You are on page 1of 9

Liver International ISSN 1478-3223

CIRRHOSIS AND LIVER FAILURE

Lipopolysaccharide precipitates hepatic encephalopathy and increases


blood–brain barrier permeability in mice with acute liver failure
langer1, Bich N. Nguyen2 and Roger F. Butterworth1
Anne Chastre1, Mireille Be
1 Neuroscience Research Unit, Saint-Luc Hospital, CRCHUM, Montreal, QC, Canada
2 Department of pathology, Saint-Luc Hospital, CHUM, Montreal, QC, Canada

Keywords Abstract
acute liver failure – azoxymethane – blood– Background & Aims: Acute liver failure (ALF) is frequently complicated by
brain barrier – endotoxemia – infection leading to precipitation of central nervous system complications
immunoglobulin G extravasation – such as hepatic encephalopathy (HE) and increased mortality. There is evi-
lipopolysaccharide – matrix dence to suggest that when infection occurs in ALF patients, the resulting
metalloproteinase-9 – pro-inflammatory pro-inflammatory mechanisms may be amplified that could, in turn, have a
cytokines – systemic inflammatory response major impact on blood–brain barrier (BBB) function. The aim of this study
was to investigate the role of endotoxemia on the progression of encephalop-
Correspondence
athy in relation to BBB permeability during ALF. Methods: Adult male C57-
Roger F. Butterworth, PhD, DSc, BL6 mice with ALF resulting from azoxymethane-induced toxic liver injury
Neuroscience Research Unit, CRCHUM, were administered trace amounts of the endotoxin component lipopolysac-
Campus Saint-Luc, Universit
e de Montr
eal, charide (LPS). Effects on the magnitude of the systemic inflammatory
1058 St-Denis Street, H2X 3J4 Montreal, response, liver pathology and BBB integrity were measured as a function of
QC, Canada progression of HE, defined as time to loss of corneal reflex (coma). Results:
Tel: (514) 890-8000 (ext 35759) Lipopolysaccharide caused additional two- to seven-fold (P < 0.001)
Fax: (514) 412-7253 increases in circulating pro-inflammatory cytokines (TNF-a, IL-1b, IL-6),
e-mail: roger.butterworth@umontreal.ca worsening liver pathology and associated increases of circulating transamin-
ases as well as increased hyperammonaemia consistent with a further loss of
Received 3 April 2013 viable hepatocytes. LPS treatment of ALF mice led to a rapid precipitation of
Revised 16 May 2013 hepatic coma and the BBB became permeable to the 25-kDa protein immu-
Accepted 31 May 2013 noglobulin G (IgG). This extravasation of IgG was accompanied by ignificant
up-regulation of matrix metalloproteinase-9 (MMP-9), an endopeptidase
DOI:10.1111/liv.12252
known to modulate opening of the BBB in a wide range of neurological dis-
orders. Conclusions: These findings represent the first direct evidence of
inflammation-related BBB permeability changes in ALF.

Hepatic encephalopathy (HE) is the hallmark neurologi- are associated with an increased incidence of HE in ALF
cal feature and a determinant of clinical outcome in (2, 3). Endotoxin treatment leads to the release of
patients with acute liver failure (ALF). On account of pro-inflammatory cytokines such as tumour necrosis
the presence of multiple immunological defects, over factor-alpha (TNF-a) and the interleukins (IL-1b and
90% of ALF patients have clinical or bacteriological IL-6), which have been shown to enhance liver injury in
evidence of infection and there is a clear temporal asso- experimental animal models of ALF (4, 5).
ciation among the acquisition of the infection, the To start to elucidate the basic mechanisms that
appearance of a systemic inflammatory response and underlie the precipitation of HE by systemic inflamma-
progression of HE in these patients (1, 2). However, the tion caused by infection in ALF, this study made use of
mechanisms responsible for the precipitation of HE by a single low dose of LPS administered to mice with ALF
infection in ALF remain poorly defined. as a result of azoxymethane-induced hepatotoxicity.
Gram-negative bacteria are frequently implicated as This is a well-characterized mouse model of ALF that
the cause of infection/sepsis in ALF and increased circu- recapitulates the nature and extent of liver injury as well
lating levels of endotoxin, a lipopolysaccharide (LPS) as systemic inflammation and neurological complica-
complex of the outer cell wall of gram-negative bacteria, tions seen in the human condition (6). In view of the
recent resurgence of interest in changes in blood–brain
A portion of the results described in this manuscript was presented
barrier (BBB) in relation to HE in ALF (7) and the role
at the annual meeting of the AASLD, Boston, October 2012, and of inflammation in the pathogenesis of BBB disruption
published in abstract form Hepatology, 956A, 2012. in a wide range of neurological disorders (8–10), BBB

Liver International (2013)


© 2013 John Wiley & Sons A/S. Publishing by John Wiley & Sons Ltd. 1
Endotoxemia during acute liver failure in mice Chastre et al.

integrity was assessed by measurement of the extravasa- CTRL-LPS and ALF-Veh mice were pair-sacrificed when
tion of immunoglobulin G (IgG) and by measurement ALF+LPS mice reached coma stage of encephalopathy,
of metalloproteinase-9 activity (MMP-9), suggested which occurred on average 8.3 h following AOM
previously to contribute to altered BBB permeability to administration (Fig. 1). LPS doses in the lg/kg range
small molecules in this model of ALF (11). In this study, were chosen to induce non-lethal endotoxemia, as much
measurement of BBB integrity was made as a function higher doses (in the range of 1–25 mg/kg) are known to
of the presence of the systemic inflammatory response mimic sepsis in mice resulting in death of approxi-
and the progression of encephalopathy. mately half of animals (12). Following AOM injection,
body temperature was carefully monitored with a rectal
probe and rigorously maintained in the range of 36.5–
Material and methods 37.5°C using heating pads and lamps. Glycaemia was
monitored and kept at 5–7 mM by subcutaneous injec-
Animals
tions of 10% dextrose. Time to coma was determined by
Adult male C57BL6 mice (20–30 g) (Charles River, time to loss of corneal reflex. After anaesthesia with a
Saint-Constant, QC, Canada) were maintained in a 12-h ketamine/xylazine cocktail (50 and 9 mg/kg, respec-
light/dark cycle, and supplied with standard laboratory tively, i.p.), plasma samples were immediately collected
chow and water ad libitum. All animals were free of from the heart into heparinized tubes, centrifuged
infection at the onset of the experiment. All procedures (10 min, 12 000 g) and kept at 70°C. Mice were then
were carried out in accordance to the Guidelines of the perfused transcardially with saline. Brains were rapidly
Canadian Council of Animal Care and protocols were removed, flash frozen and kept at 70°C until use,
approved by the Animal Research Committee at Saint- whereas livers were fixed overnight by immersion in
Luc Hospital (CHUM). 10% buffered formalin.

Animal treatments and experimental design Histological assessment of liver damage


Acute liver failure was induced by the injection of Paraffin-embedded specimens were prepared and sec-
azoxymethane (AOM) (100 lg/g, i.p.) (Sigma-Aldrich, St. tions (6 lm) were mounted on Superfrost-plus micro-
Louis, CO, USA) dissolved in 100 ll saline as previously scope slides (Fisher Scientific, Pittsburg, PA, USA). HPS
described (6). To mimic infection, mice received a single (haematoxylin-phloxin-saffron) staining was performed
dose of LPS (2 lg/kg, i.p.) (Escherichia coli 0111:B4; according to a standard protocol and liver pathology
Sigma-Aldrich), dissolved in 100 ll saline 1 h after was assessed by an investigator blinded to the experi-
AOM injection. Five groups were constituted, namely mental treatment groups.
(i) Saline-treated mice (Control-Veh), (ii) LPS-treated
normal mice (Control-LPS), (iii) Saline-treated ALF
Biochemical assays
mice (ALF-Veh), (iv) LPS-treated ALF mice at coma
stages of HE (ALF+LPS) and (v) Saline-treated ALF Plasma samples were diluted 10-fold in saline and alanine
mice at coma stages of HE (ALF-Coma). CTRL-Veh, aminotransferase (ALT) and aspartate aminotransferase

PAIR-SACRIFICE COMA
Time (h) 0 1
8.3 ± 0.5 16.4 ± 0.4

Group 1 Saline Saline Control–Veh

Group 2 Saline LPS Control–LPS

Group 3 AOM Saline ALF–Veh

Group 4 AOM LPS ALF+LPS


(Precipitated Coma)

Group 5 AOM Saline ALF–Coma


(Separate study)

Fig. 1. Experimental design. Schematic representation of the procedures and timeline followed up for each group of this study. Mice from
CTRL-Veh, CTRL-LPS and ALF-Veh groups were sacrificed along with ALF+LPS mice, when ALF+LPS mice reached coma stages of hepatic
encephalopathy, on average 8.3 ± 0.5 h following AOM injection. ALF-Coma mice were sacrificed at coma stages of HE, which occur
16.4 ± 0.4 h following AOM injection.

Liver International (2013)


2 © 2013 John Wiley & Sons A/S. Publishing by John Wiley & Sons Ltd.
Chastre et al. Endotoxemia during acute liver failure in mice

(AST) activities were assayed with an automated analy- peroxidase-conjugated secondary antibody (1/3000, Per-
ser. Ammonia levels in plasma were determined using kin-Elmer Life Sciences). Peroxidase activity was then
a commercial ammonia assay kit (Sigma-Aldrich). detected by enhanced chemiluminescence (GE Health-
Samples were diluted 40-fold in assay diluent and absor- care). Intensity of the bands was measured by densitome-
bance was read at 340 nm. try using Quantity One software (Bio-Rad Laboratories).

Plasma IL-6, IL-1b and TNF-a measurements SDS–PAGE gelatin zymography


Pro-inflammatory cytokine levels were measured in Gelatin zymography was performed as previously
plasma using enzyme-linked immunosorbent assay described, with modifications (13). Twenty micrograms
(ELISA) kits specific for mouse IL-6 (eBioscience, San of protein fraction were electrophoresed onto 10% non-
Diego, CA, USA), IL-b and TNF-a (R&D Systems, Min- denaturing SDS–PAGE containing 1 mg/ml of gelatin
neapolis, MN, USA). Detection limits were 4 pg/ml, (Bio-Rad Laboratories). Mixtures (1.5 ng) of recombi-
3 pg/ml and 5.1 pg/ml respectively. For IL-1b and TNF- nant MMP-9 and MMP-2 (US Biological, Cleveland,
a assays, samples were diluted 20-fold and 5-fold, OH, USA) were used as standards. The gels were
respectively, in calibrator diluent and incubated 2 h at processed, incubated in 2.5% Triton X-100 for 1 h,
room temperature. For IL-6 measurements, samples incubated in a zymogen buffer (50 mM Tris–HCl, pH
were diluted 17-fold in assay diluent and incubated 7.4, 5 mM CaCl2, 200 mM NaCl, 0.02% Brij.35) at
overnight at 4°C. The plates were read at 450 nm and 37°C for 40 h, stained with 0.5% Coomassie Blue R-250
values at 570 nm were subtracted. Absorbance was con- for 3 h, and destained with three changes of 30% meth-
verted to pg/ml for cytokine measurements in plasma anol, 10% acetic acid (for 15, 30 and 60 min). Gels were
using standard curves prepared with respective recombi- scanned and the bands were quantified using Quantity
nant cytokines. One Software (Bio-Rad Laboratories). Gelatinase activ-
ity appears as a clear band against a blue background.
Western blot analysis
Statistical analysis
Samples of frontal cortex were homogenized in ice-cold
buffer (50 mM Tris pH 7.5, 150 mM NaCl, 1 mM All data are expressed as the mean ± SEM. Statistical
EDTA, 1% Triton X-100, 0.1% SDS, 0.5% NaDOC) analyses were performed using one-way analysis of vari-
containing a protease inhibitor cocktail (Sigma-Aldrich) ance (ANOVA) followed by Tukey’s post hoc analysis.
and centrifuged at 12 000 g for 45 min. Proteins P value < 0.05 was considered to indicate a significant
(10–30 lg) were resolved on 8% denaturing SDS-poly- difference. Data were analysed using Prism 5.0 software
acrylamide gels and transferred for 1 h to polyvinylid- (GraphPad Prism 5.0, San Diego, CA, USA).
ene fluoride (PVDF) membranes (Bio-Rad Laboratories,
Hercules, CA, USA). The membranes were probed for
1 h with antibodies directed against ZO-1 (rabbit, Results
1/1000, #61-7300, Invitrogen, Carlsbad, CA, USA),
LPS treatment worsens hepatic damage following AOM
ZO-2 (rabbit, 1/1000, #38-9100, Invitrogen), occludin
administration
(rabbit, 1/1000, #71-1500, Invitrogen) and b-actin
(mouse, 1/25 000, #A3853, Sigma-Aldrich), then Azoxymethane administration rapidly induced diffuse
incubated for 1 h with respective horseradish peroxi- necrosis affecting primarily centrilobular and mediolob-
dase-conjugated secondary antibodies (1/10 000, Per- ular regions in ALF-Veh mice (Fig. 2C), which
kin-Elmer Life Sciences, Boston, MA, USA). After expanded at coma stages of HE to massive haemorrhag-
extensive washing, peroxidase activity was detected by ic necrosis and apoptosis with scarce neutrophil infiltra-
enhanced chemiluminescence (GE Healthcare, Arling- tion in ALF-Coma mice (Fig. 2D). In contrast,
ton Heights, IL, USA). The intensity of the bands was ALF+LPS mice displayed peliosis and increased neutro-
measured by densitometry using Quantity One software phil infiltration in addition to the massive haemorrhagic
(Bio-Rad Laboratories). necrosis and apoptosis found in ALF-Coma mice
(Fig. 2E,F). LPS treatment alone induced hepatocyte
ballooning (Fig. 2B). Both plasma transaminase activi-
IgG extravasation
ties and ammonia levels were significantly increased at
IgG (25 kDa) extravasation was performed as previously the onset of coma in ALF+LPS mice. Compared with
described, with modifications (13). Briefly, twenty CTRL-Veh mice, AST activity was increased 9.2-fold
micrograms of proteins from samples of frontal cortex (P < 0.001) in ALF+LPS mice, whereas ALT activity was
were solubilized in Laemmli buffer, boiled for 5 min, increased 29.6-fold (P < 0.001) (Table 1). Plasma
then resolved on 8% denaturing SDS–PAGE and trans- ammonia levels were increased 2.8-fold (P < 0.001) in
ferred 1 h to PVDF membranes. Membranes were incu- ALF+LPS mice compared with CTRL-Veh mice
bated for 1 h with an anti-mouse horseradish (Table 1). No changes were observed in transaminase

Liver International (2013)


© 2013 John Wiley & Sons A/S. Publishing by John Wiley & Sons Ltd. 3
Endotoxemia during acute liver failure in mice Chastre et al.

(A) (B)

(C) (D)

(E) (F)

Fig. 2. LPS worsens hepatic pathology in ALF mice. Representative liver sections from (A) CTRL-Veh mice, (B) CTRL-LPS mice, (C) ALF-Veh
mice, (D) ALF-Coma mice and (E and F) ALF+LPS mice. Mice from CTRL-Veh, CTRL-LPS and ALF-Veh groups were sacrificed along with
ALF+LPS mice, when ALF+LPS mice reached coma stages of HE, on average 8.3 ± 0.5 h following AOM injection. ALF-Coma mice were
sacrificed at coma stages of HE, which occur 16.4 ± 0.4 h following AOM injection. ALF-Veh mice show early centrolobular necrosis (C) that
progresses into extensive haemorrhagic liver necrosis at coma stages in ALF-Coma mice (D). LPS challenge during ALF results in extensive
coagulative liver necrosis with peliosis and increased neutrophil infiltration in comatose ALF+LPS mice (E and F). Black arrow shows
neutrophil infiltration. Arrow heads indicate areas of apoptosis. Representative pictures from n = 5 in each group. Scale bar: 50 lm.

Table 1. LPS increases plasma transaminase activities and ammonia levels in ALF mice
CTRL-Veh CTRL-LPS ALF-Veh ALF+LPS ALF-Coma
AST (U/L) 310 ± 46 860 ± 180 1193 ± 299 2843 ± 422* 4086 ± 484*
ALT (U/L) 96 ± 23 215 ± 87 535 ± 351 2842 ± 511* 4057 ± 2176*
Ammonia (lM) 131.8 ± 26.3 118.0 ± 11.3 144.9 ± 24.7 370.2 ± 53.0* 535.1 ± 63.0*

Mice were administered LPS 1 h after azoxymethane (AOM) or saline (vehicle). CTRL-Veh mice received only saline. CTRL-LPS mice received only LPS.
CTRL-Veh mice, CTRL-LPS mice and ALF-Veh mice were sacrificed along with ALF+LPS mice, when ALF+LPS mice reached coma stages of HE, on aver-
age 8.3 ± 0.5 h following AOM injection. ALF-Coma mice were sacrificed at coma stages of HE, which occur 16.4 ± 0.4 h following AOM injection.
Data represent mean ± SEM of n = 10 in each group.
*P < 0.01 vs. all groups.

activities or in ammonia levels in CTRL-LPS mice or in ALF-Coma mice, whereas plasma levels of ammonia
ALF-Veh (8 h) mice. Compared with CTRL-Veh mice, were increased 4.1-fold (P < 0.001). Plasma levels of
AST and ALT activities were, respectively, increased AST, ALT and ammonia in ALF-Coma mice were not
13.2-fold (P < 0.001) and 42.2-fold (P < 0.001) in significantly different from those ALF+LPS mice.

Liver International (2013)


4 © 2013 John Wiley & Sons A/S. Publishing by John Wiley & Sons Ltd.
Chastre et al. Endotoxemia during acute liver failure in mice

LPS treatment results in further increases in plasma †


pro-inflammatory cytokines in ALF mice 400 (A)
Compared with CTRL-Veh mice, significant increases in
TNF-a levels were observed in ALF-Veh mice (3.6-fold;
**
P < 0.05) and in ALF+LPS mice (7.1-fold; P < 0.001) 300

Plasma TNF-α (pg/mL)


(Fig. 3A). Interestingly, TNF-a levels in ALF+LPS mice
were 2-fold higher than those observed in ALF-Veh
mice (P < 0.01) (Fig. 3A). Similarly, IL-6 levels were
200
significantly increased in ALF-Veh mice (153.5-fold; *
P < 0.05) and in ALF+LPS mice (332.0-fold; P < 0.001)
compared with CTRL-Veh mice (Fig. 4B). IL-6 levels
were significantly higher in ALF+LPS compared with 100
ALF-Veh mice (2.2-fold; P < 0.01) (Fig. 3B). IL-1b lev-
els were significantly increased in ALF+LPS mice com-
pared with CTRL-Veh mice (48.9-fold; P < 0.001)
0
(Fig. 3C). LPS alone did not significantly increase CTRL–Veh CTRL–LPS ALF–Veh ALF+LPS
plasma levels of TNF-a, IL-1b or IL-6.

15 000

(B)
LPS treatment results in a further increase in MMP-9
activity in AOM-induced ALF mice
**
No changes of protein expression were observed for
ZO-1, ZO-2 or occludin in either group (data not
Plasma IL-6 (pg/mL)

shown). Gelatin zymography analysis showed increased 10 000


activity of brain MMP-9 in ALF-Veh mice (5.7-fold;
P < 0.05) and in ALF+LPS mice (11.3-fold; P < 0.001)
compared with CTRL-Veh mice (Fig. 4). MMP-9 activ- *
ity in brains of ALF+LPS mice was significantly higher 5000
than that in the brains of ALF-Veh mice (2-fold;
P < 0.01). LPS treatment alone had no effect on brain
MMP-9 activity. No changes in MMP-2 activity were
demonstrated in any of the treatment groups (Fig. 4).
0
CTRL–Veh CTRL–LPS ALF–Veh ALF+LPS
LPS treatment results in IgG extravasation in brain of
AOM-induced ALF mice 2000
(C)
A significant increase in brain IgG extravasation was
observed in ALF+LPS mice compared with CTRL-Veh ¥
mice (16.0-fold; P < 0.001) (Fig. 5). No significant IgG 1500
Plasma IL-1β (pg/mL)

extravasation was observed in LPS-Veh mice or in ALF-


Veh mice.

1000
Discussion
The AOM mouse model has been widely used for the
study of basic mechanisms implicated in the pathogene- 500
sis of the central nervous system (CNS) complications
of ALF (6, 7, 14–16). The model recapitulates the major
components of human ALF resulting from toxic liver
injury including characteristic liver pathology (17), 0
CTRL–Veh CTRL–LPS ALF–Veh ALF+LPS
raised serum transaminases, hyperammonemia, sys-
temic inflammation (15) and severe encephalopathy (2). Fig. 3. Circulating pro-inflammatory cytokines are further
In this study, the superposition of LPS in AOM-treated increased at coma stages of HE in LPS-treated ALF mice. Plasma
mice resulted in rapid worsening of liver function as TNF-a (A), IL-6 (B) and IL-1b (C) were quantified by ELISA. Data
assessed by histopathology, further increases in serum represent mean ± SEM of n = 8 in each group. *P < 0.05 vs. CTRL-
transaminases and accelerated progression to coma Veh; **P < 0.001 vs. CTRL-Veh; †P < 0.01 vs. ALF-Veh; ¥P < 0.001
stages of encephalopathy. Ammonia concentrations vs. all other groups.

Liver International (2013)


© 2013 John Wiley & Sons A/S. Publishing by John Wiley & Sons Ltd. 5
Endotoxemia during acute liver failure in mice Chastre et al.

Fig. 4. ALF mice challenged with LPS manifest further increases in MMP-9 activity in brain compared with ALF mice. Activity of matrix
metalloproteinases 9 (MMP-9) and 2 (MMP-2) was quantified by zymography and expressed as a percentage of control. Data represent
mean ± SEM of n = 8 in each group. *P < 0.05 vs. CTRL-Veh; **P < 0.001 vs. CTRL-Veh; †P < 0.01 vs. ALF-Veh.

clearance of ammonia. No changes in any of the above


parameters were observed in LPS-treated control mice
indicating that the low dose of LPS used in this study
had no harmful effects per se on the liver in the absence
of AOM.
Treatment of mice with AOM alone resulted in
increased concentrations of the circulating pro-inflam-
matory cytokines TNF-a and IL-6 consistent with a
systemic inflammatory response in these animals that
was comparable to that reported in patients with ALF
owing to toxic liver injury (18) and in other experimen-
tal models of ALF (19) where the precise nature and
magnitude of the cytokine increases may vary depend-
ing upon the nature of the toxin (20).
It has been proposed that liver-derived pro-inflam-
matory cytokines are involved in the pathogenesis of
intracranial hypertension in ALF. This proposal was
based on a report of a patient with ALF resulting from
toxic liver injury awaiting liver transplantation who, on
account of severe uncontrolled intracranial hyperten-
sion, underwent an emergency hepatectomy (21); liver
removal led to a rapid sustained reduction in circulating
pro-inflammatory cytokines and a marked lowering of
intracranial pressure resulting in bridging to successful
liver transplantation.
Results of this study demonstrate that LPS adminis-
tration to AOM mice caused a further two- to three-fold
Fig. 5. LPS challenge results in IgG extravasation in brain of ALF increase in serum TNF-a and IL-6 and a seven-fold
mice at coma stages of encephalopathy. IgG (light chain, 25 kDa) increase in IL-1b compared with vehicle-treated ALF
extravasation was quantified by western blot analysis, normalized
vs. b-actin and expressed as a percentage of control. Data represent
mice. In parallel with this increased systemic inflamma-
mean ± SEM of n = 10 in each group. *P < 0.001 vs. all groups. tory response, LPS treatment of AOM mice resulted in
shortening of the time to appearance of coma defined
were also increased in these animals, consistent with as loss of corneal reflex. These findings of increases in
additional decreases in the capacity for urea synthesis concentration of circulating cytokines in relation to
by injured hepatocytes leading to decreased hepatic the precipitation of encephalopathy in ALF add to a

Liver International (2013)


6 © 2013 John Wiley & Sons A/S. Publishing by John Wiley & Sons Ltd.
Chastre et al. Endotoxemia during acute liver failure in mice

growing body of evidence in support of a fundamental Matrix metalloproteinase-9 has been implicated in
role of systemic inflammation in the pathogenesis of the pathogenesis of BBB disruption in a range of neuro-
HE in ALF. For example, previous studies demonstrate logical disorders including infectious encephalitis (35),
that TNF-a gene polymorphism significantly influences stroke (36) and traumatic brain injury (37). Evidence
ALF outcome in both humans and experimental ani- for a role of MMP-9 in the pathogenesis of BBB perme-
mals (18, 22), and TNF or IL-1 receptor gene deletion ability changes in ALF has, more recently, been reported
or plasma TNF-a inhibition delay the progression of (11) where it was suggested that the injured liver, not
HE in AOM mice (14, 23). the brain, was the source of the increased MMP-9. A
Effects of ALF on BBB integrity and function have subsequent report, however, disputed the interpretation
been the subject of considerable debate over the last sev- of these findings (38). Results of this study clearly dem-
eral decades (24). Studies in autopsied brain tissue from onstrate significant and selective increases in the activity
patients with ALF who died in hepatic coma as a result of MMP-9 in the brains of AOM mice, an effect that
of acetaminophen hepatotoxicity revealed swelling of as- started early in the progression of HE (prior to the onset
trocytes but no clear evidence of structural damage to of coma). Moreover, in the presence of LPS, a further
the BBB (25), and similar negative findings were two- to three-fold increase in MMP-9 activity was
reported in animal models of ALF resulting from galac- observed in brain as the animals became comatose.
tosamine-induced toxic liver injury (26, 27). On the Together with the findings of extravasation of IgG, these
other hand, one study in the AOM mouse suggested that results represent the first direct evidence implicating
the BBB becomes increasingly permeable to small mole- alterations of the BBB in the pathogenesis of HE precip-
cules such as water, as a result of discrete changes in itated by infection/inflammation in ALF and suggest
expression of proteins related to the tight junction (TJ) that up-regulation of brain MMP-9 activity is a possible
assembly of the barrier (7). However, a subsequent mechanism involved.
study could not confirm these findings (28). Results of The findings of a lack of significant alterations in
this study likewise could find no evidence of altered per- expression of the tight junction-related proteins occlu-
meability to IgG or alterations on expression of TJ-asso- din and ZO-1 in either group confirm, on one hand,
ciated proteins in AOM-induced ALF even at coma those of an earlier study in the same animal model
stages of encephalopathy. On the other hand, when (7) and suggest, on the other hand, that BBB disrup-
trace amounts of LPS were administered to mice with tion in ALF+LPS mice did not involve tight junction
AOM-induced ALF, unequivocal evidence of IgG digestion. Other mechanisms, such as the degradation
extravasation became immediately apparent as mice of the basal lamina by MMP-9, might participate in
became comatose. the increase in BBB permeability observed in
Thus, neither AOM-induced ALF alone nor LPS ALF+LPS mice. Interestingly, cerebral MMP-9 activity
treatment alone was sufficient to cause increased BBB was specifically induced, the expression and activation
permeability to IgG, but the two insults together did so of which is known to be modulated by inflammatory
unequivocally, suggestive of a synergistic mechanism. stimuli such as TNF-a and IL-1b (39, 40), which are
These findings suggest the possibility that factors that also involved in the CNS complications of ALF (14,
occur in ALF may be acting in concert with pro-inflam- 23, 41, 42).
matory mechanisms. One such factor is ammonia. Arte- In conclusion, the aim of this study was the establish-
rial ammonia concentrations are excellent predictors of ment of an in vivo model preparation for the study of
encephalopathy and intracranial hypertension in basic mechanisms implicated in the pathogenesis of
patients with ALF resulting from toxic liver injury (29, encephalopathy precipitated by infection/inflammation
30). In this study, LPS treatment of ALF mice resulted in ALF. Time to severe encephalopathy, in particular,
in a three-fold increase in circulating ammonia. Con- was chosen as a robust clinical endpoint for the studies.
centrations of ammonia comparable to those reported Results demonstrate that the superposition of ALF
in these studies and in LPS-treated ALF mice in this resulting from AOM-induced toxic liver injury together
study are known to facilitate ammonia diffusion into with endotoxemia (LPS) results in worsening of both
cerebrovascular endothelial cells (31), an action that is hepatic function and hyperammonaemia as well as
consistent with increased BBB permeability. Moreover, breakdown of the BBB and precipitation of HE. These
millimolar concentrations of ammonia inhibit glucose findings represent the first direct evidence for BBB per-
oxidation in brain mitochondria (32) resulting in accu- meability changes in ALF owing to toxic liver injury and
mulation of lactate. Brain lactate concentrations as well suggest a role for these changes in the pathogenesis of
as lactate synthesis rates correlate significantly with HE encephalopathy in this condition.
grade in experimental ALF (33). Moreover, exposure of
both astrocytes and microglial cells to lactate leads to
Acknowledgement
the release of pro-inflammatory cytokines (34) provid-
ing a possible explanation of synergism between ammo- Financial support: Studies from the authors’ laboratory
nia and neuroinflammation in relation to the were funded by operating grants from The Canadian
pathogenesis of encephalopathy in ALF. Institutes of Health Research.

Liver International (2013)


© 2013 John Wiley & Sons A/S. Publishing by John Wiley & Sons Ltd. 7
Endotoxemia during acute liver failure in mice Chastre et al.

18. Bernal W, Donaldson P, Underhill J, Wendon J, Williams


References R. Tumor necrosis factor genomic polymorphism and out-
1. Rolando N, Wade J, Davalos M, et al. The systemic come of acetaminophen (paracetamol)-induced acute liver
inflammatory response syndrome in acute liver failure. failure. J Hepatol 1998; 29: 53–9.
Hepatology 2000; 32: 734–9. 19. Streetz K, Leifeld L, Grundmann D, et al. Tumor necrosis
2. Vaquero J, Polson J, Chung C, et al. Infection and the pro- factor alpha in the pathogenesis of human and murine ful-
gression of hepatic encephalopathy in acute liver failure. minant hepatic failure. Gastroenterology 2000; 119: 446–
Gastroenterology 2003; 125: 755–64. 60.
3. Iwai H, Nagaki M, Naito T, et al. Removal of endotoxin 20. Bemeur C, Butterworth R F. Liver-brain proinflammatory
and cytokines by plasma exchange in patients with acute signalling in acute liver failure: Role in the pathogenesis of
hepatic failure. Crit Care Med 1998; 26: 873–6. hepatic encephalopathy and brain edema. Metab Brain Dis
4. Galanos C, Freudenberg MA, Reutter W. Galactosamine- 2013; 28: 145–50.
induced sensitization to the lethal effects of endotoxin. 21. Jalan R, Pollok A, Shah SH, Madhavan K, Simpson KJ.
Proc Natl Acad Sci USA 1979; 76: 5939–43. Liver derived pro-inflammatory cytokines may be impor-
5. Takada Y, Ishiguro S, Fukunaga K, et al. Increased intra- tant in producing intracranial hypertension in acute liver
cranial pressure in a porcine model of fulminant hepatic failure. J Hepatol 2002; 37: 536–8.
failure using amatoxin and endotoxin. J Hepatol 2001; 34: 22. Tsuchiya N, Tokushige K, Yamaguchi N, et al. Influence
825–31. of TNF gene polymorphism in patients with acute and ful-
6. Belanger M, Cote J, Butterworth RF. Neurobiological minant hepatitis. J Gastroenterol 2004; 39: 859–66.
characterization of an azoxymethane mouse model of 23. Chastre A, Belanger M, Beauchesne E, et al. Inflammatory
acute liver failure. Neurochem Int 2006; 48: 434–40. cascades driven by tumor necrosis factor-alpha play a
7. Shimojima N, Eckman CB, McKinney M, et al. Altered major role in the progression of acute liver failure and its
expression of zonula occludens-2 precedes increased neurological complications. PLoS ONE 2012; 7: e49670.
blood-brain barrier permeability in a murine model of ful- 24. Blei AT, Larsen FS. Pathophysiology of cerebral edema in
minant hepatic failure. J Invest Surg 2008; 21: 101–8. fulminant hepatic failure. J Hepatol 1999; 31: 771–6.
8. Wispelwey B, Lesse AJ, Hansen EJ, Scheld WM. Haemo- 25. Kato M, Hughes RD, Keays RT, Williams R. Electron
philus influenzae lipopolysaccharide-induced blood brain microscopic study of brain capillaries in cerebral edema
barrier permeability during experimental meningitis in the from fulminant hepatic failure. Hepatology 1992; 15:
rat. J Clin Invest 1988; 82: 1339–46. 1060–6.
9. Moor AC, de Vries HE, de Boer AG, Breimer DD. The 26. Gove CD, Hughes RD, Ede RJ, Williams R. Regional cere-
blood-brain barrier and multiple sclerosis. Biochem Phar- bral edema and chloride space in galactosamine-induced
macol 1994; 47: 1717–24. liver failure in rats. Hepatology 1997; 25: 295–301.
10. de Vries HE, Blom-Roosemalen MC, van Oosten M, 27. Traber PG, Dal Canto M, Ganger DR, Blei AT. Electron
et al. The influence of cytokines on the integrity of the microscopic evaluation of brain edema in rabbits with
blood-brain barrier in vitro. J Neuroimmunol 1996; 64: galactosamine-induced fulminant hepatic failure:
37–43. ultrastructure and integrity of the blood-brain barrier.
11. Nguyen JH, Yamamoto S, Steers J, et al. Matrix metallo- Hepatology 1987; 7: 1272–7.
proteinase-9 contributes to brain extravasation and edema 28. Bemeur C, Chastre A, Desjardins P, Butterworth RF. No
in fulminant hepatic failure mice. J Hepatol 2006; 44: changes in expression of tight junction proteins or blood-
1105–14. brain barrier permeability in azoxymethane-induced
12. Warren HS. Editorial: mouse models to study sepsis syn- experimental acute liver failure. Neurochem Int 2010; 56:
drome in humans. J Leukoc Biol 2009; 86: 199–201. 3.
13. Beauchesne E, Desjardins P, Butterworth RF, Hazell AS. 29. Clemmesen JO, Larsen FS, Kondrup J, Hansen BA, Ott P.
Up-regulation of caveolin-1 and blood-brain barrier Cerebral herniation in patients with acute liver failure is
breakdown are attenuated by N-acetylcysteine in thiamine correlated with arterial ammonia concentration. Hepatolo-
deficiency. Neurochem Int 2010; 57: 830–7. gy 1999; 29: 648–53.
14. Bemeur C, Qu H, Desjardins P, Butterworth RF. IL-1 or 30. Tofteng F, Hauerberg J, Hansen BA, et al. Persistent
TNF receptor gene deletion delays onset of encephalopa- arterial hyperammonemia increases the concentration of
thy and attenuates brain edema in experimental acute liver glutamine and alanine in the brain and correlates with
failure. Neurochem Int 2010; 56: 213–5. intracranial pressure in patients with fulminant hepatic
15. Bemeur C, Vaquero J, Desjardins P, Butterworth RF. failure. J Cereb Blood Flow Metab 2006; 26: 21–7.
N-acetylcysteine attenuates cerebral complications of non- 31. Duchini A, Govindarajan S, Santucci M, Zampi G, Hof-
acetaminophen-induced acute liver failure in mice: antiox- man FM. Effects of tumor necrosis factor-alpha and inter-
idant and anti-inflammatory mechanisms. Metab Brain leukin-6 on fluid-phase permeability and ammonia
Dis 2010; 25: 241–9. diffusion in CNS-derived endothelial cells. J Investig Med
16. Rangroo TV, Thrane AS, Chanag J, et al. Real-time analy- 1996; 44: 474–82.
sis of microglial activation and motility in hepatic and 32. Lai JC, Cooper AJ. Brain alpha-ketoglutarate dehydroge-
hyperammonemic encephalopathy. Neuroscience 2012; nase complex: kinetic properties, regional distribution,
220: 247–55. and effects of inhibitors. J Neurochem 1986; 47: 1376–86.
17. Matkowskyj KA, Marrero JA, Carroll RE, et al. Azoxyme- 33. Zwingmann C, Chatauret N, Leibfritz D, Butterworth RF.
thane-induced fulminant hepatic failure in C57BL/6J mice: Selective increase of brain lactate synthesis in experimental
characterization of a new animal model. Am J Physiol acute liver failure: results of a [H-C] nuclear magnetic
1999; 277: G455–62. resonance study. Hepatology 2003; 37: 420–8.

Liver International (2013)


8 © 2013 John Wiley & Sons A/S. Publishing by John Wiley & Sons Ltd.
Chastre et al. Endotoxemia during acute liver failure in mice

34. Andersson AK, Ronnback L, Hansson E. Lactate induces 39. Quagliarello V, Scheld WM. Bacterial meningitis: patho-
tumour necrosis factor-alpha, interleukin-6 and interleu- genesis, pathophysiology, and progress. N Engl J Med
kin-1beta release in microglial- and astroglial-enriched 1992; 327: 864–72.
primary cultures. J Neurochem 2005; 93: 1327–33. 40. Sharief MK, Thompson EJ. In vivo relationship of tumor
35. Sporer B, Koedel U, Paul R, et al. Human immunodefi- necrosis factor-alpha to blood-brain barrier damage in
ciency virus type-1 Nef protein induces blood-brain patients with active multiple sclerosis. J Neuroimmunol
barrier disruption in the rat: role of matrix metallopro- 1992; 38: 27–33.
teinase-9. J Neuroimmunol 2000; 102: 125–30. 41. Wright G, Shawcross D, Olde Damink SW, Jalan R. Brain
36. Fujimura M, Gasche Y, Morita-Fujimura Y, et al. Early cytokine flux in acute liver failure and its relationship with
appearance of activated matrix metalloproteinase-9 and intracranial hypertension. Metab Brain Dis 2007; 22: 375–
blood-brain barrier disruption in mice after focal cerebral 88.
ischemia and reperfusion. Brain Res 1999; 842: 92–100. 42. Jiang W, Desjardins P, Butterworth RF. Cerebral inflam-
37. Rosenberg GA. Matrix metalloproteinases in brain injury. mation contributes to encephalopathy and brain edema in
J Neurotrauma 1995; 12: 833–42. acute liver failure: protective effect of minocycline. J Neu-
38. Palenzuela L, Mas A, Montaner J, Cordoba J. Matrix rochem 2009; 109: 485–93.
metalloproteinase-9 in fulminant hepatic failure.
Hepatology 2010; 51: 1475–6; author reply 76.

Liver International (2013)


© 2013 John Wiley & Sons A/S. Publishing by John Wiley & Sons Ltd. 9

You might also like