ArticlePDF Available

Lung Matrix Metalloproteinase Activation following Partial Hepatic Ischemia/Reperfusion Injury in Rats

Authors:

Abstract and Figures

Purpose: Warm hepatic ischemia-reperfusion (I/R) injury can lead to multiorgan dysfunction. The aim of the present study was to investigate whether acute liver I/R does affect the function and/or structure of remote organs such as lung, kidney, and heart via modulation of extracellular matrix remodelling. Methods: Male Sprague-Dawley rats were subjected to 30 min partial hepatic ischemia by clamping the hepatic artery and the portal vein. After a 60 min reperfusion, liver, lung, kidney, and heart biopsies and blood samples were collected. Serum hepatic enzymes, creatinine, urea, Troponin I and TNF-alpha, and tissue matrix metalloproteinases (MMP-2, MMP-9), myeloperoxidase (MPO), malondialdehyde (MDA), and morphology were monitored. Results: Serum levels of hepatic enzymes and TNF-alpha were concomitantly increased during hepatic I/R. An increase in hepatic MMP-2 and MMP-9 activities was substantiated by tissue morphology alterations. Notably, acute hepatic I/R affect the lung inasmuch as MMP-9 activity and MPO levels were increased. No difference in MMPs and MPO was observed in kidney and heart. Conclusions: Although the underlying mechanism needs further investigation, this is the first study in which the MMP activation in a distant organ is reported; this event is probably TNF-alpha-mediated and the lung appears as the first remote organ to be involved in hepatic I/R injury.
This content is subject to copyright. Terms and conditions apply.
Research Article
Lung Matrix Metalloproteinase Activation following Partial
Hepatic Ischemia/Reperfusion Injury in Rats
Giuseppina Palladini,1Andrea Ferrigno,1Vittoria Rizzo,2
Eleonora Tarantola,3Vittorio Bertone,3Isabel Freitas,3,4 Stefano Perlini,1
Plinio Richelmi,1and Mariapia Vairetti1
1Department of Internal Medicine and erapeutics, Fondazione IRCCS Policlinico S. Matteo, University of Pavia,
Via Ferrata 9A, 27100 Pavia, Italy
2Department of Molecular Medicine, Fondazione IRCCS Policlinico S. Matteo, University of Pavia, 27100 Pavia, Italy
3Department of Biology and Biotechnology “Lazzaro Spallanzani, University of Pavia, 27100 Pavia, Italy
4Institute of Molecular Genetics of the C.N.R. (IGM-CNR), Histochemistry and Cytometry Section, University of Pavia,
27100Pavia,Italy
Correspondence should be addressed to Mariapia Vairetti; mariapia.vairetti@unipv.it
Received  August ; Accepted  November ; Published  January 
Academic Editors: A. K. Nussler and G. Ramadori
Copyright ©  Giuseppina Palladini et al. is is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly
cited.
Purpose. Warm hepatic ischemia-reperfusion (I/R) injury can lead to multiorgan dysfunction. e aim of the present study
was to investigate whether acute liver I/R does aect the function and/or structure of remote organs such as lung, kidney, and
heart via modulation of extracellular matrix remodelling. Methods. Male Sprague-Dawley rats were subjected to  min partial
hepatic ischemia by clamping the hepatic artery and the portal vein. Aer a  min reperfusion, liver, lung, kidney, and heart
biopsies and blood samples were collected. Serum hepatic enzymes, creatinine, urea, Troponin I and TNF-alpha, and tissue matrix
metalloproteinases (MMP-, MMP-), myeloperoxidase (MPO), malondialdehyde (MDA), and morphology were monitored.
Results. Serum levels of hepatic enzymes and TNF-alpha were concomitantly increased during hepatic I/R. An increase in hepatic
MMP- and MMP- activities was substantiated by tissue morphology alterations. Notably, acute hepatic I/R aect the lung
inasmuch as MMP- activity and MPO levels were increased. No dierence in MMPs and MPO was observed in kidney and heart.
Conclusions. Although the underlying mechanism needs further investigation, this is the rst study in which the MMP activation in
a distant organ is reported; this event is probably TNF-alpha-mediated and the lung appears as the rst remote organ to be involved
in hepatic I/R injury.
1. Introduction
Reperfusionfollowingprolongedischemiamaycausepara-
doxical damage at several levels. is phenomenon, dened
as ischemia-reperfusion (I/R) injury, has been described in
the heart as well in other organs, such as the liver []. Among
theseveralinvolvedmechanisms,oxygentoxicityandfree
radical production play an important role []. Warm hepatic
I/Rinjurymayoccurnotonlyduringlivertransplantation,
but also following surgical resections requiring hepatic inow
occlusion for bleeding control. Via systemic yet poorly
understood mechanisms, I/R injury in one organ can also
lead to multiorgan dysfunction that is to tissue damage in
remote organs away from the body district where the I/R
damage is taking place. For example, I/R intestine damage
has been described to cause multiple organ dysfunction due
to uncontrolled production and release of cytokines and
other proinammatory molecules []. Recent data suggested
that also hepatic I/R injury may cause damage to remote
organs such as kidney [], heart [], and lung []. Indeed,
damaged liver tissue releases destructive proinammatory
cytokines and oxygen-derived radicals into the circulation
thatarelikelycausingfurtherdamagetoremoteorgans[].
In an experimental model, Colletti et al. [], showed that
Hindawi Publishing Corporation
e Scientific World Journal
Volume 2014, Article ID 867548, 10 pages
http://dx.doi.org/10.1155/2014/867548
e Scientic World Journal
hepatic I/R injury is associated with lung dysfunction such as
neutrophil inltration, edema, intraalveolar hemorrhage and
endothelial activation.
Inammatory cytokines such as TNF-alpha participate
to extracellular matrix (ECM) degradation following liver
injury, and hepatic TNF-alpha expression has been demon-
stratedtobeparallelwithinductionofmatrixmetallopro-
teinases (MMPs) [], collagenolytic zinc-dependent enzymes
that degrade several ECM constituents such as collagen,
gelatin, elastin, and bronectin []. A recent study []
reports increased liver MMP- expression following I/R
injury, and a correlation between serum MMP- and sever-
ity/progression of liver damage has been described in the
setting of I/R injury [], acute allogra rejection [], and
chronic viral hepatitis []. Also in the kidney, it has been
recently shown that MMPs play a role in the development
of endothelial damage during ischemia-reperfusion injury,
with increased MMP- activity paralleling degradation of
endothelial cells and the subsequent increase in vascular
permeability []. Cardiovascular dysfunction frequently
occurs aer major surgery or liver transplantation. Several
investigators have reported that liver ischemia is associated
with the release of vasoactive substance that inicts remote
cardiac damage [].
e mechanisms leading to the initiation of multior-
ganinjuryhavenotyetbeenelucidated.eaimofthe
present study was therefore to investigate whether acute liver
ischemia and reperfusion do aect the function and the
structure of remote organs such as kidney, heart, and lung via
the modulation of ECM remodelling. As indicators of remote
and local tissue damage and leukocyte inltration we used
malondialdehyde (MDA), an indicator of lipid peroxidation
rate, and myeloperoxidase (MPO), a neutrophil marker.
2. Material and Methods
2.1. Animals. e use of animals in this experimental study
was approved by the National Institute for Research, and the
animals were cared for according to its guidelines. irty-
two Male Sprague-Dawley rats (– g) with free access
to water and food were used.
2.2. Materials. All reagents were of the highest grade of purity
available and were purchased from Sigma Aldrich.
2.3. Ischemia-Reperfusion (I/R) Procedure. e eects of I/R
were studied in vivo in a partial normothermic hepatic I/R
model that has been previously reported [,]. Briey, the
abdomen was opened by a median incision while the rats
were anesthetized with pentobarbital ( mg/kg). Ischemia
totheleandmedianlobewasinducedbyclampingthe
portal vein and hepatic artery with microvascular clips for
 min, and the abdomen was temporary closed with a suture.
Aer  min of ischemia, the abdomen was reopened, the
clips were removed, the abdomen was closed again, and the
liverwasreperfusedformin.Topreventpostsurgicaldehy-
dration and hypotension mL of normal saline was injected
in the inferior vena cava immediately aer the removal of
the clips. e duration of the injection was approximately
 s. During I/R the animals (𝑛=17) were maintained on
warm support to prevent heat loss: rectal temperature was
maintained at 37 ± 0.1C. Sham-operated control animals
(𝑛=15) were similarly treated as compared with I/R
group for all the aspects of the experimental model: rats
were maintained under heat support during anaesthesia of
equal length of time, injected with cc saline and submitted
tosimilarmanipulationoftheliverhiluswithoutvascular
occlusion. Blood samples were obtained aer reperfusion and
immediately centrifuged to isolate serum. At the end of the
reperfusion period, tissue samples of the liver, ischemic lobes
(le), of the kidney (cortex and medulla), of the heart (le
ventricle),andofthelungweresnapfrozeninliquidnitrogen.
2.4. Biochemical Assays. Liver and kidney injury was assessed
by serum release of alanine transaminase (ALT), aspartate
transaminase (AST), lactate dehydrogenase (LDH), crea-
tinine, and urea by an automated Hitachi  analyser
(Roche/Hitachi, Indianapolis, IN, USA).
Heart injury was assessed by serum evaluation of highly
specic marker of myocardial cell damage such as Troponin I
(cTN) by an automated Hitachi  analyser (Roche/Hitachi,
Indianapolis, IN, USA).
e amounts of released TNF-alpha in serum were quan-
tied by commercial rat TNF-alpha ELISA kit from R&D
Systems (Minneapolis, USA) according to the manufacture
instructions.
e amounts of malondialdehyde (MDA) formation
were quantied by HPLC method using the Chromsystems
assay kit (Chromsystems GmbH, M¨
unchen). e assay was
performed according to the manufacturer instructions with
some modications: briey, the derivatized samples were
incubated for  minutes at C and nally used aer cen-
trifugation [].
Myeloperoxidase (MPO) activity was measured with
a uorimetric detection kit (Cayman Chemical) aer an
adequate tissue preparation. Briey liver, lung, kidney, and
heart tissues were homogenized (IKA-Ultraturrax T) in a
cooled . M potassium phosphate buer (pH .). Aer
addition of another equal volume of cooled . M potassium
phosphate buer, the homogenate was centrifuged at C
for  min at . rpm in order to pellet insoluble cellular
debris []. Pellets were resuspended in a cooled . M
potassium phosphate buer (pH ) containing .% hexa-,-
bis-decyltrimethylammonium bromide (HTAB) and homog-
enized. Samples were sonicated for  sec and submitted to
two cycles of freeze/thaw. Finally samples were centrifuged at
C for  min at . rpm and supernatants were imme-
diately frozen at Cforlateruser.OneunitofMPO
activity was dened as the amount of enzyme that caused the
formation of nmol of uorophore per minute at C.
2.5. Tissue Sources for MMPs Analysis. Aer sacrice liver,
kidney, heart, and lung were quickly excised and placed in
cold (C) buer ( mM Histidine,  mM sucrose, and
mM EDTA, pH .) to remove blood. Liver and lung were
weighed and subsequently cut, frozen in liquid nitrogen and
stored at C, until use. Kidney was cleaned of external
tissue; the renal cortex and medulla were separated and
e Scientic World Journal
subsequently frozen in liquid nitrogen and stored at C,
until use. Heart was separated in atria, right and le ventricle.
e le ventricle tissue samples were then frozen in liquid
nitrogenandstoredatC, until use.
2.5.1. Hepatic Protein Isolation. Hepatic MMPs were extrac-
ted by homogenisation (IKA-Ultraturrax T) of frozen liver
tissue, in an ice-cold extraction buer ( :  wt/vol) con-
taining % Triton X-,  mmol/L Tris-HCl,  mmol/L
NaCl, and  mmol/L CaCl2,pH.[]. e homogenate
was then centrifuged ( min at . rpm at C) and the
protein concentration of the supernatant was measured with
the colorimetric Lowry method []. Samples were stored at
Cbeforeuse.
2.5.2. Renal Protein Isolation. Fiy milligrams of cortex and
medulla were used to homogenize in a dissociation buer
containing  mmol/L cacodylic acid, . mmol/L NaCl,
mmol/L ZnCl
2,  mmol/L CaCl2,.mmol/LNaN
3,and
.% Triton X-, pH . []. e homogenate was then
shaken at Cforhandtheproteinconcentrationof
the supernatant was measured with the colorimetric Lowry
method []. Samples were stored at Cbeforeuse.
2.5.3. Cardiac and Pulmonary Protein Isolation. Le ventricle
myocardial and lung samples were homogenized in an ice-
cold extraction buer [] ( :  wt/vol) containing cacodylic
acid ( mmol/L), NaCl ( mmol/L), ZnCl2( mmol/L),
CaCl2( mmol/L), NaN3(. mmol/L), and Triton X-
.% vol/vol (pH ). e homogenate was then centrifuged
( min at . rpm) and the supernatant protein concentra-
tion was measured with the colorimetric Lowry method [].
Samples were stored at Cbeforeuse.
2.6. MMPs Zymography. In order to detect MMPs activity
present in the samples, the homogenate protein content was
normalized by a nal concentration of  𝜇g/mL in sample
loading buer (. M Tris-HCl, % sucrose w/v, % SDS
w/v, and .% bromphenol blue w/v, pH .). Aer dilution
the samples were loaded onto electrophoretic gels (SDS-
PAGE) containing mg/mL of gelatin under nonreducing
conditions [,], followed by zymography as described
previously [].
e zymograms were analyzed by densitometer (GS 
Densitometer BIORAD, Hercules, CA, USA) and data were
expressed as optical density (OD), reported to mg/mL
protein content.
2.7. Tissue Morphology. Liver and lung tissues were xed in
% paraformaldehyde in . M phosphate buer at pH .
for  hours, processed routinely and embedded in Paraplast
wax. Sections ( 𝜇m thick) were stained with hematoxylin
and eosin (H&E). To appraise the severity of hepatic injury,
H&E-stained sections were evaluated as follows: Grade ,
minimal or no evidence of injury; Grade , mild injury
consisting of cytoplasmic vacuolation and focal nuclear
pyknosis; Grade , moderate-to-severe injury with extensive
nuclear pyknosis, cytoplasmic hypereosinophilia, and loss of
intercellular borders; Grade , severe necrosis with disinte-
gration of hepatic cords, hemorrhage, occasional granulomas,
cytoplasmic pallor, and cellular swelling [].
To appraise the severity of lung injury, the number of
granulocytes was evaluated in H&E-stained sections and
calculated per microscopic eld []: the prepared sections
were coded and examined by an independent histologist in a
single-blind scoring procedure.
Kidney tissue was frozen in liquid nitrogen and stored
at C. Sections ( 𝜇m thick) were stained with H&E.
Microscopic criteria for tubular damage are tubular brush
border loss, cytoplasmic swelling, and cellular debris.
2.8. Statistical Analysis. Results are expressed as mean ±
error standard as specied. Comparisons between groups
were performed by unpaired 𝑡-test. When data distribution
was not normal, according to the Kolmogorov-Smirnov test,
Mann-Whitney test was used. All statistical procedures were
performed using the MedCalc statistical soware package
(... version). Value of 𝑃 < 0.05 was considered
signicant.
3. Results
3.1. Liver I/R Injury. Asexpected,serumlevelsofAST,ALT,
and LDH increased signicantly in animals submitted to
ischemia ( min) and reperfusion ( min) as compared
with sham-operated group (Tab l e ).
3.2. Kidney Function. Serum creatinine and urea did not
signicantly dier between sham and I/R groups (Table ).
3.3. Heart Biomarkers. Serum Troponin I (cTN), did not
show any dierence between groups (I/R versus sham-
operated) (Ta b l e ).
3.4. Biochemical Parameters (TNF-Alpha, MDA, and MPO).
e serum concentrations of TNF-alpha, an index of Kupf-
fer cell activation, increased aer ischemia/reperfusion as
reported in Figure . No dierence in MDA formation, as
products of lipid peroxidation, was observed in liver and
lung (Table ). e same trend was found in kidney medulla
and heart samples as compared with sham group (Heart:
0.113 ± 0.005 versus 0.122 ± 0.009,resp.;Kidneymedulla
0.492 ± 0.098 versus 0.528 ± 0.129, resp.). e MDA levels
were signicantly higher in ischemic group compared with
sham animals only in kidney cortex (0.205 ± 0.011 versus
0.142 ± 0.022 nmoli/mg/prot, 𝑃 < 0.05).
Tissue MPO activity, an indirect evidence of neutrophil
inltration,wasmeasuredinliverandlung;meanMPO
activity was increased in liver and lung comparing the
ischemic group with sham animals (Tab l e ). No changes in
the MPO levels were found in kidney and heart tissue during
hepatic I/R injury, as compared with sham group (data not
shown).
3.5. Gelatinolytic Activity. e activity of gelatinase-A
(MMP-) and gelatinase-B (MMP-) was evaluated to inves-
tigate the extent of hepatic, renal, lung, and heart MMPs
e Scientic World Journal
T : Serum levels of AST, ALT, LDH, creatinine, urea, and Troponin I in animals submitted to ischemia/reperfusion (I/R). Sham-operated
group (control) has been compared with I/R group: 𝑃 < 0.05. ese are the mean results of  dierent experiments ±S.E.M.
SHAM (𝑛=15)I/R(𝑛=17)
AST mU/mL 118.92 ± 9.31 670.83 ± 180.33
ALT mU/mL 41.83 ± 3.48 609.17 ± 191.16
LDH mU/mL 2169.4 ± 486.2 8022.7 ± 2197.8
Creatinine mg/dL 0.67 ± 0.05 0.74 ± 0.04
Urea mg/dL 48.33 ± 1.94 48.17 ± 1.7
Troponin I ng/mL 0.02 ± 0.01 0.02 ± 0.01
T : Liver and lung levels of MDA and MPO in animals submitted to ischemia/reperfusion (I/R). Sham-operated group (control) has
been compared with I/R group: 𝑃 < 0.05. ese are the mean results of  dierent experiments ±S.E.M.
SHAM (𝑛=15)I/R(𝑛=17)
MDA
Liver nmoli/mg/prot 0.292 ± 0.072 0.293 ± 0.062
Lung nmoli/mg/prot 0.206 ± 0.022 0.210 ± 0.044
MPO
Liver nmoli/min/mL 1.7 ± 0.07 2.0 ± 0.06
Lung nmoli/min/mL 1.1 ± 0.01 1.2 ± 0.01
0
2
4
6
8
10
12
14
16
(pg/mL)
SHAM
I/R
TNF-𝛼
F : Serum levels of TNF-𝛼in animals submitted to
ischemia/reperfusion (I/R). Sham operated group (control, 𝑛=15)
hasbeencomparedwithI/Rgroup(𝑛=17): 𝑃 < 0.05.eseare
the mean results of  dierent experiments ±S.E.M.
activity, potentially inducing interstitial degradation (Figure
). Both I/R and sham-operated groups showed detectable
MMP- and MMP- activities with the exception of the heart
where MMP- was not detectable.
In the liver, I/R was associated with a signicant increase
of gelatinase activity in the ischemic lobe (Figure ). Inter-
estingly, acute hepatic I/R was associated with lung MMP-
activation, while in kidney (cortex and the medulla) and in
heart no signicant dierence was observed between groups
(I/R versus sham). Only a no signicant increase in MMP-
was observed in kidney medulla (Figure ).
3.6. Liver Histology. A semiquantitative evaluation of liver
lesions showed a statistically signicant dierence in the
extent of liver damage when comparing sham-operated rats
and animals subjected to I/R (Score –: 0.6± 0.1 versus 2.4±
0.2,resp.).Intheischemiclobeoftheanimalssubmittedto
I/R several lobules showed a detectable damage. In particular,
hepatocyte necrosis and sinusoidal disarrangement (Figures
(b)-(c), Grade ) when compared with sham-operated
animals (Figure (a)).
3.7. Kidney Histology. When compared with the cortex of
sham-operated animals (Figure (a) andinset),thecortexof
animals submitted to hepatic I/R showed dilated interstitium.
Rats submitted to I/R showed damage to tubules with loss of
brushborderandpresenceofcellulardebris(Figure (b) and
inset). No signicant damageto glomeruli was detected. With
respect to the outer medulla of sham animals (Figure (c)),
the outer medulla of rats submitted to hepatic I/R showed
wider areas of interstitial uid accumulation in the inter-
stitium and disarrangement of thick limbs of Henle’s loop
(Figure (d)). In the inner medulla of animals submitted to
I/R, a few thin limbs of Henles loop appear dilated and with
increased cellularity in the stromal (Figure (f))withrespect
to sham animals (Figure (e)).
3.8. Lung Histology. A signicantly higher number of gran-
ulocyteswerefoundinthewallandlumenofalveoliofrats
submitted to I/R as compared with sham rats (Figure ). In
contrast with the lungs of sham-operated animals (Figures
(a),(b),and(c)), in the lungs of animals submitted to
hepatic I/R alveoli appear to be dilated (Figures (d) and
(e)), rare erythrocytes in alveolar capillaries (Figure (e)),
dilated lymphatics (Figure (d)), and abundant granulocytes
e Scientic World Journal
0
0.1
0.2
0.3
0.4
0.5
MMP-2 MMP-9
Liver
SHAM
I/R
(OD mm2/mg/mL protein)
(a)
MMP-2 MMP-9
0
0.25
0.5
0.75
1
1.25
1.5 Lung
SHAM
I/R
(OD mm2/mg/mL protein)
(b)
0
0.25
0.5
0.75
1
MMP-2
Heart
SHAM
I/R
(OD mm2/mg/mL protein)
(c)
MMP-2 MMP-9
0
0.25
0.5
0.75
1Kidney medulla
SHAM
I/R
(OD mm2/mg/mL protein)
(d)
MMP-2 MMP-9
0
0.25
0.5
0.75
1Kidney cortex
SHAM
I/R
(OD mm2/mg/mL protein)
(e)
F : Bar graphs of MMP- and MMP- activity in ischemic liver lobe (a), lung (b), heart (c), kidney cortex (d), and medulla (e). Sham-
operated group (control, 𝑛=15) has been compared with I/R group (𝑛=17): 𝑃 < 0.05.Dataareshownasmeanvalues±SEM.
e Scientic World Journal
CL
CL
P
P
P
(a)
CL
P
P
(b)
P
P
(c)
F : Representative light micrographs of the le liver lobe of sham-operated rats (a) and of the le lobe of rats submitted to
ischemia/reperfusion (I/R) (b and c). Hematoxylin and eosin staining. e sham animal shows normal hepatocyte and sinusoid morphology
(a). In animals submitted to I/R the lower magnication picture (b) shows decreased eosinophilia of hepatocytes, hepatocyte vacuolation,
disarrangement of hepatocyte cords, and altered sinusoidal dilatation (b). An area of extensive hepatocyte necrosis and plate disintegration
(black stars) is shown under higher magnication in (c), example of grade . P: portal vein; CL: centrolobular vein.
DCT
DCT
G
50 𝜇m
010𝜇m
(a)
DCT
50 𝜇m
010𝜇m
(b)
THL
THL
50 𝜇m
(c)
THL
THL
50 𝜇m
(d)
tHL
tHL
50 𝜇m
(e)
tHL
tHL 50 𝜇m
(f)
F : Representative light micrographs of kidney samples obtained from sham-operated rats (a, b, and c) and from rats submitted to
hepatic ischemia/reperfusion (I/R) (d, e, and f). Cortex (a, b), outer medulla (c, d), and inner medulla (e; f) are illustrated. Hematoxylin and
eosin staining. With respect to the normal morphology of the cortex of sham animals (a), the cortex of animals submitted to hepatic I/R (b)
shows dilated interstitium and injury to a few tubules. e insets in (a) show a normal distal convolute tubule (DCT) and in (b) patchy areas
of dilatation. With respect to the normal morphology of the outer medulla of sham animals (c), the outer medulla of animals submitted to
hepatic I/R shows extended areas of interstitial uid (asterisk) apparently displacing thick limbs of Henles loop (THL). Respect to the inner
medulla of sham animals (e), the inner medulla of animals submitted to I/R (f), shows slightly dilated thin limbs of Henle’s loop (tHL) and
increased cellularity in the stromal.
e Scientic World Journal
0
2
4
6
8
10
Cells/fields
Granulocytes
SHAM
I/R
F : Changes of lung granulocytes in response to hepatic I/R
injury. Lung samples were obtained from sham-operated rats, not
submittedtohepaticI/R,andfromratswhosehepaticlobeswere
submitted to  min of ischemia and hence reperfused for  min.
e number of granulocytes was calculated per microscopic eld.
𝑃 < 0.05. Data are shown as mean values ±SEM.
and mononuclear cells in the lumen and stroma of arteries
(Figures (e) and (f)) and in the lumen of bronchi (not
shown).
4. Discussion
isstudyshowsthatmoderateacutehepaticischemia/
reperfusion (I/R) injury increases MMPs activity not only in
theischemicliverregionbutalsointhelung,associatedwith
histological damage in liver, lung, and kidney. e concomi-
tant increase in serum TNF-alpha suggests a potential role for
this cytokine in the development of multiorgan dysfunction
arising from isolated hepatic I/R injury. A moderate hepatic
I/R injury is able to increase MMPs activity not only in the
ischemic region, as previously reported [,], but also
in the nonischemic lobe associated with several histological
signs of interstitial and cellular damage []. is event is
probably TNF-alpha-mediated, fully supporting the hypoth-
esis that a direct connection exists between the events taking
placeinboththedamagedlobeandthenonischemicliver.
4.1. e Involvement of Lung in the Hepatic I/R Injury. e
results of the present work suggest that a sizeable release of
hepatic enzymes in the blood stream following acute liver
I/R injury is associated with increased MMP activity, in
particular MMP- also in distant organs, such as the lung.
Multiorgan failure (MOF) is the simultaneous dysfunction of
several organs, and it represents one of the most intriguing
clinical problems arising in patients admitted to the Inten-
sive Care Unit []. A central mechanism leading to MOF
seems to be I/R injury []. Oxygen-derived free radicals,
cytokines, and activated neutrophils have been found to
be involved in the I/R liver damage []triggeringthe
systemic inammatory response that contributes to distant
organ injury []. Although the involved mechanism is still
unclear, the observed increase in MMP- activity appears
to be connected with the high serum levels of TNF-alpha
representing the connection between the hepatic I/R damage
and “at-a-distance” lung alterations.
MMPs are not expressed during normal conditions but
their expression and activity increase during inammation
[]. MMP- is one of the families of MMPs, which degrades
ECM, and it is induced by many inammatory factors,
including IL-beta, IL-, and TNF-alpha. MMP- is stored in
the tertiary granules of polymorphonuclear leukocytes which
are key eectors in acute inammatory diseases. In addition
MMP- can actively assist MPO activation, an index of
neutrophil inltration []. Once an inammatory response
is initiated, neutrophils are the rst cells to be recruited to the
sites of injury or []infection[]. Aer hepatic I/R injury
liver MPO activity increased in the ischemic tissue and a
few neutrophils were occasionally seen in edematous portal
spaces, and/or forming small granulomas around necrotic
cells in ischemic lobes. Interestingly, a similar increase of
MPO levels was also observed in the lung tissue, associated
with a high number of granulocytes as compared with the
control group.
Previous data also show increased neutrophil inltration
in the liver and in other organs such as the lung aer hepatic
I/R, suggesting that neutrophils contribute to MOF induced
by hepatic I/R []. In the present study, I/R injury was not
associated with an oxidative damage in the liver and in the
lung despite early signs of tissue damage. No signicant
dierencewasobservedinMDAlevels,alipidperoxidation
product, conrming our previous work in which liver MMP
activation was shown to be an MDA-independent event [].
A more prolonged reperfusion time is required to obtain a
signicant increase in MDA levels aer hepatic I/R damage
[].
4.2. Kidney Involvement in the Hepatic I/R Injury. No increase
in MMPs and MPO was observed in the kidney. Previ-
ously, Miranda et al. [] demonstrated that  min hepatic
ischemia associated with or hours reperfusion induced
an increase in MPO and MDA in distant organ such as
the lung and the kidney. In the present work we conrm
the increase in pulmonary MPO and the absence of these
alterations in other organs such as the kidney. A possible
explanation is strictly connected with the short duration of
both ischemia ( min) and reperfusion ( min) period in
our experimental model, as well as with the much lower
transaminase levels, -times lower than those observed by
Miranda et al. []. No changes in MDA formation was
found in distant organs such as the kidney, thus conrming
the limited damage induced in our experimental setting, as
supported by previous reports showing that the hepatic MDA
levels aer  min ischemia followed by min reperfusion
were comparable to those observed in sham animals [].
We did only nd increased MDA levels in the kidney
cortex. Further studies will be performed to explain this
nding. e histological analysis reveals some alterations
such as focal patchy areas of dilatation and much higher uid
accumulation in rats submitted to I/R versus control group.
e Scientic World Journal
alv
50 𝜇m
(a)
25 𝜇m
alv
(b)
25 𝜇m
(c)
alv
B
AL
50 𝜇m
(d)
25 𝜇m
alv
A
(e)
alv
B
A25 𝜇m
(f)
F : Representative sections of lung tissue from sham-operated animals (a, b, and c) and from animals submitted to hepatic
ischemia/reperfusion (I/R) (d, e, and f). Hematoxylin and eosin staining. e sections from sham animals show the typical morphology
of bronchi (B), blood vessels, and alveoli (alv) with associated capillaries; alveolar capillaries are recognizable in high magnication (c), by
erythrocytes in the lumen. In the lung tissues of animals submitted to hepatic I/R alveoli appear to be dilated. (d, e) Dilated lymph vessels (L)
surrounding arteries (A) and an abundant number of inammatory cells (arrowheads) in the lumen and stroma of blood vessels (f).
4.3. Heart Involvement in the Hepatic I/R Injury. No changes
in heart MMPs, MDA, and MPO were observed, probably
because cardiac dysfunction has been reported to follow only
major liver surgical procedures when the liver is subjected to
an important decrease in blood ow or aer transplantation
[]. Meyer et al. [] demonstrated that hepatic I/R induced
the upregulation of ICAM, one of the adhesion molecules,
mediated by TNF-alpha in distant organs such as the heart
and the kidney. ey evaluated the damage aer -hour
reperfusion and probably this is the reason why we did
not nd any cardiac alteration aer -hour reperfusion; our
hypothesis is supported by the strict correlation that exists
between lipid peroxidation formation and ischemia time in
the rat liver: only aer hours of reperfusion a marked MDA
was shown [].
4.4. Hepatic I/R Injury and MOF. Multiple organ dysfunc-
tion syndrome/failure is an important cause of death in
the surgical intensive care unit. As a syndrome, MODS is
dened as altered organ function in the setting of sepsis,
septic shock, or systemic inammatory response syndrome.
Our data show that also aer hepatic I/R, biochemical and
histological changes do occur in distant organs and these
events are traceable very early during reperfusion aer a brief
transient ischemia. A histopathology examination showed
that hepatic, pulmonary, and renal tissue were more injured,
albeit slightly, in the I/R rats than in sham animal. In
particular increased MMP- activity was associated with
early lung injury. Recently, some studies have shown that
signicant increases in active MMP- are associated with a
multiple organ dysfunction in an infection model []. Our
results suggest that aer hepatic I/R an increased MMP-
activation in distant organ can occur representing (a) a step
forward the comprehension of the mechanisms involved in
MOFand(b)aninnovativetargetforlimitingtheMOF
progression.
e Scientic World Journal
Interestingly, Rahman et al. recently demonstrated a
novel role of MMPs in regulating inltration of neutrophils
by controlling platelets secretion of CDL, a factor involved
in the septic lung injury. eir results suggested that targeting
MMPs may be a useful strategy for limiting lung injury [].
Experiments are in progress in our laboratory for increasing
both ischemia and reperfusion period.
Interestingly, the present work highlights that already
aer a relatively short I/R period an acute distant organ
damage occurs and that the lung is the rst organ involved,
suggesting that the increase in lung MMP- activity may
represent a key and early event involved in the pathogenesis
of hepatopulmonary syndrome, whereas kidney injury may
occur later and cardiac alterations may be observed only aer
a period of reperfusion longer than hour [].
e likelihood that an ischemic and reperfused organ
can directly induce a remote organ failure is of a signicant
clinical importance: these eects must be taken into account
when treating patients aer liver transplantation and these
ndings may have important practical applications in the
clinical management of liver transplantation, as well as in the
procedures involving no ow-reow conditions.
Abbreviations
MMP: Matrix metalloproteinases
I/R: Ischemia-reperfusion
MDA: Malondialdehyde
MPO: Myeloperoxidase.
Conflict of Interests
e authors state that no conict of interest or nancial dis-
closure exists.
Authors’ Contribution
Giuseppina Palladini and Andrea Ferrigno contributed
equally in this paper.
Acknowledgments
e authors thank Mr. Gaetano Viani and Massimo Costa
for the skillful technical assistance, Dr. Enrico Scoglio for the
technicalsupportduringtheHPLCassay,andMrs.Nicoletta
Breda for the editing assistance. is work was funded by
F.A.R. –—University of Pavia.
References
[] P. A. Grace, Ischemia-Reperfusion Injury,BlackwellScience,
London, UK, .
[] D. L. Carden and D. N. Granger, “Pathophysiology of ischa-
emia-reperfusion injury, Journal of Pathology,vol.,no.,
pp.,.
[] M. Oltean, S. Mera, R. Olofsson et al., “Transplantation of pre-
conditioned intestinal gras is associated with lower inamma-
tory activation and remote organinjury in rats, Trans pla ntati on
Proceedings,vol.,no.,pp.,.
[] M.Behrends,R.Hirose,Y.H.Parketal.,“Remoterenalinjury
following partial hepatic ischemia/reperfusion injury in rats,
Journal of Gastrointestinal Surgery,vol.,no.,pp.,
.
[] V.G.Nielsen,S.Tan,M.S.Baird,P.N.Samuelson,A.T.McCam-
mon, and D. A. Parks, “Xanthine oxidase mediates myocardial
injury aer hepatoenteric ischemia-reperfusion, Critical Care
Medicine, vol. , no. , pp. –, .
[] C.Peralta,J.C.Perales,R.Bartronsetal.,“ecombination
of ischemic preconditioning and liver Bcl- overexpression is a
suitable strategy to prevent liver and lung damage aer hepatic
ischemia-reperfusion, American Journal of Pathology,vol.,
no. , pp. –, .
[] H.Jiang,F.Meng,W.Li,L.Tong,H.Qiao,andX.Sun,“Splenec-
tomy ameliorates acute multiple organ damage induced by liver
warm ischemia reperfusion in rats, Surgery,vol.,no.,pp.
–, .
[]L.M.Colletti,S.L.Kunkel,A.Walzetal.,“Chemokine
expression during hepatic ischemia/reperfusion-induced lung
injury in the rat. e role of epithelial neutrophil activating
protein, Journal of Clinical Investigation,vol.,no.,pp.
, .
[]T.Knittel,M.Mehde,D.Kobold,B.Saile,C.Dinter,andG.
Ramadori, “Expression patterns of matrix metalloproteinases
and their inhibitors in parenchymal and non-parenchymal cells
of rat liver: regulation by TNF-𝛼and TGF-𝛽, Journal of
Hepatology,vol.,no.,pp.,.
[] P. Mignatti and D. B. Riin, “Nonenzymatic interactions
between proteinases and the cell surface: novel roles in normal
and malignant cell physiology, Advances in Cancer Research,
vol. , pp. –, .
[] C. Moore, X.-D. Shen, F. Gao, R. W. Busuttil, and A. J. Coito,
“Fibronectin-𝛼𝛽 integrin interactions regulate metallopro-
teinase- expression in steatotic liver ischemia and reperfusion
injury, American Journal of Pathology,vol.,no.,pp.
, .
[] J. P. Kuy venhoven, J. Ringers, H. W. Verspaget, C. B. H. W.
Lamers, and B. Van Hoek, “Serum matrix metalloproteinase
MMP- and MMP- in the late phase of ischemia and reperfu-
sion injury in human orthotopic liver transplantation, Tran s-
plantation Proceedings,vol.,no.,pp.,.
[] J. P. Kuyvenhoven, H. W. Verspaget, Q. Gao et al., Assessment
of serum-matrix metalloproteinases MMP- and MMP- aer
human liver transplantation: increased serum MMP- level in
acute rejection, Tran spl ant at ion ,vol.,no.,pp.,
.
[] V. Leroy, F. Monier, S. Bottari et al., Circulating matrix
metalloproteinases , , and their inhibitors TIMP- and
TIMP- as serum markers of liver brosis in patients with
chronic hepatitis C: comparison with PIIINP and hyaluronic
acid, American Journal of Gastroenterology,vol.,no.,pp.
–, .
[] T. Kuroda, Y. Yoshida, J. Kamiie et al., “Expression of MMP- in
mesangial cells and its changes in anti-GBM glomerulonephri-
tis in WKY rats, Clinical and Experimental Nephrology,vol.,
no.,pp.,.
[] E.Hochhauser,Z.Ben-Ari,O.Pappo,Y.Chepurko,andB.A.
Vidne, “TPEN attenuates hepatic apoptotic ischemia/reperfu-
sion injury and remote early cardiac dysfunction,” Apoptosis,
vol.,no.,pp.,.
[] R. Imberti, M. Vairetti, M. R. Gualea et al., “e eects of
thyroid hormone modulation on rat liver injury associated
 e Scientic World Journal
with ischemia-reperfusion and cold storage, Anesthesia and
Analgesia, vol. , no. , pp. –, .
[] G. Palladini, A. Ferrigno, V. Rizzo et al., “Lobe-specic hetero-
geneity and matrix metalloproteinase activation aer ischemia/
reperfusion injury in rat livers, Toxicologic Pathology,vol.,
pp. –, .
[] M. B. Grisham, L. Anzueto Hernandez, and D. N. Granger,
“Xanthine oxidase and neutrophil inltration in intestinal
ischemia, American Journal of Physiology: Gastrointestinal and
Liver Physiology,vol.,no.,pp.GG,.
[] A. E. Kossakowska, D. R. Edwards, S. S. Lee et al., Altered
balance between matrix metalloproteinases and their inhibitors
in experimental biliary brosis,” American Journal of Pathology,
vol. , no. , pp. –, .
[]O.H.Lowry,N.J.Rosebrough,A.L.Farr,andR.J.Randall,
“Protein measurement with the Folin phenol reagent, e
JournalofBiologicalChemistry,vol.,no.,pp.,.
[] T. M. Camp, L. M. Smiley, M. R. Hayden, and S. C. Tyagi,
“Mechanism of matrix accumulation and glomerulosclerosis in
spontaneously hypertensive rats, Journal of Hypertension,vol.
,no.,pp.,.
[] M. L. Coker, C. V. omas, M. J. Clair et al., “Myocardial matrix
metalloproteinase activity and abundance with congestive heart
failure, American Journal of Physiology. Heart and Circulatory
Physiology,vol.,no.,pp.HH,.
[] D. E. Kleiner and W. G. Stetler-Stevenson, “Quantitative
zymography: detection of picogram quantities of gelatinases,
Analytical Biochemistry,vol.,no.,pp.,.
[]S.C.Tyagi,S.E.Campbell,H.K.Reddy,E.Tjahja,andD.
J. Voelker, “Matrix metalloproteinase activity expression in
infarcted, noninfarcted and dilated cardiomyopathic human
hearts, Molecular and Cellular Biochemistry,vol.,no.,pp.
–, .
[] R. Tozzi, G. Palladini, S. Fallarini et al., “Matrix metalloprotease
activity is enhanced in the compensated but not in the decom-
pensated phase of pressure overload hypertrophy, American
Journal of Hypertension,vol.,no.,pp.,.
[] A. Seraf´
ın, J. Rosell´
o-Catafau, N. Prats, E. Gelp´
ı, J. Rod´
es,
and C. Peralta, “Ischemic preconditioning aects interleukin
release in fatty livers of rats undergoing ischemia/reperfusion,
Hepatology, vol. , no. , pp. –, .
[]E.Cozzi,S.Hazarika,H.W.StallingsIIIetal.,“Ultrane
particulate matter exposure augments ischemia-reperfusion
injury in mice, American Journal of Physiology. Heart and
Circulatory Physiology, vol. , no. , pp. H–H, .
[]T.Hamada,C.Fondevila,R.W.Busuttil,andA.J.Coito,
“Metalloproteinase- deciency protects against hepatic ische-
mia/reperfusion injury, Hepatolog y,vol.,no.,pp.,
.
[] T. Hamada, S. Duarte, S. Tsuchihashi, R. W. Busuttil, and A.
J. Coito, Inducible nitric oxide synthase deciency impairs
matrix metalloproteinase- activity and disrupts leukocyte
migration in hepatic ischemia/reperfusion injury, American
Journal of Pathology,vol.,no.,pp.,.
[] A. E. Baue, R. Durham, and E. Faist, “Systemic inammatory
response syndrome (SIRS), multiple organ dysfunction syn-
drome (MODS), multiple organ failure (MOF): Are we winning
the battle?” Shock,vol.,no.,pp.,.
[] K. Meyer, M. F. Brown, G. Zibari et al., “ICAM- upregulation
in distant tissues aer hepatic ischemia/reperfusion: a clue to
the mechanism of multiple organ failure, Journal of Pediatric
Surgery, vol. , no. , pp. –, .
[] C.-F. Chen, D. Wang, C. P. Hwang et al., “e protective eect
of niacinamide on ischemia-reperfusion-induced liver injury,
Journal of Biomedical Science,vol.,no.,pp.,.
[] M. Bhatia and S. Moochhala, “Role of inammatory mediators
in the pathophysiology of acute respiratory distress syndrome,
Journal of Pathology,vol.,no.,pp.,.
[] H. Nagase, R. Visse, and G. Murphy, “Structure and func-
tion of matrix metalloproteinases and TIMPs, Cardiovascular
Research, vol. , no. , pp. –, .
[] J. A. Smith, “Neutrophils, host defense, and inammation: a
double-edged sword, Journal of Leukocyte Biology,vol.,no.
, pp. –, .
[] M. Fukai, T. Hayashi, R. Yokota et al., “Lipid peroxidation
during ischemia depends on ischemia time in warm ischemia
and reperfusion of rat liver, Free Radical Biology and Medicine,
vol.,no.,pp.,.
[]L.E.C.Miranda,V.K.Capellini,G.S.Reis,A.C.Celotto,
C.G.CarlottiJr.,andP.R.B.Evora,“Eectsofpartialliver
ischemia followed by global liver reperfusion on the remote
tissue expression of nitric oxide synthase: lungs and kidneys,
Transplantation Proceedings, vol. , no. , pp. –, .
[] E. Hochhauser, I. Alterman, A. Weinbroum et al., “Eects of
vasoactive substances released from ischemic reperfused liver
on the isolated rat heart, ExperimentalandClinicalCardiology,
vol.,no.,pp.,.
[] L. Teng, M. Yu, J.-M. Li et al., “Matrix metalloproteinase-
as new biomarkers of severity in multiple organ dysfunction
syndrome caused by trauma and infection, Molecular and
Cellular Biochemistry,vol.,no.-,pp.,.
[] M. Rahman, J. Roller, S. Zhang et al., Metalloproteinases
regulate CDL shedding from platelets and pulmonary
recruitment of neutrophils in abdominal sepsis, Inammation
Research,vol.,no.,pp.,.
... We have previously documented that moderate acute hepatic ischemia (30 min) followed by reperfusion (60 min) increases MMPs activity not only in the ischemic liver region but also in the lung, associated with histological damage in the liver, lung, and kidney [9]. Clinical studies have reported that, in patients with acute liver failure induced by I/R, the incidence of acute kidney injury (AKI) ranges from 40 to 85%, and up to 95% in liver transplantation [10]. ...
... We have already reported that liver I/R injury is associated with MMP activation with profound effects on tissue integrity [15]; this event also influences the function of many remote organs [9,16]. In the present study, we evaluated the MMP-9-dimer activity in the kidney cortex after hepatic ischemia (60 min) followed by reperfusion (120 min): a significant increase in MMP-9-dimer activity was found in the I/R group compared with sham-operated rats (Figure 1a). ...
... We have previously documented that moderate acute hepatic ischemia (30 min) followed by reperfusion (60 min) increases MMPs activity not only in the ischemic liver region but also in the lung, associated with histological damage in the liver, lung, and kidney [9]. Additionally, no significant difference in MMPs was observed in other distant organs such as the kidney and heart. ...
Article
Full-text available
We have previously demonstrated that the farnesoid X receptor (FXR) agonist obeticholic acid (OCA) protects the liver via downregulation of hepatic matrix metalloproteinases (MMPs) after ischemia/reperfusion (I/R), which can lead to multiorgan dysfunction. The present study investigated the capacity of OCA to modulate MMPs in distant organs such as the kidney. Male Wistar rats were dosed orally with 10 mg/kg/day of OCA (5 days) and were subjected to 60-min partial hepatic ischemia. After 120-min reperfusion, kidney biopsies (cortex and medulla) and blood samples were collected. Serum creatinine, kidney MMP-2, and MMP-9-dimer, tissue inhibitors of MMPs (TIMP-1, TIMP-2), RECK, TNF-alpha, and IL-6 were monitored. MMP-9-dimer activity in the kidney cortex and medulla increased after hepatic I/R and a reduction was detected in OCA-treated I/R rats. Although not significantly, MMP-2 activity decreased in the cortex of OCA-treated I/R rats. TIMPs and RECK levels showed no significant differences among all groups considered. Serum creatinine increased after I/R and a reduction was detected in OCA-treated I/R rats. The same trend occurred for tissue TNF-alpha and IL-6. Although the underlying mechanisms need further investigation, this is the first study showing, in the kidney, beneficial effects of OCA by reducing TNF-alpha-mediated expression of MMPs after liver I/R.
... In previous studies, thickening in the interalveolar areas of the lungs, edema, congestion, mononuclear cell infiltration [28], inflammation, hemorrhage, atelectasis, hyaline membrane formation, and bronchiole epithelial degeneration have been described [26,27,29]. In our study, apoptotic cell death in the lung tissue and capillary congestion in the interalveolar septum were detected. ...
... Besides, renal tubular necrosis, decrease in glomerular filtration rate, inflammation, and apoptotic cell death by proinflammatory cytokines occur [6], urea and creatinine levels increase [30]. In addition, dilatation of the renal tubules, platelet accumulation in the glomeruli, increased fibrinogen, disappearance of the striated border, and epithelial cell degeneration have been reported [29]. In our study, significant congestion in intertubular capillaries in the renal cortex, shedding of proximal tubule epithelial cells, dilatation of tubules, and apoptosis were observed in the IR group. ...
Article
Full-text available
Liver ischemia/reperfusion (IR) often affects distant organs, such as small intestine, kidney, and lung. Coriandrum sativum (CS) has an antioxidant and anti-inflammatory effect on liver damage. The aim of this study was to investigate the anti-inflammatory and antiapoptotic effects of CS extract on small intestine, lung, and kidney after the liver IR injury. Small intestine, lung, and kidney tissues were evaluated and scored in terms of cell degeneration, inflammation, and congestion, as well as caspase-3 (Cas-3) and cluster of differentiation 31 (CD31) immunostainings were carried out. Renal enzymes, creatinine and urea levels were measured biochemically in serum. After IR, a decrease in villi size, diffuse degeneration, epithelial cell shedding and extensive congestion in the capillaries were observed. Meanwhile, the number of degenerated villi and congestion decreased in the IR+CS group. Due to IR, increased congestion was detected in the interalveolar septum of the lungs and in the capillaries between the kidney tubules. It was also observed that the positively stained cells with Cas-3 and CD31 were increased in the lung, kidney, and small intestine tissues of the IR group, and decreased in the IR+CS group. Kidney enzymes, urea and creatinine levels were significantly increased in the IR group and decreased in the IR+CS group. In conclusion, it was observed that liver IR caused changes in distant organs, especially in the small intestine, lung, and kidneys. Damaging effects of IR as well as apoptosis and inflammation were found to be decreased in the groups treated with CS.
... Progression of NAFLD to NASH is also modulated by TNF-alpha. It is known that SCD1-deficiency attenuates the induction of TNF-alpha [49], involved in the control of MMPs [50]; in addition, silencing of TNF-alpha in myeloid cells is able to prevent the development of NASH [51]. The current study supports a strong association between desaturases, TNF-alpha and MMP-2 in rat models of NAFLD. ...
Article
Full-text available
We investigated changes in fatty acid desaturases, D5D, D6D, D9-16D and D9-18D, and their relationship with oxidative stress, matrix metalloproteinases (MMPs) and serum TNF-alpha in two rat models of non-alcoholic fatty liver disease NAFLD. Eight-week-old male Wistar rats fed for 3 weeks with methionine-choline–deficient (MCD) diet and eleven-week-old Obese male Zucker rats were used. Serum levels of hepatic enzymes and TNF-alpha were quantified. Hepatic oxidative stress (ROS, TBARS and GSH content) and MMP-2 and MMP-9 (protein expression and activity) were evaluated. Liver fatty acid profiling, performed by GC-MS, was used for the quantification of desaturase activities. Higher D5D and D9-16D were found in Obese Zucker rats as well as an increase in D9-18D in MCD rats. D6D was found only in MCD rats. A negative correlation between D5D and D9-16D versus TBARS, ROS and TNF-alpha and a positive correlation with GSH were shown in fatty livers besides a positive correlation between D9-18D versus TBARS, ROS and TNF-alpha and a negative correlation with GSH. A positive correlation between D5D or D9-16D or D9-18D versus protein expression and the activity of MMP-2 were found. NAFLD animal models showed comparable serum enzymes. These results reinforce and extend findings on the identification of therapeutic targets able to counteract NAFLD disorder.
... In the four groups of rats in the study, at seven hours following hepatic I/R, myeloperoxidase (MPO) activity in the liver homogenates was determined using the assay described by Palladini and colleagues [11]. Briefly, 200 mg of fresh liver tissue in 1 ml PBS was homogenized and resuspended in PBS (1 ml) containing 0.5% hexa-1,6-bis-decyltrimethylammonium bromide (HTAB) (Sigma-Aldrich, St. Louis, MO, USA) and 5 mM ethylenediaminetetraacetic acid (EDTA) (Sigma-Aldrich, St. Louis, MO, USA). ...
Article
Full-text available
Background Several clinical conditions can cause hepatic ischemia/reperfusion (I/R) injury. This study aimed to determine the mechanism of the protective effect of hyperbaric oxygen preconditioning (HBO2P) on hepatic ischemia/reperfusion (I/R) injury in a rat model, and to investigate the effects on HBO2P and I/R injury of blocking HSP70 using antibody (Ab) pretreatment. Material/Methods Male Sprague-Dawley rats underwent HBO2P for 60 min at 2.0 atmosphere absolute (ATA) pressure for five consecutive days before surgical hepatic I/R injury, performed by clamping the portal vein and hepatic lobe. Four groups studied included: the non-HBO2P+ non-I/R group, which underwent sham surgery (N=10); the non-HBO2P + I/R group (N=10); the HBO2P + I/R group (N=10); and the HBO2P + HSP70-Ab + I/R group (N=10) received one dose of HSP70 antibody one day before hepatic I/R injury. Serum lactate dehydrogenase (LDH), aspartate aminotransferase (AST), alanine aminotransferase (ALT), and pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), and hepatic malondialdehyde (MDA) and myeloperoxidase (MPO) were measured biochemically. Rat liver tissues were examined histologically. Results In rats with hepatic I/R injury without HSP70 antibody pre-treatment, HBO2P significantly reduced hepatic injury and levels of LDH, AST, ALT, TNF-α, IL-6, MDA, and MPO levels; in comparison, the group pre-treated with an antibody to inhibit HSP70 (the HBO2P + HSP70-Ab + I/R group) showed significant reversal of the beneficial effects of HBO2P on hepatic I/R injury (p<0.05). Conclusions In a rat model of hepatic I/R injury with HBO2P, HSP70 reduced hepatic inflammatory and oxidative damage.
... Besides, a decreased level of serum lactate was found in irisin-treated mice in contrast to vehicle-treated mice (Fig. 2G), indicating that irisin improves the metabolism condition after hepatic I/R. Hepatic I/R is often associated with lung injury [19,20]. As shown in Fig. 2H and I, vehicle-treated mice had marked increase in lung injury scores at 24 h after hepatic I/R. ...
Article
Full-text available
Current management of liver ischemia-reperfusion (I/R) injury is mainly based on supportive care and no specific treatment is available. Irisin, a recently identified hormone, plays pivotal roles in energy expenditure and oxidative metabolism; however, it remains unknown whether irisin has any protective effects on hepatic I/R injury. In this study, we found that serum and liver irisin levels were markedly decreased at 24 h after hepatic I/R. Treatment with exogenous irisin improved liver function, reduced liver necrosis and cell apoptosis, and relieved inflammatory response after hepatic I/R. Meanwhile, exogenous irisin markedly inhibited mitochondrial fission related protein dynamin related protein 1 (drp-1) and fission 1 (Fis-1) expression in hepatic I/R. Additionally, treatment with exogenous irisin increased mitochondrial content and increased mitochondrial biogenesis related peroxisome proliferative activated receptor-γ (PPARγ) co-activator 1α (PGC-1α) and mitochondrial transcription factor (TFAM) expression. Furthermore, irisin decreased oxidative stress by upregulating uncoupling proteins (UCP) 2 expression in hepatic I/R. The results reveal that treatment with exogenous irisin alleviated hepatic I/R injury by restraining mitochondrial fission, promoting mitochondrial biogenesis and relieving oxidative stress. Irisin treatment appears to be a novel and promising therapeutic approach for hepatic I/R injury.
... By inducing a cross-linking reaction between proteins and their respective primary amino groups, MDA leads to the destruction of cell function. Therefore, by measuring the serum content of MDA, the degree of cell membrane damage can be indirectly determined (27). In the present study, the MDA increase was significantly lower in the MT group, as compared with that of the IRI group, thus suggesting that MT may promote the degradation of oxygen free radicals into non-toxic or low toxic substances, and thus enhance antioxidant capacity. ...
Article
Full-text available
The present study aimed to investigate the effects of melatonin (MT) on liver function and lipid peroxidation following hepatic ischemia-reperfusion injury (IRI). A total of 66 male Sprague-Dawley rats were randomly assigned into three groups: Normal control (N) group, ischemia-reperfusion (IR) group and the MT-treated group. A hepatic IRI model was developed by blocking the first porta hepatis, and subsequently restoring hepatic blood inflow after 35 min. Following reperfusion, changes in the levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione (GSH) were detected by a chemical method at various time points. In the MT group, the MDA levels were significantly reduced (P<0.05) at all time points, as compared with the IR group. Furthermore, SOD activity was significantly increased (P<0.05) in the MT group, as compared with the IR group at all time points; and the levels of GSH in the MT group were significantly higher (P<0.05) than those of the IR group at 2, 4, and 8 h post-reperfusion. The levels of ALT, AST and LDH were significantly reduced in the MT group at each time point, as compared with that of the IR group (P<0.05). In conclusion, MT exhibits potent antioxidant properties that may create favorable conditions for the recovery of liver function following IRI. © 2016, Experimental and Therapeutic Medicine. All rights reserved.
Article
Full-text available
Oxymatrine (OMT) is the primary active component of Sophora flavescens Ait., and is widely used for the treatment of diabetic complications. The present study aimed to investigate the effects of OMT on acute lung injury (ALI) in diabetic rats subjected to myocardial ischemia/reperfusion (I/R). ALI in a myocardial I/R model was established in streptozocin‑induced diabetic rats. Enzyme‑linked immunosorbent assays were used to evaluate the levels of creatine kinase isoenzyme MB and lactate dehydrogenase, and the inflammatory response was assessed via leukocyte counts and the levels of tumor necrosis factor (TNF)‑α, interleukin (IL)‑6 and IL‑8 in the bronchoalveolar lavage (BAL) fluid. Hematoxylin and eosin staining was used to determine pathological changes to the lung tissue, and the autophagy‑related proteins LC‑3II/LC‑3I, Beclin‑1, autophagy protein 5 (Atg5) and p62 were detected by western blotting. Diabetic rats subjected to myocardial I/R showed increased levels of ALI with a higher lung injury score and WET/DRY ratio, and lower partial pressure of oxygen. This was accompanied by aberrant autophagy, indicated by an increased LC‑3II/LC‑3I ratio, decreased p62 expression levels, increased Atg5 and beclin‑1 expression levels, decreased superoxide dismutase activity and increased 15‑F2t‑isoprostane formation in lung tissues, as well as increased levels of leukocytes, TNF‑α, IL‑6 and IL‑8 in the BAL fluid. Administration of the autophagy inducer rapamycin significantly accelerated these alterations, while the autophagy inhibitor 3‑Methyladenine exerted the opposite effects. These results indicated that diabetic lungs are more vulnerable to myocardial I/R, which was associated with aberrant autophagy. Furthermore, oxymatrine was observed to reverse and alleviate ALI in diabetic rats with myocardial I/R in a concentration‑dependent manner, the mechanism of which may be associated with the inhibition of autophagy.
Article
Lung injury is a serious condition encountered following hepatic ischemia/reperfusion (IR). This study aimed to explore whether a dipeptidyl peptidase-4 inhibitor agent vildagliptin (V) could alleviate the lung injury caused by hepatic IR in a rat model and if so elucidate its molecular protective mechanism. Three groups of rats were used. Sham group: received normal saline and exposed to a sham operation, IR group: received normal saline and subjected to the operation of hepatic I (45 min)/ R (180 min), V+IR group: received for 10 days intraperitoneal injection of V (10 mg/kg/day). After reperfusion, liver and lung were collected for biochemical and histological evaluation. Hepatic IR exhibited significant elevation in serum alanine aminotransferase (ALT), alkaline phosphatase (ALP), and lactate dehydrogenase (LDH) enzyme levels, serum and lung malondialdehyde (MDA) and tumor necrosis factor-alpha (TNF-α) in addition to lung nitric oxide (NO) levels, hypoxia-inducible factor 1-alpha (HIF-1α) mRNA and protein levels, hepatocyte growth factor (HGF) mRNA expression, and inducible nitric oxide synthase (iNOS) mRNA and protein expressions in lung tissue along with a marked reduction in the serum and lung content of catalase in comparison to the sham group. Moreover, liver and lung injury in the IR group was detected by histopathological examination. Vildagliptin ameliorated markedly the biochemical changes as well as liver and lung architecture in comparison to the IR group. Vildagliptin mitigated the induced lung injury by hepatic IR via suppression of oxidative stress markers, pro-inflammatory cytokine TNF-α as well as the HIF1-α/iNOS/HGF expressions in lung tissue.
Article
Full-text available
Ischemia–reperfusion (IR) injury is a kind of injury resulting from the restoration of the blood supply after blood vessel closure during liver transplantation and is the main cause of graft failure. The pathophysiological mechanisms of hepatic IR include a variety of oxidative stress responses. Hepatic IR is characterized by ischemia and hypoxia inducing oxidative stress, immune response and apoptosis. Fat-denatured livers are also used as donors due to the lack of liver donors. Fatty liver is less tolerant to IR than normal liver. Heme oxygenase (HO) is an enzyme that breaks down hemoglobin to bilirubin, ferrous iron and carbon monoxide (CO). Inducible HO subtype HO-1 is an important protective molecule in mammalian cells used to improve acute and chronic liver injury owing to its characteristic anti-inflammatory and anti-apoptotic qualities. HO-1 degrades heme, and its reaction product CO has been shown to reduce hepatic IR injury and increase the survival rate of grafts. As an induced form of HO, HO-1 also exerts a protective effect against liver IR injury and may be useful as a new strategy of ameliorating this kind of damage. This review summarizes the protective effects of HO-1 in liver IR injury, especially in fatty liver.
Article
Objective: To describe new technologies (biomarkers and tests) used to assess and monitor the severity and progression of multiple organ dysfunction syndrome in children as discussed as part of the Eunice Kennedy Shriver National Institute of Child Health and Human Development MODS Workshop (March 26-27, 2015). Data sources: Literature review, research data, and expert opinion. Study selection: Not applicable. Data extraction: Moderated by an experienced expert from the field, investigators developing and assessing new technologies to improve the care and understanding of critical illness presented their research and the relevant literature. Data synthesis: Summary of presentations and discussion supported and supplemented by relevant literature. Conclusions: There are many innovative tools and techniques with the potential application for the assessment and monitoring of severity of multiple organ dysfunction syndrome. If the reliability and added value of these candidate technologies can be established, they hold promise to enhance the understanding, monitoring, and perhaps, treatment of multiple organ dysfunction syndrome in children.
Article
Full-text available
Studies assessing the effects of partial-hepatic ischemia/reperfusion (I/R) injury focused on the damage to the ischemic-lobe, whereas few data are available on non-ischemic lobe. This study investigated whether acute liver I/R does affect non-ischemic lobe function via modulation of extracellular matrix remodeling. Male Sprague-Dawley rats underwent left lateral- and median-lobe ischemia for 30 min and reperfusion for 60 min or sham operation. After reperfusion, blood samples and hepatic biopsies from both the ischemic (left-lobe, LL) and the non-ischemic lobe (right-lobe, RL) were collected. Serum hepatic enzymes and TNF-alpha, tissue matrix metalloproteinases (MMP-2, MMP-9), liver morphology, malondialdehyde (MDA), and myeloperoxidase (MPO) were evaluated. Liver I/R injury was confirmed by altered increased hepatic enzymes and TNF-alpha. I/R induced an altered morphology and an increase in MMP-2 and MMP-9 activity not only in left-ischemic lobe (LL) but also in the right-non-ischemic (RL) lobe. A lobar difference was detected for MDA formation and MPO activity in both sham and I/R submitted rats, with higher levels in the left lobe for both groups. This study indicates that an increase in MMPs, which may be TNF-alpha-mediated, occurs in both the ischemic- and the non-ischemic lobes; the heterogeneous lobe concentrations of MDA and MPO suggest that the random sampling of liver tissue should be avoided.
Article
Full-text available
Cardiovascular dysfunction frequently occurs after major vascular surgery or liver transplantation. To evaluate the effects on myocardial activity of vasoactive agents released from ischemic-reperfused liver. Isolated rat livers were perfused with Krebs-Henseleit solution (KH), propranolol 10(-5) M, losartan 2x10(-5) M and indomethacin 10(-5) M, then made globally ischemic for 120 min (37 degrees C) and reperfused. Isolated hearts from other rats were stabilized with KH and reperfused for 15 min with the perfusate exiting the livers. Livers were disconnected, and the hearts continued to be recirculated with the accumulated liver and heart effluent for an additional 50 min. Enzyme leakage, different vasoactive substances, left ventricular developed pressure (LVP) and coronary flow were measured during the experimental protocol. Hepatic release of adrenaline, noradrenaline, angiotensin II, prostaglandin E(2) and thromboxane B(2) was significantly increased in the liver effluent following ischemia. When this effluent was directed to the heart, LVP was significantly raised in the first 10 min of reperfusion (137+/-5%) followed by marked decreased (46+/-6%) during the following 65 min of myocardial reperfusion. In the ischemic-reperfused drug-treated groups, the initial positive effect on LVP was milder than in controls (propranolol 112+/-12%, losartan 111+/-11%, indomethacin 113+/-9%) and the final LVP was lower (propranolol 29+/-6%, losartan 27+/-7% [P<0.05 versus ischemic control], indomethacin 46 +/-12%). During the initial phase of reperfusion, vasoactive substances released in the hepatic effluent potentiated LVP of the hearts exposed to this effluent. When the three inhibitory drugs were added to KH, this initial augmentation was not sustained. Propranolol and losartan, but not indomethacin, further depressed LVP. Vasoactive substances released from ischemic reperfused livers directly influenced heart function.
Article
Although matrix metalloproteinases (MMPs) and their specific inhibitors (TIMPs) play an essential role in liver injury associated with tissue remodeling, the cellular origin of MMPs/TMPs within the liver remains to be clarified. Different liver cell populations were analysed with respect to their expression by reverse transcription-polymerase chain reaction, Northern blot analysis and zymography. MMP and TIMP coding transcripts were detectable in all liver cell types by reverse transcription-polymerase chain reaction; however, the cellular expression levels were markedly different as assessed by Northern blot analysis. Gelatinase-B was predominantly expressed in Kupffer cells, gelatinase-A in hepatic stellate cells and rat liver myofibroblasts and stromelysins-1, -2 as well as collagenase in hepatic stellate cells. Membrane type-1 MMP (MMP-14) was found in significant amounts in all liver cells. TIMP-1 coding m-RNAs were present mainly in hepatic stellate cells and rat liver myofibroblasts, TIMP-2 additionally in Kupffer cells, while TIMP-3 expression was detectable only in hepatocytes. During in vitro activation of hepatic stellate cells, MMP expression was mostly downregulated, while TIMP expression was enhanced, thereby providing an explanation for matrix accumulation co-localised with these cells during chronic liver injury. In general, TNF-alpha stimulated both MMP and TIMP expression of hepatic stellate cells, while TGF-beta1 induced TIMP expression only. Collectively these data demonstrate that all resident liver cells are involved in matrix degradation to some extent and that hepatic stellate cells play an important role in matrix breakdown in addition to matrix synthesis. The cytokine-specific regulation of MMP/TIMP expression in hepatic stellate cells suggests that the initial matrix breakdown following liver injury might be enhanced by TNF-alpha, while diminished matrix degradation during chronic tissue injury might be due to the action of TGF-beta1 through TIMP induction.
Article
Platelets promote sepsis-induced activation of neutrophils via secretion of CD40L. However, the mechanism regulating the release of platelet-derived CD40L is not known. We hypothesized that matrix metalloproteinases (MMPs) might regulate shedding of platelet-expressed CD40L and neutrophil activation in sepsis. Wild-type C57BL/6 mice were subjected to cecal ligation and puncture (CLP). Animals were pretreated with a broad-range MMP inhibitor, GM6001, prior to CLP induction. Edema formation, CXC chemokine and myeloperoxidase (MPO) levels and bronchoalveolar neutrophils in the lung as well as plasma levels of CD40L were quantified. Flow cytometry was used to determine expression of Mac-1 on neutrophils and CD40L on platelets. Intravital fluorescence microscopy was used to analyze leukocyte-endothelial cell interactions in the pulmonary microcirculation. The MMP inhibitor reduced sepsis-induced release of CD40L and maintained normal levels of CD40L on platelets. Inhibition of MMP decreased CLP-induced neutrophil expression of Mac-1, formation of CXC chemokines and edema as well as neutrophil infiltration in the lung. Intravital fluorescence microscopy revealed that the MMP inhibitor attenuated leukocyte adhesion in venules whereas capillary trapping of leukocytes was not affected by MMP inhibition. We describe a novel role of metalloproteinases in regulating platelet-dependent activation and infiltration of neutrophils in septic lung injury which might be related to controlling CD40L shedding from platelets. We conclude that targeting metalloproteinases may be a useful strategy for limiting acute lung injury in abdominal sepsis.
Article
Multiple organ dysfunction syndrome (MODS) is an important cause of morbidity and mortality in intensive care unit. A severe insult in the form of infection or trauma primes the host immune system so that a subsequent, relatively trivial insult produces systemic inflammation response syndrome, which can lead to MODS and death. Matrix metalloproteinase-9 (MMP-9) is stored in the tertiary granules of polymorphonuclear leukocytes. These cells are key effectors in acute inflammatory diseases, such as sepsis and MODS. Endotoxin leads to rapid release of MMP-9 from these granules in vitro and in vivo. However, the role of this enzyme in MODS, and whether it is associated with organ injury at the early stage of MODS remains unclear. This present work may study role of MMP-9 with the MODS rats that caused by trauma and infection and investigate the mechanism of organ injury at the early stage of MODS. Here, we developed a rat model for MODS caused by trauma and infection and analyzed the dynamic level of MMP-9 and determined the relationship between MMP-9 level and early phase of organ injury in MODS rat. The histological changes in pulmonary, renal, and hepatic tissue were observed by light microscope. The expressions of plasma MMP-9 proteins were detected by an enzyme linked immunosorbent assay and its levels in the pulmonary, renal, and hepatic tissue were detected by using immunohistochemistry, respectively. The results indicated that there were no significant improvements in histopathology of rats in control group. However, the pulmonary, renal, and hepatic damage were serious in MODS groups. The concentration of MMP-9 in plasma and tissues of MODS rats increased markedly at the early stage and were higher than that of the control group. Moreover, the MMP-9 level in plasma positively correlated with the levels of pulmonary, renal, and hepatic tissue. This study clearly shows that MMP-9 is good biomarker to predict the severity of injury organ at the early phase of MODS.
Article
Hepatic ischemia followed by reperfusion (IR) results in mild to severe remote organ injury. Oxidative stress and nitric oxide (NO) seem to be involved in the IR injury. Our aim was to investigate the effects of liver I/R on hepatic function and lipid peroxidation, leukocyte infiltration and NO synthase (NOS) immunostaining in the lung and the kidney. We randomized 24 male Wistar rats into 3 groups: 1) control; 2) 60 minutes of partial (70%) liver I and 2 hours of global liver R; and 3) 60 minutes of partial (70%) liver I and 6 hours of global liver R. Groups 2 and 3 showed significant increases in plasma alanine and aspartate aminotransferase levels and in tissue malondialdehyde and myeloperoxidase contents. In the kidney, positive endothelial NOS (eNOS) staining was significantly decreased in group 3 compared with group 1. However, staining for inducible NOS (iNOS) and neuronal NOS (nNOS) did not differ among the groups. In the lung, the staining for eNOS and iNOS did not show significant differences among the groups; no positive nNOS staining was observed in any group. These results suggested that partial liver I followed by global liver R induced liver, kidney, and lung injuries characterized by neutrophil sequestration and increased oxidative stress. In addition, we supposed that the reduced NO formation via eNOS may be implicated in the moderate impairment of renal function, observed by others at 24 hours after liver I/R.
Article
Matrix metalloproteinase 9 (MMP-9) is a critical mediator of leukocyte migration in hepatic ischemia/reperfusion (I/R) injury. To test the relevance of inducible nitric oxide synthase (iNOS) expression on the regulation of MMP-9 activity in liver I/R injury, our experiments included both iNOS-deficient mice and mice treated with ONO-1714, a specific iNOS inhibitor. The inability of iNOS-deficient mice to generate iNOS-derived nitric oxide (NO) profoundly inhibited MMP-9 activity and depressed leukocyte migration in livers after I/R injury. While macrophages expressed both iNOS and MMP-9 in damaged wild-type livers, neutrophils expressed MMP-9 and were virtually negative for iNOS; however, exposure of isolated murine neutrophils and macrophages to exogenous NO increased MMP-9 activity in both cell types, suggesting that NO may activate MMP-9 in leukocytes by either autocrine or paracrine mechanisms. Furthermore, macrophage NO production through the induction of iNOS was capable of promoting neutrophil transmigration across fibronectin in a MMP-9-dependent manner. iNOS expression in liver I/R injury was also linked to liver apoptosis, which was reduced in the absence of MMP-9. These results suggest that MMP-9 activity induced by iNOS-derived NO may also lead to detachment of hepatocytes from the extracellular matrix and cell death, in addition to regulating leukocyte migration across extracellular matrix barriers. These data provide evidence for a novel mechanism by which MMP-9 can mediate iNOS-induced liver I/R injury.