, and/or differences in immunohistochemical proto-
cols. Our ndings suggest that nitrotyrosine formation may
serve as a biomarker for the effects of reactive nitrogen species
and may not be relevant to the injury mechanism, because no
evidence of necrosis was noticed in the PP hepatocytes. In
addition, it may be speculated that peroxynitrite was reacted
with hepatic glutathione (endogenous scavengers of peroxyni-
trite), which are still reserved in the PP hepatocytes and
thereby protected against peroxynitrite-mediated oxidant stress
injury in these hepatocytes.
In contrast to L-NIL, L-NAME, a nonselective NOS inhibi-
tor, exacerbated APAP-induced liver injury. These ndings are
consistent with the ndings by others (11) showing that N-
monomethyl-L-arginine worsened the injury. The enhanced
liver injury was associated with aggravated APAP-induced
hepatic microvascular injury including KC phagocytic activity.
L-NAME caused upregulated expression of nitrotyrosine in the
hepatic CL sinusoids 2 and 6 h after APAP gavage, indicating
that peroxynitrite could lead to SEC injury. This possibility
was supported by the results of the present study showing that
the extrasinusoidal area occupied by inltrated erythrocytes
was further increased by L-NAME. These results suggest that
NO-mediated nitration of cellular proteins in SECs at least in
part contributes to the progression of liver injury elicited by
APAP and that peroxynitrite is an important mediator of injury
to parenchymal cells (18) as well as SECs. However, as
mentioned above, whether the presence of nitrotyrosine-pro-
tein adducts is related to the injury remains to be elucidated.
The enhanced hemorrhagic necrosis by L-NAME also suggests
that endogenous NO derived from cNOS plays a critical role in
maintaining the functional integrity of SECs, because cNOS
appears to be localized exclusively to the SECs (32). cNOS has
been shown to associate with cytoskeletal proteins (1). Thus
inhibition of cNOS with L-NAME could result in disruption of
cytoskeleton, leading to changes in cell shape or to the forma-
tion of gaps. In addition, it has been reported that APAP
increased levels of hepatic lipid peroxidation in mice treated
with the iNOS inhibitor aminoguanidine (11) or in mice de-
cient in iNOS (26). This suggests that exacerbated liver injury
elicited by APAP is mediated by lipid peroxidation during
NOS inhibition. However, Knight et al. (17) recently have
shown that there is no evidence of enhanced lipid peroxidation
in the liver treated with APAP in combination with aminogua-
nidine, indicating that lipid peroxidation is not involved in the
injury mechanism. Because of the antioxidant properties of
cNOS-derived NO, inhibition of cNOS could increase super-
oxide (9). Thus it is plausible that activated KCs generated
larger amounts of superoxide and thereby formed peroxynitrite
in the sinusoids. However, the mechanisms by which NO from
cNOS suppresses the activation of KCs remain unknown.
Recent studies reported that nonselective NOS inhibitors
reduced the sinusoidal perfusion and sinusoidal diameters dur-
ing endotoxemia (5, 30), indicating that cNOS inhibition
caused vasoconstriction and thereby augmented liver injury.
However, this is not seen in the current study, in which
L-NAME did not further reduce the sinusoidal blood ow as
well as sinusoidal diameter following APAP treatment. Thus
aggravation of APAP-induced liver injury by L-NAME is not
attributed to vasoconstriction.
In summary, selective inhibition of iNOS with L-NIL atten-
uated liver microcirculatory dysfunction elicited by APAP
gavage. In contrast, inhibition of cNOS by L-NAME potenti-
ated APAP-induced liver injury due to its effect on the liver
microcirculation including KC function. These results suggest
that NO derived from iNOS contributes to APAP-induced
parenchymal cell injury and hepatic microcirculatory distur-
bances, whereas NO-derived cNOS exerts a protective role in
liver microcirculation against APAP intoxication and prevents
enhanced liver injury.
GRANTS
This study was supported in part by National Institute on Alcohol Abuse
and Alcoholism Grant RO1-AA-12436.
REFERENCES
1. Ayajiki K, Kindermann M, Hecker M, Fleming I, and Busse R.
Intracellular pH and tyrosine phosphorylation but not calcium determine
shear stress-induced nitric oxide production in native endothelial cells.
Circ Res 78: 750758, 1995.
2. Beckman JS, Beckman TW, Chen J, Marshall PA, and Freeman BA.
Apparent hydroxyl radical production by peroxynitrite: implications for
endothelial injury from nitric oxide and superoxide. Proc Natl Acad Sci
USA 87: 16201624, 1990.
3. Bourdi M, Masubuchi Y, Reilly TP, Amouzadeh HR, Martin JL,
George JW, Shah AG, and Pohl LR. Protection against acetaminophen-
induced liver injury and lethality by interleukin 10: role of inducible nitric
oxide synthase. Hepatology 35: 289298, 2002.
4. Cleeter MWJ, Cooper JM, Darkey-Usmar VM, Moncada S, and
Schapira AHV. Reversible inhibition of cytochrome c oxidase, the
terminal enzyme of the mitochondrial respiratory chain, by nitric oxide.
FEBS Lett 345: 5054, 1994.
G66 NO AND MICROVASCULAR INJURY
AJP-Gastrointest Liver Physiol VOL 286 JANUARY 2004 www.ajpgi.org
5. Corso CO, Gundersen Y, Dorger M, Lilleaasen P, Aasen AO, and
Messmer K. Effects of the nitric oxide synthase inhibitors N(G)-nitro-
L-
arginine methyl ester and aminoethyl-isothiourea on the liver microcircu-
lation in rat endotoxemia. J Hepatol 28: 6169, 1998.
6. Ganthier TW, Davenpeck KL, and Lefer AM. Nitric oxide attenuates
leukocyte-endothelial interaction via P-selectin in splanchnic ischemia-
reperfusion. Am J Physiol Gastrointest Liver Physiol 267: G562G568,
1994.
7. Gardner CR, Heck DE, Yang CS, Thomas PE, Zhang XJ, DeGeorge
GL, Laskin JD, and Laskin DL. Role of nitric oxide in acetaminophen-
induced hepatotoxicity in the rat. Hepatology 27: 748754, 1998.
8. Gardner CR, Laskin JD, Dambach DM, Sacco M, Durham SK, Bruno
MK, Cohen SD, Gordon MK, Gerecke DR, Zhou P, and Laskin DL.
Reduced hepatotoxicity of acetaminophen in mice lacking inducible nitric
oxide synthase: potential role of tumor necrosis factor- and interleukin-
10. Toxicol Appl Pharmacol 184: 2736, 2002.
9. Grisham MB, Jourd’Heuil D, and Wink DA. Nitric oxide. I. Physio-
logical chemistry of nitric oxide and its metabolites: implications in
inammation. Am J Physiol Gastrointest Liver Physiol 276: G315G321,
1999.
10. Gross SS, Stuehr DJ, Aisaka K, Jaffe EA, Levi R, and Griffith OW.
Macrophage and endothelial cell nitric oxide synthesis: cell-type selective
inhibition by NG-aminoarginine, NG-nitroarginine and NG-methylargin-
ine. Biochem Biophys Res Commun 170: 96103, 1990.
11. Hinson JA, Bucci TJ, Irwin LK, Michael SL, and Mayeux PR. Effect
of inhibitors of nitric oxide synthase on acetaminophen-induced hepato-
toxicity in mice. Nitric Oxide 6: 160167, 2002.
12. Hinson JA, Pike SL, Pumford NR, and Mayeux PR. Nitrotyrosine-
protein adducts in hepatic centrilobular areas following toxic doses of
acetaminophen in mice. Chem Res Toxicol 11: 604607, 1998.
13. Horie Y, Wolf R, and Granger DN. Role of nitric oxide in gut
ischemia-reperfusion-induced hepatic microvascular dysfunction. Am J
Physiol Gastrointest Liver Physiol 273: G1007G1013, 1997.
14. Ishida Y, Kondo T, Ohshima T, Fujiwara H, Iwakura Y, and Mu-
kaida N. A pivotal involvement of IFN- in the pathogenesis of acet-
aminophen-induced acute liver injury. FASEB J 16: 12271236, 2002.
15. Ito Y, Bethea NW, Abril ER, and McCuskey RS. Early hepatic
microvascular injury in response to acetaminophen toxicity. Microcircu-
lation 10: 391400, 2003.
16. Kang JL, Lee K, and Castranova V. Nitric oxide up-regulates DNA-
binding activity of nuclear factor-B in macrophages stimulated with
silica and inammatory stimulants. Mol Cell Biochem 215: 19, 2000.
17. Knight TR, Fariss MW, Farhood A, and Jaeschke H. Role of lipid
peroxidation as mechanism of liver injury after acetaminophen overdose in
mice. Toxicol Sci. In press.
18. Knight TR, Ho YS, Farhood A, and Jaeschke H. Peroxynitrite is a
critical mediator of acetaminophen hepatotoxicity in murine livers: pro-
tection by glutathione. J Pharmacol Exp Ther 303: 468475, 2002.
19. Knight TR, Kurtz A, Bajt ML, Hinson JA, and Jaeschke H. Vascular
and hepatocellular peroxynitrite formation during acetaminophen toxicity:
role of mitochondrial oxidant stress. Toxicol Sci 62: 212220, 2001.
20. Kubes P, Suzuki M, and Granger DN. Nitric oxide: an endogenous
modulator of leukocyte adhesion. Proc Natl Acad Sci USA 88: 46514655,
1991.
21. Laskin DL, Gardner CR, Price VF, and Jollow DJ. Modulation of
macrophages functioning abrogates the acute hepatotoxicity of acetamin-
ophen. Hepatology 21: 10451050, 1995.
22. Liu J, Li C, Waalkes MP, Clark J, Myers P, Saavedra JE, and Keefer
LK. The nitric oxide donor, V-PYRRO/NO, protects against acetamino-
phen-induced hepatotoxicity in mice. Hepatology 37: 324333, 2003.
23. Liu J, Saavedra JE, Lu T, Song JG, Clark J, Waalkes MP, and Keefer
LK. O
2
-vinyl 1-(pyrrolidin-1-yl)diazen-1-ium-1,2-diolate protection
against
D-galactosamine/endotoxin-induced hepatotoxicity in mice:
genomic analysis using microarrays. J Pharmacol Exp Ther 300: 1825,
2002.
24. McCuskey RS. Microscopic methods for studying the microvasculature
of internal organs. In: Physical Techniques in Biology and Medicine
Microvascular Technology, edited by Baker CH and Nastuk WF. New
York: Academic, 1986, p. 247264.
25. McCuskey RS, Machen NW, Wang X, McCuskey MK, Abril ER,
Earnest DL, and Deleve LD. A single ethanol binge exacerbates early
acetaminophen-induced centrilobular injury to the sinusoidal endothelium
and alters sinusoidal blood ow. In: Cells of the Hepatic Sinusoid VIII,
edited by Wisse E, Knook DL, and Arthur M. Leiden: Kupffer Cell
Foundation, 2001, p. 6870.
26. Michael SL, Mayeux PR, Bucci TJ, Warbritton AR, Irwin LK,
Pumford NR, and Hinson JA. Acetaminophen-induced hepatotoxicity in
mice lacking inducible nitric oxide synthase activity. Nitric Oxide 5:
432441, 2001.
27. Michael SL, Pumford NR, Mayeux PR, Niesman MR, and Hinson JA.
Pretreatment of mice with macrophage inactivators decreases acetamino-
phen hepatotoxicity and the formation of reactive oxygen and nitrogen
species. Hepatology 30: 186195, 1999.
28. Mitchell J, Jollow D, Potter W, Gillette J, and Brodie B. Acetamino-
phen-induced hepatic necrosis. IV. Protective role of glutathione. J Phar-
macol Exp Ther 187: 211217, 1973.
29. Moore WM, Webber RK, Jerome GM, Tjoeng FS, Misko TP, and
Currie MG. L-N
6
-(1-iminoethyl)-lysine: a selective inhibitor of inducible
nitric oxide synthase. J Med Chem 37: 38863888, 1994.
30. Nishida J, McCuskey RS, McDonnell D, and Fox EB. Protective role of
NO in hepatic microcirculatory dysfunction during endotoxemia. Am J
Physiol Gastrointest Liver Physiol 267: G1135G1141, 1994.
31. Roberts DW, Bucci TJ, Benson RW, Warbritton AR, McRae TA,
Pumford NR, and Hinson JA. Immunohistochemical localization and
quantication of the 3-(cystein-S-yl)-acetaminophen protein adduct in
acetaminophen hepatotoxicity. Am J Pathol 138: 359371, 1991.
32. Shah V, Haddad FG, Garcia-Cardena G, Frangos JA, Mennone A,
Groszmann RJ, and Sessa WC. Liver sinusoidal endothelial cells are
responsible for nitric oxide modulation of resistance in the hepatic sinu-
soids. J Clin Invest 100: 29232930, 1997.
33. Storto M, Ngomba RT, Battaglia G, Freitas I, Griffini P, Richelmi P,
Nicoletti F, and Vairetti M. Selective blockade of mGlu5 metabotropic
glutamine receptors is protective against acetaminophen hepatotoxicity in
mice. J Hepatol 38: 179187, 2003.
34. Thomas SH. Paracetamol (acetaminophen) poisoning. Phamacol Ther 60:
91120, 1993.
35. Thummel KE, Slattery JT, and Nelson SD. Mechanisms by which
ethanol diminishes the hepatotoxicity of acetaminophen. J Pharmacol Exp
Ther 245: 129136, 1987.
36. Walker RM, Racz WJ, and McElligott TF. Scanning electron micro-
scopic examination of acetaminophen-induced hepatotoxicity and conges-
tion in mice. Am J Pathol 113: 321330, 1983.
37. Zingarelli B, Hake PW, Yang Z, O’Connor M, Denenberg A, and
Wong HR. Absence of inducible nitric oxide synthase modulates early
reperfusion-induced NF-kappaB and AP-1 activation and enhances myo-
cardial damage. FASEB J 16: 327342, 2002.
G67NO AND MICROVASCULAR INJURY
AJP-Gastrointest Liver Physiol VOL 286 JANUARY 2004 www.ajpgi.org
... After liver contusion, some authors have reported an enhancement of MMP-2 at 6 h, with a peak at 24 h, then a gradual reduction with normalization of the levels after 7 days (71). ECM degradation by MMP is added to the hepatic microcirculation disturbances caused by NOx excess (72). Intracellular damage induced by APAP overdose is amplified by increased MMP and elicits a robust inflammatory response with the release of pro-inflammatory cytokines that will attract more neutrophils at the injury site (73). ...
Article
Full-text available
Background/aim: The hepatoprotective role of various molecules in drug-induced hepatotoxicity arouses great interest. We investigated the effect of liposomal curcumin (LCC) on experimental acetaminophen (APAP)-induced hepatotoxicity. Materials and methods: Rats were randomly allocated into 5 groups, and the effect of two LCC concentrations was studied: group 1 - 1 ml intraperitoneal (i.p.) saline, group 2 - APAP pretreatment, group 3 - APAP+silymarin (extract of the silybum marianum with anti-inflammatory, anti-oxidant, and anti-fibrotic properties), group 4 - APAP+LCC1, group 5 - APAP+LCC2. The biomarkers of oxidative stress (nitric oxide and malondialdehyde) and antioxidant status of plasma (thiols and catalase), TNF-α, MMP-2 and MMP-9 serum levels were evaluated. Results: An improvement in oxidative stress, antioxidant status, and TNF-α, MMP-2 and MMP-9 levels was obtained in groups pretreated with LCC compared to silymarin treatment, in a dose-dependent manner. Histopathological examination reinforced the results. Conclusion: Liposomal curcumin improves the oxidative stress/antioxidant balance and alleviates inflammation in experimental APAP-induced hepatotoxicity.
... 17 Such toxicity is commonly observed following acetaminophen overdosing in adults and involves the liver, kidneys, and auditory cells. [18][19][20] Acetaminophen-related tissue toxicity can also occur in neonates, and in a recently reported case, it was associated with liver tissue injury. 21 Acetaminophen-induced vascular tissue injury has not been previously reported. ...
Article
Background: Acetaminophen is widely prescribed to both neonates and young children for a variety of reasons. In adults, therapeutic usage of acetaminophen induces systemic arterial pressure changes and exposure to high doses promotes tissue toxicity. The pulmonary vascular effects of acetaminophen at any age are unknown. Hypothesizing that, early in life, it promotes vasomotor tone changes via oxidative stress, we tested the in vitro acetaminophen effects on intrapulmonary and carotid arteries from newborn and adult rats. Methodology: We measured the acetaminophen dose-response in isometrically mounted arteries and pharmacologically evaluated the factors accounting for its vasomotor effects. Results: Acetaminophen induced concentration- and age-dependent vasomotor tone changes. Whereas a progressive increase in vasomotor tone was observed in the newborn, the adult arteries showed mostly vasorelaxation. Inhibition of endogenous nitric oxide generation with L-NAME and the use of the peroxynitrite decomposition catalyst FeTPPS (Fe(III)5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrinato chloride) mostly abolished the drug-induced increase in newborn pulmonary vasomotor tone CONCLUSIONS: In newborn rats, acetaminophen increases pulmonary vasomotor tone via peroxynitrite generation. Given its therapeutic usage, further clinical studies are warranted to assess the acetaminophen effects on the newborn pulmonary and systemic vascular resistance.
... This is all the more relevant considering the role of nitric oxide in the process. While studies have been conducted on the role of NO in mediating APAP induced SEC damage (Ito et al., 2004), they lacked a mitochondrial focus. This is relevant since nitric oxide would have to be produced close to the location where superoxide is being produced for efficient reaction to peroxynitrite. ...
Chapter
Acetaminophen (APAP) is a highly effective analgesic, which is safe at therapeutic doses. However, an overdose can cause hepatotoxicity and even liver failure. APAP toxicity is currently the most common cause of acute liver failure in the United States. Decades of research on mechanisms of liver injury have established the role of mitochondria as central players in APAP-induced hepatocyte necrosis and this chapter examines the various facets of the organelle's involvement in the process of injury as well as in resolution of damage. The injury process is initiated by formation of a reactive metabolite, which binds to sulfhydryl groups of cellular proteins including mitochondrial proteins. This inhibits the electron transport chain and leads to formation of reactive oxygen species, which induce the activation of redox-sensitive members of the MAP kinase family ultimately causing activation of c-Jun N terminal kinase, JNK. Translocation of JNK to the mitochondria then amplifies mitochondrial dysfunction, ultimately resulting in mitochondrial permeability transition and release of mitochondrial intermembrane proteins, which trigger nuclear DNA fragmentation. Together, these events result in hepatocyte necrosis, while adaptive mechanisms such as mitophagy remove damaged mitochondria and minimize the extent of the injury. This oscillation between recovery and necrosis is predominant in cells at the edge of the necrotic area in the liver, where induction of mitochondrial biogenesis is important for liver regeneration. All these aspects of mitochondria in APAP hepatotoxicity, as well as their relevance to humans with APAP overdose and development of therapeutic approaches will be examined in detail in this chapter.
... Previous studies demonstrated that acetaminophen (APAP, or Paracetamol) acutely alters microvascular flow in mice liver, quantified as the fraction of perfused sinusoids per microscope field (Ito et al., 2003;Ito et al., 2004). APAP hepatotoxicity occurs primarily in centrilobular liver regions suggesting that effects of APAP on microcirculatory flow might likewise manifest in the regions immediately surrounding the central veins. ...
... Also, Jiang et al [45] and Vinas et al [46] found that the upregulation of iNOS and or NO synthesis through the iNOS pathway induced apoptosis during in vitro and in vivo ischemia-reperfusion studies, respectively. In addition, genetic manipulation that inhibits the production of iNOS [11] and treatment with specific iNOS inhibitors [47,48] during IR results in decreased injury and improved recovery. However, there were controversial studies supporting the benefit of iNOS [49, 50, and 51]. ...
Article
Full-text available
Background: Adropin is a peptide hormone that plays an important role in energy homeostasis and endothelial functions. Nitric oxide is a potent vasodilator synthesized in the vascular endothelium from the nitric oxide synthase enzyme (NOS). There are controversial studies about the role of NOS and its isoforms the endothelial (eNOS) and the inducible (iNOS) forms in ischemia-reperfusion (IR) injury. It was found that adropin influences gene expression of eNOS and enhances endothelial cell function. However, few researchers have studied the link between adropin and iNOS activity during different phases of IR-injury.
... Notably, this APAP toxicity is associated with a cytokine response that includes upregulation of IL-1β and TNFα (23,39). Previous reports demonstrated that, during murine APAP intoxication, iNOS protein is well detectable (33,40) in hepatocytes at regions with centrilobular injury (41,42). Evaluation of iNOS knockout mice indicated that iNOS-derived NO may promote injury during early intoxication (detected by serum ALT) (43,44). ...
Article
Full-text available
Cytokine regulation of high-output nitric oxide (NO) derived from inducible NO synthase (iNOS) is critically involved in inflammation biology and host defense. Herein, we set out to characterize the role of type I interferon (IFN) as potential regulator of hepatic iNOS in vitro and in vivo. In this regard, we identified in murine Hepa1-6 hepatoma cells a potent synergism between pro-inflammatory interleukin-β/tumor necrosis factor-α and immunoregulatory IFNβ as detected by analysis of iNOS expression and nitrite release. Upregulation of iNOS by IFNβ coincided with enhanced binding of signal transducer and activator of transcription-1 to a regulatory region at the murine iNOS promoter known to support target gene expression in response to this signaling pathway. Synergistic iNOS induction under the influence of IFNβ was confirmed in alternate murine Hepa56.1D hepatoma cells and primary hepatocytes. To assess iNOS regulation by type I IFN in vivo, murine acetaminophen (APAP)-induced sterile liver inflammation was investigated. In this model of acute liver injury, excessive necroinflammation drives iNOS expression in diverse liver cell types, among others hepatocytes. Herein, we demonstrate impaired iNOS expression in type I IFN receptor-deficient mice which associated with diminished APAP-induced liver damage. Data presented indicate a vital role of type I IFN within the inflamed liver for fine-tuning pathological processes such as overt iNOS expression.
Article
Acetaminophen (APAP), a widely used antipyretic and analgesic drug in clinics, is relatively safe at therapeutic doses; however, APAP overdose may lead to fatal acute liver injury. Currently, N-acetylcysteine (NAC) is clinically used as the main antidote for APAP poisoning, but its therapeutic effect remains limited owing to rapid disease progression and the general diagnosis of advanced poisoning. As is well known, APAP-induced hepatotoxicity (AIH) is mainly caused by the toxic metabolite N-acetyl- p-benzoquinone imine (NAPQI), and the toxic mechanisms of AIH are complicated. Several cellular processes are involved in the pathogenesis of AIH, including liver metabolism, mitochondrial oxidative stress and dysfunction, sterile inflammation, endoplasmic reticulum stress, autophagy, and microcirculation dysfunction. Mitochondrial oxidative stress and dysfunction are the major cellular events associated with APAP-induced liver injury. Many biomolecules involved in these biological processes are potential therapeutic targets for AIH. Therefore, there is an urgent need to comprehensively clarify the molecular mechanisms underlying AIH and to explore novel therapeutic strategies. This review summarizes the various cellular events involved in AIH and discusses their potential therapeutic targets, with the aim of providing new ideas for the treatment of AIH.
Article
Full-text available
(1) Aim of the Study: In this study, we aimed to evaluate the vascular damage and the effects of nitric oxide synthase (NOS) enzyme inhibitors in hepatic damage caused by high doses of acetaminophen (APAP). (2) Material and methods: Fifty-three Swiss albino male mice were used for this study. Hepatic and thoracic aorta toxicity caused by 2 or 6 h exposures to APAP (300 mg/kg intraperitoneally (i.p.)) were evaluated. The general NOS inhibitor NG-nitro-L-arginine methyl ester (L-NAME: 25 mg/kg and 50 mg/kg, i.p.) and the neuronal NOS inhibitor 7-nitroindazole (7-NI: 15 mg/kg, i.p.) were administered one hour before APAP exposure. (3) Results: Significant morphological deteriorations were observed after 6 h of APAP exposure in histopathological examinations of hepatic sections. Pre-treatment with L-NAME (at 50 mg/kg) or 7-NI before a 6 h APAP exposure significantly decreased hepatic toxicity (p < 0.05). Significant increases in ALT levels in 6 h of APAP exposure were decreased by both L-NAME (with the 25 mg/kg but not at 50 mg/kg) and 7-NI pre-treatments. No significant change was observed in the measured nitrate/nitrite levels and total antioxidant status in either serum or liver homogenates. No significant deteriorations were observed during either hematoxylin-eosin or immunohistochemical staining in thoracic aorta sections. In the thoracic artery sections, no statistical difference was found in acetylcholine-mediated relaxation, which may indicate endothelial dysfunction. (4) Conclusions: This study demonstrated that APAP-induced hepatic toxicity, especially neuronal NOS inhibitors, may decrease hepatic toxicity. It was also shown that APAP-induced hepatic toxicity was not accompanied by vascular dysfunction.
Article
Acute kidney injury (AKI) is a progressive renal complication which significantly affects the patient's life with huge economic burden. Untreated acute kidney injury eventually progresses to a chronic form and end-stage renal disease. Although significant breakthroughs have been made in recent years, there are still no effective pharmacological therapies for the treatment of acute kidney injury. Toll-like receptor 4 (TLR4) is a well-characterized pattern recognition receptor, and increasing evidence has shown that TLR4 mediated inflammatory response plays a pivotal role in the pathogenesis of acute kidney injury. The expression of TLR4 has been seen in resident renal cells, including podocytes, mesangial cells, tubular epithelial cells and endothelial cells. Activation of TLR4 signaling regulates the transcription of numerous pro-inflammatory cytokines and chemokines, resulting in renal inflammation. Therefore, targeting TLR4 and its downstream effectors could serve as an effective therapeutic intervention to prevent renal inflammation and subsequent kidney damage. For the first time, this review summarizes the literature on acute kidney injury from the perspective of TLR4 from year 2010 to 2020. In the current review, the role of TLR4 signaling pathway in AKI with preclinical evidence is discussed. Furthermore, we have highlighted several compounds of natural and synthetic origin, which have the potential to avert the renal TLR4 signaling in preclinical AKI models and have shown protection against AKI. This scientific review provides new ideas for targeting TLR4 in the treatment of AKI and provides strategies for the drug development against AKI.
Article
On renal ischemia-reperfusion (I/R) injury, recruitment of neutrophils during the inflammatory process promotes local generation of oxygen and nitrogen reactive species, which, in turn, are likely to exacerbate tissue damage. The mechanism by which inducible nitric oxide synthase (iNOS) is involved in I/R has not been elucidated. In this work, the selective iNOS inhibitor l- N6-(1-iminoethyl)lysine (l-NIL) and the NOS substrate l-arginine were employed to understand the role of NOS activity on the expression of particular target genes and the oxidative stress elicited after a 30-min of bilateral renal ischemia, followed by 48-h reperfusion in Balb/c mice. The main findings of the present study were that pharmacological inhibition of iNOS with l-NIL during an I/R challenge of mice kidney decreased renal injury, prevented tissue loss of integrity, and improved renal function. Several novel findings regarding the molecular mechanism by which iNOS inhibition led to these protective effects are as follows: 1) a prevention of the I/R-related increase in expression of Toll-like receptor 4 (TLR-4), and its downstream target, IL-1β; 2) reduced oxidative stress following the I/R challenge; noteworthy, this study shows the first evidence of glutathione S-transferase (GST) inactivation following kidney I/R, a phenomenon fully prevented by iNOS inhibition; 3) increased expression of clusterin, a survival autophagy component; and 4) increased expression of nuclear factor of activated T cells 5 (NFAT-5) and its target gene aquaporin-1. In conclusion, prevention of renal damage following I/R by the pharmacological inhibition of iNOS with l-NIL was associated with the inactivation of proinflammatory pathway triggered by TLR-4, oxidative stress, renoprotection (autophagy inactivation), and NFAT-5 signaling pathway.
Article
Full-text available
Paracetamol (acetaminophen) was marketed in the 1950s as a nonprescription analgesic/antipyretic without any preclinical toxicity studies. It became used increasingly for self‐poisoning, particularly in the UK and was belatedly found to cause acute liver damage, which could be fatal. Management of poisoned patients was difficult as maximum abnormalities of liver function were delayed for 3 days or more after an overdose. There was no treatment and the mechanism of hepatotoxicity was not known. The paracetamol half‐life was prolonged with liver damage occurring when it exceeded 4 h and the Rumack–Matthew nomogram was an important advance that allowed stratification of patients into separate zones of risk. It is used to guide prognosis and treatment and its predictive value could be increased by combining it with the paracetamol half‐life. The problems of a sheep farmer in Australia in the early 1970s led to the discovery of the mechanism of paracetamol hepatotoxicity, and the first effective treatment of overdosage with intravenous (IV) cysteamine. This had unpleasant side effects and administration was difficult. N‐acetylcysteine soon became the treatment of choice for paracetamol overdose and given early it was very effective when administered either IV or orally. N‐acetylcysteine could cause anaphylactoid reactions, particularly early during IV administration when the concentrations were highest. Simpler and shorter regimes with slower initial rates of infusion have now been introduced with a reduced incidence of these adverse effects. In addition, there has been a move to use larger doses of N‐acetylcysteine given over longer periods for patients who are more severely poisoned and those with risk factors. There has been much interest recently in the search for novel biomarkers such as microRNAs, procalcitonin and cyclophilin that promise to have greater specificity and sensitivity than transaminases. Paracetamol–protein adducts predict hepatotoxicity and are specific biomarkers of toxic paracetamol metabolite exposure. Another approach would be measurement of the plasma levels of cysteine and inorganic sulfate. It is 50 years since the first effective treatment for paracetamol poisoning and, apart from liver transplantation, there is still no effective treatment for patients who present late.
Article
Full-text available
The objective of this study was to determine whether endogenous nitric oxide (NO) inhibits leukocyte adhesion to vascular endothelium. This was accomplished by superfusing a cat mesenteric preparation with inhibitors of NO production, NG-monomethyl-L-arginine (L-NMMA) or NG-nitro-L-arginine methyl ester (L-NAME), and observing single (30-microns diameter) venules by intravital video microscopy. Thirty minutes into the superfusion period the number of adherent and emigrated leukocytes, the erythrocyte velocity, and the venular diameter were measured; venular blood flow and shear rate were calculated from the measured parameters. The contribution of the leukocyte adhesion glycoprotein CD11/CD18 was determined using the CD18-specific monoclonal antibody IB4. Both inhibitors of NO production increased leukocyte adherence more than 15-fold. Leukocyte emigration was also enhanced, whereas venular shear rate was reduced by nearly half. Antibody IB4 abolished the leukocyte adhesion induced by L-NMMA and L-NAME. Incubation of isolated cat neutrophils with L-NMMA, but not L-NAME, resulted in direct upregulation of CD11/CD18 as assessed by flow cytometry. Decrements in venular shear rate induced by partial occlusion of the superior mesenteric artery in untreated animals revealed that only a minor component of L-NAME-induced leukocyte adhesion was shear rate-dependent. The L-NAME-induced adhesion was inhibited by L-arginine but not D-arginine. These data suggest that endothelium-derived NO may be an important endogenous modulator of leukocyte adherence and that impairment of NO production results in a pattern of leukocyte adhesion and emigration that is characteristic of acute inflammation.
Article
The role of nitric oxide (NO) in inflammation represents one of the most studied yet controversial subjects in physiology. A number of reports have demonstrated that NO possesses potent anti-inflammatory properties, whereas an equally impressive number of studies suggest that NO may promote inflammation-induced cell and tissue dysfunction. The reasons for these apparent paradoxical observations are not entirely clear; however, we propose that understanding the physiological chemistry of NO and its metabolites will provide a blueprint by which one may distinguish the regulatory/anti-inflammatory properties of NO from its deleterious/proinflammatory effects. The physiological chemistry of NO is complex and encompasses numerous potential reactions. In an attempt to simplify the understanding of this chemistry, the physiological aspects of NO chemistry may be categorized into direct and indirect effects. This type of classification allows for consideration of timing, location, and rate of production of NO and the relevant targets likely to be affected. Direct effects are those reactions in which NO interacts directly with a biological molecule or target and are thought to occur under normal physiological conditions when the rates of NO production are low. Generally, these types of reactions may serve regulatory and/or anti-inflammatory functions. Indirect effects, on the other hand, are those reactions mediated by NO-derived intermediates such as reactive nitrogen oxide species derived from the reaction of NO with oxygen or superoxide and are produced when fluxes of NO are enhanced. We postulate that these types of reactions may predominate during times of active inflammation. Consideration of the physiological chemistry of NO and its metabolites will hopefully allow one to identify which of the many NO-dependent reactions are important in modulating the inflammatory response and may help in the design of new therapeutic strategies for the treatment of inflammatory tissue injury.
Article
The question whether nitric oxide protects or impairs organ perfusion during early endotoxemia has not been completely answered. To evaluate the regulative function of nitric oxide on organ microvascular perfusion and leukocyte accumulation during endotoxemia, we studied the influence of a non-selective nitric oxide inhibitor and a preferential inducible nitric oxide synthase inhibitor (respectively, N(G)-nitro-L-arginine methyl ester and aminoethyl-isothiourea) on liver microcirculation (intravital fluorescence microscopy) in a rat model. Two hours after intraportal injection of lipopolysaccharide (5 mg/kg in 10 min) the rats were randomly treated and received a bolus dose of N(G)-nitro-L-arginine methyl ester (10 mg/kg, n = 7), aminoethyl-isothiourea (10 mg/kg, n = 6) or normal saline, (n = 7). After 1 h, N(G)-nitro-L-arginine methyl ester blockade yielded a higher rate of non-perfused sinusoids than normal saline (27 +/- 2% vs 19 +/- 5%, p < 0.05). LPS-induced leukocyte stagnation in sinusoids was further increased (p < 0.05) in all groups after 1 h treatment, but N(G)-nitro-L-arginine methyl ester clearly accentuated leukocyte accumulation in sinusoids as compared to normal saline (69 +/- 19% vs 16 +/- 4%, p < 0.05). Both modalities of nitric oxide blockade elicited a significant enhancement in the number of leukocytes adherent to the postsinusoidal venules in contrast to normal saline (N(G)-nitro-L-arginine methyl ester 48 +/- 17%, aminoethyl-isothiourea 33 +/- 9% vs normal saline 1 +/- 5%, p < 0.05). We conclude that complete nitric oxide blockade aggravates lipopolysaccharide-induced hepatic microvascular perfusion failure and enhances leukocyte accumulation, in both sinusoids and post-sinusoidal venules. The preferential inducible nitric oxide synthase inhibitor aminoethyl-isothiourea has a moderate negative effect, favoring leukocyte adhesion in postsinusoidal venules, and its usefulness demands further research, especially concerning its late effects.
Article
Acetaminophen is a mild analgesic and antipyretic agent known to cause centrilobular hepatic necrosis at toxic doses. Although this may be due to a direct interaction of reactive acetaminophen metabolites with hepatocyte proteins, recent studies have suggested that cytotoxic mediators produced by parenchymal and nonparenchymal cells also contribute to the pathophysiological process. Nitric oxide is a highly reactive oxidant produced in the liver in response to inflammatory mediators. In the present studies we evaluated the role of nitric oxide in the pathophysiology of acetaminophen-induced liver injury. Treatment of male Long Evans Hooded rats with acetaminophen (1 g/kg) resulted in damage to centrilobular regions of the liver and increases in serum transaminase levels, which were evident within 6 hours of treatment of the animals and reached a maximum at 24 hours. This was correlated with expression of inducible nitric oxide synthase (iNOS) protein in these regions. Hepatocytes isolated from both control and acetaminophen-treated rats were found to readily synthesize nitric oxide in response to inflammatory stimuli. Cells isolated from acetaminophen-treated rats produced more nitric oxide than cells from control animals. Production of nitric oxide by cells from both control and acetaminophen-treated rats was blocked by aminoguanidine, a relatively specific inhibitor of iNOS. Arginine uptake and metabolism studies revealed that the inhibitory effects of aminoguanidine were due predominantly to inhibition of iNOS enzyme activity. Pretreatment of rats with aminoguanidine was found to prevent acetaminophen-induced hepatic necrosis and increases in serum transaminase levels. This was associated with reduced nitric oxide production by hepatocytes. Inhibition of toxicity was not due to alterations in acetaminophen metabolism since aminoguanidine had no effect on hepatocyte cytochrome P4502E1 protein expression or N-acetyl-p-benzoquinone-imine formation. Taken together, these data demonstrate that nitric oxide is an important mediator of acetaminophen-induced hepatotoxicity.