ArticlePDF AvailableLiterature Review

Molecular pathways in protecting the liver from ischaemia/reperfusion injury: a 2015 update

Authors:

Abstract and Figures

Ischaemia/reperfusion injury is an important cause of liver damage during surgical procedures such as hepatic resection and liver transplantation, and represents the main cause of graft dysfunction post-transplantation. Molecular processes occurring during hepatic ischaemia/reperfusion are diverse, and continuously include new and complex mechanisms. The present review aims to summarize the newest concepts and hypotheses regarding the pathophysiology of liver ischaemia/reperfusion, making clear distinction between situations of cold and warm ischaemia. Moreover, the most updated therapeutic strategies including pharmacological, genetic and surgical interventions, as well as some of the scientific controversies in the field are described.
Content may be subject to copyright.
Clinical Science (2015) 129, 345–362 doi: 10.1042/CS20150223
Molecular pathways in protecting the liver from
ischaemia/reperfusion injury: a 2015 update
Jordi Gracia-Sancho,Aran
´
ı Casillas-Ram´
ırez†‡ and Carmen Peralta†
Barcelona Hepatic Hemodynamic Laboratory, Institut d’Investigacions Biom`
ediques August Pi i Sunyer (IDIBAPS), Centro de Investigaci´
on Biom´
edica
en Red en Enfermedades Hep´
aticas y Digestivas (CIBEREHD), Barcelona, Spain
†Institut d’Investigacions Biom`
ediques August Pi i Sunyer (IDIBAPS), Centro de Investigaci´
on Biom´
edica en Red en Enfermedades Hep´
aticas y
Digestivas (CIBEREHD), Barcelona, Spain
‡Hospital Regional de Alta Especialidad de Ciudad Victoria “Bicentenario 2010”, Cd. Victoria, Mexico
Abstract
Ischaemia/reperfusion injury is an important cause of liver damage during surgical procedures such as hepatic
resection and liver transplantation, and represents the main cause of graft dysfunction post-transplantation.
Molecular processes occurring during hepatic ischaemia/reperfusion are diverse, and continuously include new and
complex mechanisms. The present review aims to summarize the newest concepts and hypotheses regarding the
pathophysiology of liver ischaemia/reperfusion, making clear distinction between situations of cold and warm
ischaemia. Moreover, the most updated therapeutic strategies including pharmacological, genetic and surgical
interventions, as well as some of the scientific controversies in the field are described.
Key words: cold storage, hepatocytes, liver microcirculation, liver sinusoidal endothelial cells, LSEC, warm ischaemia.
INTRODUCTION
The pathophysiology of I/R (ischaemia/reperfusion) injury has
been comprehensively studied, and reviewed, by several authors
in the past [1,2]. One might suggest that similar underlying events
happen in all types of clinical situations where I/R occurs, and that
the mediators and mechanisms involved are solidly established.
Nevertheless, the more we discover, the less we certainly know
about I/R injury, and thus the further away seems to be its solution
at the bedside.
The complexity in mechanisms and cellular components im-
plicated in I/R has led to several controversies, and even discrep-
ancies, in our understanding of this pathology. Carefully reading
the literature, we hypothesize that disagreements in I/R mechan-
isms, and therefore in therapeutic targets, were probably due to
slight (or evident) differences in experimental procedures. The
present mini-review aims to summarize some of the most recently
described cellular and molecular mechanisms underlying hepatic
I/R injury, as well as derived therapeutic options, making a patent
distinction between those results obtained in experimental mod-
els of cold and warm ischaemia. In addition, some of the most
Abbreviations: B7-H1, B7 homologue 1; DAMP, damage-associated molecular pattern; eNOS, endothelial nitric oxide synthase; HDmiR, hepatocyte-derived miRNA; I/R,
ischaemia/reperfusion; IRF, interferon-regulatory factor; KLF2, Kr ¨
uppel-like factor 2; LSEC, liver sinusoidal endothelial cells; MAPK, mitogen-activated protein kinase; MMP, matrix
metalloproteinase; NMP, nor mothermic machine perfusion; Nrf2, nuclear factor-erythroid 2-related factor 2; PAMP, pathogen-associated molecular pattern; PH, par tial hepatectomy;
RBP4, retinol-binding protein 4; TLR, Toll-like receptor.
Correspondence: Dr Jordi Gracia-Sancho (email jgracia@clinic.ub.es).
controversial (and long-lasting) observations in the field are also
discussed.
COLD STORAGE
Mechanisms of injury
Hepatocyte and sinusoidal injuries
Nowadays, it is accepted that hepatic endothelium damage occur-
ring during cold preservation represents the initial factor leading
to hepatic I/R injury, determining poor graft microcirculation,
platelet activation, persistent vasoconstriction, up-regulation of
adhesion molecules, cytokine release, oxidative stress, Kupffer
cell activation, neutrophil infiltration and hepatocyte death, thus
contributing to the development of primary non-function or im-
paired primary function after liver transplantation (Figure 1).
Indeed, and although hepatocyte function and viability might be
preserved under in vitro cold storage conditions up to 72 h, the
LSEC (liver sinusoidal endothelial cells) phenotype is rapidly
deregulated during cold storage, becoming activated after 6 h,
and highly apoptotic and pro-inflammatory thereafter [3,4]. In
C
The Authors Journal compilation C
2015 Biochemical Society 345
Clinical Science www.clinsci.org
J. Gracia-Sancho, A. Casillas-Ram´
ırez and C. Peralta
Figure 1 Underlying mechanisms of hepatic cold ischaemia and warm reperfusion injury
Pathways recently described are coloured, whereas classic mechanisms are summarized in grey. DAMPs, damage-as-
sociated molecular patterns; eNOS, endothelial nitric oxide synthase; ET, endothelin; IL, interleukin; IRF-1, interferon
regulatory factor-1; KC, Kupffer cells; KLF2, Kr¨
uppel-like factor 2; LSEC, liver sinusoidal endothelial cells; Nrf2, nuclear
factor-erythroid 2-related factor 2; PAMPs, pathogen-associated molecular patterns; ROS, reactive oxygen species; TLR4,
Toll-like receptor 4; TM, thrombomodulin; TNF-α, tumour necrosis factor α.
fact, experimental studies of liver preservation for transplant-
ation showed acute endothelial dysfunction development after
16 h of cold storage, associated with significant hepatocellular
injury and death [4]. Considering the intimate cellular cross-talk
between LSEC and hepatocytes, it is very possible that LSEC
injury due to cold storage may negatively affect hepatocyte viab-
ility, and vice versa.
Liver sinusoidal cells
It is well known that LSEC of cold-stored livers are seriously
compromised during cold storage and transplantation. Indeed,
rapid repopulation of LSEC is observed after transplantation
in association with up-regulation of diverse pro-angiogenic and
endothelial-survival mechanisms. In this setting, potential re-
cruitment and engraftment of bone-marrow-derived endothelial
precursors may contribute to revascularization sites [5]. In the last
few years, several studies have revealed new signalling mechan-
isms involved in cold ischaemia injury where sinusoidal cells
play a key role. The lack of biomechanical stimuli occurring dur-
ing cold preservation for transplantation markedly deteriorates
the LSEC protective phenotype by down-regulating the expres-
sion of the transcription factor KLF2 (Kr¨
uppel-like factor 2),
which orchestrates the transcription of a variety of protective
genes including the eNOS (endothelial nitric oxide synthase),
the anti-thrombotic molecule thrombomodulin or the antioxid-
ant transcription factor Nrf2 (nuclear factor-erythroid 2-related
factor 2) [4,6].
Concomitantly to LSEC deregulation, Kupffer cells suffer
from a profound activation process that is promoted by the release
of DAMPs (damage-associated molecular patterns) from neigh-
bouring necrotic hepatic cells and, under conditions of sepsis
or endotoxaemia, also PAMPs (pathogen-associated molecular
patterns). TLRs (Toll-like receptors) recognize both PAMPs and
DAMPs, leading to activation of downstream signalling cascades.
Evidence from experimental studies of cold ischaemia and trans-
plantation indicates that an increase in TLR4 could be correlated
with hepatocellular damage [7]. Although this occurs in non-
steatotic livers, up-regulation of the TLR4 pathway is protective
in steatotic livers undergoing transplantation [8]. This divergent
role for TLR4 could be explained by differences in the pathogenic
mechanisms of I/R injury between steatotic and non-steatotic
livers.
Emerging mechanisms of injury
The autophagy machinery is activated under stress conditions,
such as transient hypoxia or starvation, to ensure cell survival
and limit cell death. Under experimental conditions of cold I/R
injury, it has been found that defective liver autophagy correlates
with liver damage [9]. In fact, strategies aimed at up-regulating
autophagy reduced the development of hepatocellular necrosis
346 C
The Authors Journal compilation C
2015 Biochemical Society
Underlying mechanisms of hepatic I/R injury
and improved graft survival; nevertheless, better characteriza-
tion of this process, focusing on each hepatic cell type, is still
necessary. In this regard, a recent communication by Biel and col-
leagues presented at the AASLD Liver Meeting 2014 (abstract
105) demonstrated that hepatocyte autophagy defect during I/R
leads to cell death, therefore proposing activation of hepatocyte
autophagy as a new therapeutic option.
Small non-coding RNAs, and particularly miRNAs, have
emerged as important genetic regulators of cellular processes,
including tissue injury and repair responses. Recently, it has been
demonstrated that the levels of the HDmiRs (hepatocyte-derived
miRNAs) miR-122,miR-148a and miR-194 are significantly el-
evated in serum from patients after liver transplantation, posit-
ively correlating with aminotransferase levels [10]. As experi-
mental studies have shown that miRNAs are feasible targets for
therapeutic interventions designed to minimize injury caused by
ischaemic insults, further studies targeting HDmiRs might elu-
cidate their real role in cold I/R injury.
IRF (interferon-regulatory factor)-1, a transcription factor
originally identified as a regulator of IFN (interferon) α/β,is
involved in many aspects of innate and adaptive immune re-
sponses. It has been described that IRF-1-mediated inflammation
after liver transplantation is involved in multiple mechanisms
that are vital to the cellular stress response, including produc-
tion of inflammatory mediators, activation of apoptotic pathways
and activation of MAPK (mitogen-activated protein kinase) path-
ways [11]. Importantly, antagonism or silencing of IRF-1 gene
expression may be a potential strategy to ameliorate liver damage
associated with cold preservation injury [12].
B7-H1 (B7 homologue 1) is a recently identified member
of the B7 family with important regulatory functions in cell-
mediated immune responses. In experimental models of liver
transplantation, B7-H1 is up-regulated in both parenchymal and
non-parenchymal cells ameliorating cold I/R injury. Indeed, B7-
H1 inhibition markedly worsens I/R injury through increased
infiltration and reduced apoptosis of CD8+T-cells in liver grafts
[13].
Finally, it is worth considering recent findings indicating that
brain death itself strongly reduces the tolerance of liver grafts
to cold I/R injury [14,15]. Indeed, donor brain death triggers a
systemic inflammatory response and increased immunogenicity
of the graft that could potentially lead to reduced organ func-
tion. Injurious effects of brain death are mainly mediated through
Kupffer cell activation, leading to TNFα(tumour necrosis factor
α) and TLR4 amplification. Considering that 80 % of organs used
in the clinic come from donors who suffered brain death [14], it
seems reasonable to include brain death as part of the protocol
in experimental studies investigating cold I/R injury. This pat-
ent difference in the protocol might positively influence the ratio
of therapeutic strategies that are successfully translated to the
bedside.
Controversies
Role of warm ischaemia (after cold) in hepatic injury
The injury process that begins during hypothermia is afterwards
fostered by the re-warming process during graft implantation.
Concretely, in the liver, when temperature and cell metabolism
ramps up, damage switches from the more cold-sensitive LSEC
to the warm ischaemia-sensitive hepatocytes [16]. In the light of
these findings, one might assume that normothermic preservation
[using NMP (normothermic machine perfusion)] would probably
be the most appropriate strategy to preserve grafts in the clinical
setting of liver transplantation. Indeed, it would avoid the trans-
ition of cold to warm ischaemia, maintaining the liver graft at a
single temperature from retrieval until implantation. The under-
lying principle of NMP is the combination of continuous circu-
lation of metabolic substrates for ATP regeneration and removal
of waste products, together with biomechanical stimulation of
the sinusoids. Accordingly, accumulating evidence demonstrates
the superiority of the more physiological approach of normother-
mia, in association with an oxygenated blood-based perfusion
solution, in comparison with cold storage procedures [17–19].
Furthermore, in situ normothermic regional perfusion of dona-
tion after cardiac death is gaining in popularity and has resulted
in a number of successful liver transplantations [20–24].
Therapeutic approaches
On the basis of the above-described mechanisms, a variety of
therapeutic strategies have been developed. As seen in Table 1, in
the last 2 years the development of strategies to protect marginal
liver grafts has predominated, thus indicating the concern about
the increase in waiting list times and the shortage of liver grafts
available for transplantation; also, considerable efforts have been
devoted to the improvement in perfusion machine techniques,
and the first strategies based on cell therapy against cold I/R
injury have been reported. Interestingly, most of these original
investigations devoted their efforts to molecular pathways and
pathophysiological events markedly different from those studied
in previous decades (i.e. note the switch from strategies broadly
targeting adhesion molecules to strategies specifically focused
on upstream events of inflammation, such as adipocytokine sig-
nalling). In addition, in the specific scenario of machine perfu-
sion, a significant part of work aimed to characterize the pos-
sible use of oxygenated perfusion solutions, thus challenging the
dogma of noxious effects of oxygen during reperfusion injury.
In fact, very recently, a new preservation modality has been de-
scribed that combines machine perfusion at subnormothermic
conditions with a new haemoglobin-based oxygen carrier solu-
tion, triggering regenerative and cell protective responses result-
ing in improved allograft function [25].
WARM ISCHAEMIA
Mechanisms of injury
Warm I/R injury is responsible for significant organ dysfunction
and failure after liver resection and haemorrhagic shock. Histor-
ically, it has been considered that hepatocytes are more sensitive
to warm I/R, whereas LSEC are more sensitive to cold I/R. How-
ever, limitation of injury to a particular cell type is rare under
pathophysiological conditions; indeed evidencesuggest profound
sinusoidal deregulation during warm I/R injury [26]. Similarly
to liver cold I/R injury, Kupffer cells play a key role in warm
C
The Authors Journal compilation C
2015 Biochemical Society 347
J. Gracia-Sancho, A. Casillas-Ram´
ırez and C. Peralta
Table 1 Therapeutic approaches against cold I/R injury
AMPK, AMP-activated protein kinase; Bcl-2, B-cell lymphoma 2; DCD, donations after cardiac death; eNOS, endothelial nitric oxide synthase; ER, endoplasmic reticulum; ERK,
extracellular-signal-regulated kinase; ET-1, endothelin 1; GABAR, γ-aminobutyric acid receptor; HO-1, haem oxygenase-1; IFN, interferon; IL, interleukin; iNOS, inducible nitric
oxide synthase; LDH, lactate dehydrogenase; LSEC, liver sinusoidal endothelial cells; MMP, matrix metalloproteinase; MSC-CM, mesenchymal stem cell-conditioned medium;
NHBD, non-heart-beating donors; NF-κB, nuclear factor κB; NKT, natural killer T-cell; NMP, normothermic machine perfusion; Nrf2, nuclear factor-erythroid 2-related factor 2; PI3K,
phosphoinositide 3-kinase; PGE1, prostaglandin E1; PPAR, peroxisome-proliferator-activated receptor; rMnSOD, recombinant MnSOD; SLC, sinusoidal lining cell; SOD, superoxide
dismutase; SOWP, short oxygenated warm perfusion; STAT, signal transducer and activator of transcription; TIM-1, T-cell immunoglobulin mucin 1; TLR, Toll-like receptor; TNF,
tumour necrosis factor; UW, University of Wisconsin; VEGF, vascular endothelial growth factor.
(a) Pharmacological strategies
Drug Species Experimental model
Cold ischaemia
time Effect
Adiponectin [40] (adipocytokine) Rat Liver transplantation (steatotic grafts) 6 h Hepatic injury; PI3K/Akt
ανβ6 Antibody [41] (ανβ6 integrin) Mouse Liver transplantation 12 h Progression of biliary fibrosis; liver function
Biliverdin [42] (green tetrapyrrolic bile pigment) Pig Ex vivo liver perfusion 19 h Hepatic injury, neutrophil infiltration and apoptosis; liver
function
Bortezomib [43] (26S proteasome) Rat Liver transplantation (steatotic grafts) 2 h Hepatic injury, NF-κB, MMP and pro-inflammatory cytokines
Cardiotrophin-1 [44] (cytokine) Pig Liver transplantation 4 h Hepatic injury, caspase 3, IL-1β, IL-6, TNFα, oxidative stress;
survival, Akt, ERK, STAT3
Edaravone [45] (neuroprotective agent) Pig Liver transplantation (NHBD liver grafts) 4 h Hepatic injury and SLC damage; survival
Fructose [46] (monosaccharide) Rat Ex vivo liver perfusion 26 h Hepatic injury; ATP
GABAR agonist [47] (γ-aminobutyric acid) Rat Split liver transplantation 2 h Hepatic injury, oxidative stress and apoptosis
Ketanserin [48] (serotonin 2A receptor antagonist) Rat Liver transplantation (DCD liver grafts) 4 h Hepatic injury, biliary fibrosis, liver serotonin and hidroxyproline;
biliary function
Magnesium [49] Human Living donor liver transplantation Not reported Hepatic injury; regulation of Th1-to-Th2 cytokine balance towards
Th2
5-Methylthioadenosine [50] (nucleoside) Rat Liver transplantation Not reported Hepatic injury, NF-κB and MAPK activation
N-acetylcysteine [51] (antioxidant) Human Liver transplantation 2–10 h Post-operative complications; survival
Perfluorodecalin [52] (perfluorocarbon) Rat Hypothermic perfusion (DCD liver grafts) 8 h Hepatic injury; survival
Resistin [40] (adipocytokine) Rat Liver transplantation (steatotic grafts) 6 h Hepatic injury; PI3K/Akt
rMnSOD [37] (antioxidant) Rat Ex vivo liver perfusion 16 h Hepatic injury and inflammation; hepatic microcirculation and
endothelial function
RMT1-10 [53] (TIM-1 blocker) Mice Liver transplantation 20 h Hepatic injury and neutrophil and macrophage activation; IL-4,
IL-10, IL-22, Bcl-2
Simvastatin [54] (vasoprotector) Rat Ex vivo liver perfusion (steatotic grafts) 16 h Hepatic injury, apoptosis and inflammation; hepatic
microcirculation and endothelial function
(b) Gene and cell therapy strategies
Target gene/cell therapy strategy Species Experimental model
Cold ischaemia
time Effect
Ad-hTERT [55] (human telomerase transcriptase) Rat Liver transplantation (aged grafts) 2 h Hepatic injury and apoptosis; telomerase activity
CD39/in myeloid dendritic cells [56] Mouse Liver transplantation 24 h Hepatic injury; pro-inflammator y cytokines
CD39-transgenic [57] (CD39-overexpression) Mouse Liver transplantation 18 h Hepatic injury, IL-6, CD4+T-cells and invariant NKT cells
348 C
The Authors Journal compilation C
2015 Biochemical Society
Underlying mechanisms of hepatic I/R injury
Table 1 Continued
(b) Gene and cell therapy strategies
Target gene/cell therapy strategy Species Experimental model
Cold ischaemia
time Effect
Keap1-deficient [58] (Keap1 hepatocyte-specific
knockout)
Mouse Liver transplantation 20 h Hepatic injury, oxidative stress and inflammation and apoptosis;
Nrf2, thioredoxin and PI3K/Akt
MSC-CM [59] (mesenchymal stem cells) Rat Reduced-size liver transplantation 55–65 min Activation of Kupffer cells and neutrophils, pro-inflammatory
cytokines and apoptosis; VEGF, MMP-9 and proliferating
hepatocytes and LSEC
(c) Additives for preservation solutions
Drug or additive Species Experimental model
Cold ischaemia
time Effect
Bortezomib in UW or IGL solution [60,61]
(proteasome inhibitor) Rat Ex vivo liver perfusion (steatotic grafts) 24 h Hepatic injury, oxidative stress, IL-1 and TNF-α;NO and AMPK
Desferrioxamine in UW solution [62] (bacterial
siderophore) Rat Ex vivo liver perfusion 20 h Hepatic injury
Melatonin and trimetazidine in IGL-1 solution [63]
(hormone and anti-ischaemic drug) Rat Ex vivo liver perfusion (steatotic grafts) 24 h Hepatic injury and ER stress; autophagy
rMnSOD in Celsior solution [37] (antioxidant) Human Ex vivo preservation 16 h Oxidative stress
SNO-HSA in UW solution [64] (S-nitrosated human
serum albumin) Rat Ex vivo liver perfusion 72 h Hepatic injury, LDH, oxidative stress and apoptosis
SOWP and PGE1in UW solution [65] (short
oxygenated warm perfusion and prostaglandin E1)Rat
Ex vivo liver perfusion (uncontrolled NHBD
grafts) 6 h Hepatic injury, necrosis and apoptosis
(d) Surgical and machine perfusion-based strategies
Strategy Species Experimental model/clinical setting
Cold ischaemia
time Effect
HOPE [66,67] (hypothermic oxygenated perfusion) Rat Liver transplantation (DCD liver grafts) 4 h
Hepatic injury, biliary injury, Kupffer and endothelial cell
activation and oxidative stress; survival
HOPE [68] (hypothermic oxygenated perfusion) Pig
Ex vivo liver perfusion model (DCD liver
grafts) 6 h Hepatic injury and endothelial and mitochondrial damage
IPC [69] (ischaemic pre-conditioning) Rat Liver transplantation 50 min
Hepatic injury and bacterial translocation; intestinal microbiota
and mucosal ultrastructure
NMP [70] (normothermic machine perfusion) Rat Liver transplantation (DCD liver grafts)
No cold
ischaemia
Organ stability over 5–6 h of perfusion; restoration of warm
ischaemic liver to a likely transplantable state after 2 h of perfusion
Machine perfusion [71] Pig Liver transplantation (DCD liver grafts) 2 h Hepatic injury; recovery and resuscitation of DCD liver grafts
RIPostC [72] (remote ischaemic post-conditioning) Human Living donor liver transplantation 76-83 min
Acute kidney injury; no improvements in postoperative liver graft
function or clinical outcomes
Subnormothermic machine perfusion combined with
haemoglobin-based oxygen carrier (HBOC) solution
[25] Pig Liver transplantation 9 h
Hepatic injury, IFNα,IFNγ,TNFα,IL-1β, IL-4; liver function,
survival
VSOP-NO [73,74] (venous systemic oxygen
persufflation using NO gas) Rat
Liver transplantation (small or steatotic
grafts) 3 h
Hepatic injury, iNOS and peroxinitrite; regeneration, eNOS
expression, viability and microcirculation
VSOP-NO [75] (venous systemic oxygen
persufflation using NO gas) Rat Liver transplantation (DCD liver grafts) 3 h
Hepatic injury, oxidative stress, TNFα, IL-6, eNOS, ET-1,
hepatocyte and LSEC damage
C
The Authors Journal compilation C
2015 Biochemical Society 349
J. Gracia-Sancho, A. Casillas-Ram´
ırez and C. Peralta
I/R injury being a source of many pro-inflammatory mediators.
Neutrophils are also activated and recruited into the liver, where
several factors and conditions determine that neutrophils migrate
out of the sinusoids and affect parenchymal cells [26].
A central underlying mechanism for parenchymal injury due
to warm I/R injury is the impairment of mitochondria. Interest-
ingly, recent data indicate that cardiolipin (diphosphatidylgly-
cerol), a phospholipid required for efficient mitochondrial func-
tion, may play an essential role in such injury. Indeed, warm I/R
causes a significant decrease in hepatic cardiolipin content that,
together with an increase in its oxidized form, impairs mitochon-
drial function, altogether worsening liver injury [27]. Strategies
that support cardiolipin synthesis, or prevent its oxidation, may
be beneficial to reduce I/R injury in the liver.
Emerging evidence reveals the role of acetylation in funda-
mental biological processes. Protein acetylation by histone acet-
yltransferases enhances gene expression via relaxation of chro-
matin and transcription activation. Acetylation homoeostasis is
often disrupted in several pathologies. Indeed, warm hepatic I/R
injury is accompanied by a significant reduction in histone acet-
ylation in the liver, suggesting that histone acetylation may play
an important role in I/R injury representing a new therapeutic
strategy to ameliorate this pathology [28].
MMP (matrix metalloproteinase)-9, an inducible gelatinase,
is emerging as a central mediator of leucocyte trafficking into
inflamed tissues. In a mouse model of steatotic liver undergo-
ing warm I/R injury, it was demonstrated that MMP-9 activ-
ity disrupts vascular integrity through a PECAM-1 (platelet en-
dothelial cell adhesion molecule-1)-dependent mechanism, and
interferes with liver regeneration [29]. Similar findings were ob-
served characterizing MMP-10 [30], thus suggesting that MMP-
9/10-targeted therapies would allow more patients to undergo
successful liver resection, especially those with fatty liver, who
exhibit higher susceptibility to I/R.
Adipose tissue and adipocytokines
It has been suggested that, in conditions of PH (partial hepatec-
tomy) without I/R, adipose tissue supplies the energy needed by
the remnant liver. Considering that, in clinical practice, PH is
usually accompanied by I/R, a recent study has evaluated the
contribution of adipose tissue to liver injury and regeneration ob-
served in these surgical conditions. This study demonstrated that
adipose tissue is not required for the regeneration of non-steatotic
livers; however, it is necessary to promote regeneration and di-
minish injury in steatotic ones. Interestingly, adipose tissue does
not seem to be an energy source for the steatotic liver; indeed
ATP levels were maintained after lipectomy, but adipose tissue is
a source of different adipocytokines, which are essential signals
for liver regeneration [31].
Several adipocytokines may play a role in warm I/R injury;
however, very little is known about them. In the setting of I/R in-
jury without PH, adiponectin protects non-steatotic livers by an
AMPK (AMP-activated protein kinase)/eNOS mechanism [32].
Contrarily, this hormone is accumulated in steatotic livers upon
warm I/R injury through a MAPK-mediated mechanism, pro-
moting exacerbation of oxidative stress and liver injury [33].
Therefore the response of livers to adiponectin may very much
depend on their basal phenotype, involving different transduction
pathways.
RBP4 (retinol-binding protein 4), resistin and visfatin have
been characterized in experimental models of PH under warm
I/R. In this sense, RBP4 exerts injurious effects in steatotic and
non-steatotic livers, and its modulation further worsens the liver
outcome, thus it is not advised as a therapeutic strategy [34].
More recently, a relationship between resistin and visfatin has
been described [35]. Although no evident role for these adipo-
cytokines was observed in non-steatotic livers, in steatotic livers
endogenous resistin maintained low levels of visfatin by blocking
its hepatic uptake from the circulation, thus regulating the visfatin
detrimental effects on hepatic damage and regenerative failure.
NAD biosynthetic activity, rather than inflammatory response-
like activity, was responsible for the injurious effects of visfatin
in steatotic livers. This study indicates the clinical potential of
visfatin-blocking-based therapies in steatotic livers submitted to
I/R and regeneration.
See Figure 2 for a summary of mechanisms underlying warm
ischaemia and reperfusion injury.
Controversies
Differences between experimental models and clinical
practice
Numerous experimental animal models have been used in the
field of warm (and cold) I/R injury. There are many advantages
of animal studies: large numbers of animals can be studied, in-
terventional studies can be performed, and tools for targeted ma-
nipulation of gene expression provide insight into the function of
mediators in hepatic I/R injury. Comparison of the results of an-
imal studies and their extrapolation to human beings is feasible,
but with limitations such as differences in ischaemia tolerance,
anatomy of the liver of various species, surgical conditions used
in clinical practice and those used in the experimental models,
and administration, dosage and metabolic breakdown of the drugs
under investigation. Importantly, studies performed in small an-
imals are of limited applicability to human beings due to their
different size and anatomy of the liver and their faster metabol-
ism. Large animals exhibit greater similarity in their anatomy and
physiology to humans; however, their use is restricted by serious
logistical and financial difficulties, ethical concerns and limited
availability of immunological tools for use in large animal species
[36].
Despite the limitations of the experimental animal models,
these are the best options to study hepatic I/R, especially con-
sidering that the progress of human studies is slow, the majority
of human tissues are not routinely accessible for research, and
there is very limited opportunity for interventional studies. The
clinical application of strategies designed at benchside will de-
pend on the use of experimental models that resemble as much as
possible the clinical conditions in which the strategy intends to
be applied. As stated above, the pathophysiological mechanisms
very much differ depending on the type of ischaemia (cold or
warm). In fact, it should be considered that the extent and time
of ischaemia, the type of liver submitted to I/R, and the presence
of liver regeneration all lead to differences in the mechanisms
of hepatic I/R injury and in the effects of therapeutic strategies
350 C
The Authors Journal compilation C
2015 Biochemical Society
Underlying mechanisms of hepatic I/R injury
Figure 2 Underlying mechanisms of hepatic warm ischaemia and reperfusion injury
Pathways recently described are coloured, whereas classic mechanisms are summarized in grey. eNOS, endothelial
nitric oxide synthase; ET, endothelin; IL, interleukin; I/R wo hepatectomy, ischaemia/reperfusion without hepatectomy;
KC, Kupffer cells; KLF2, Kr¨
uppel-like factor 2; LSEC, liver sinusoidal endothelial cells; MMP, matrix metalloproteinase;
Nrf2, nuclear factor-erythroid 2-related factor 2; RBP4, retinol-binding protein 4; ROS, reactive oxygen species; TM,
thrombomodulin; TNF-α, tumour necrosis factor α.
evaluated [36]. Probably related to these factors, no satisfactory
treatment is currently available to prevent warm hepatic I/R injury
in the clinical practice. It should be considered that the effect-
iveness of a certain strategy could be different depending on the
surgical conditions evaluated: warm I/R injury itself or partial
hepatectomy under warm I/R injury. It would be extremely use-
ful to make a clear distinction between the mechanisms for each
surgical situation to design therapies that demonstrate its effect-
iveness under experimental conditions similar to what happens
in clinical practice. This will probably lead to translation of those
strategies to clinical practice in the short-term.
Lack of knowledge regarding sinusoidal cells in warm I/R
As stated above, non-parenchymal cells play essential roles in
liver function, resulting in being profoundly affected under I/R
conditions. Nevertheless, and although recent data unmasked the
phenotypic deregulations that LSEC suffer during cold ischaemia
and warm reperfusion [4,37], very little is known about sinusoidal
cell modifications due to warm ischaemia. An excellent study
by Jaeschke et al. [38] demonstrated that Kupffer cells become
activated after warm I/R, strongly contributing to liver injury.
These data, together with the recruitment of circulating neutro-
phils after I/R, suggest that the hepatic endothelium may rapidly
become dysfunctional upon warm I/R, therefore mediating the
inflammatory response. In this regard, a recent study from our
team described for the first time that the KLF2-derived vasopro-
tective pathways are rapidly deregulated in LSEC submitted to
warm I/R, leading to endothelial dysfunction development and
massive inflammation [39]. Undoubtedly, future studies will cla-
rify the effects of warm I/R on the phenotype and function of
LSEC, and desirably also of hepatic stellate cells.
Therapeutic approaches
In the last 2 years, pharmacological therapies with very different
targets have predominated in the research on novel strategies to
minimize the injurious effects of warm I/R (see Table 2). It is
also evident that there is an increasing interest in cellular and
surgical strategies. However, and as discussed above, it is import-
ant to note that strategies should be developed in experimental
models that resemble as much as possible the conditions present
in clinical practice, such as the use of intermittent clamping, the
combination of PH and I/R injury, and the use of pathological
livers such as steatotic or aged. In this sense, few studies have
been developed that comply with these characteristics, which can
represent an obstacle to the clinical application of other proposed
strategies. Accordingly, only one clinical trial analysing a therapy
to reduce warm I/R injury has been published in this period of
time.
C
The Authors Journal compilation C
2015 Biochemical Society 351
J. Gracia-Sancho, A. Casillas-Ram´
ırez and C. Peralta
Table 2 Therapeutic approaches against warm I/R injur y
AMPK, AMP-activated protein kinase; ATF4, activating transcription factor 4; CHOP, C/EBP (CCAAT/enhancer-binding protein)-homologous protein; eNOS, endothelial nitric oxide
synthase; ER, endoplasmic reticulum; FoxO1, forkhead box O1; GPR120, G-protein-coupled receptor 120; GSK3, glycogen synthase kinase 3; HMGB1, high-mobility group
protein B1; HO-1, haem oxygenase-1; Hsp70, heat-shock protein 70; IL, interleukin; IPC, ischaemic pre-conditioning; iNOS, inducible nitric oxide synthase; LSEC, liver sinusoidal
endothelial cells; MPO, myeloperoxidase; mTOR, mammalian target of rapamycin; NF-κB, nuclear factor κB; NKT, natural killer T-cell; NMP, normothermic machine perfusion; Nrf2,
nuclear factor-erythroid 2-related factor 2; PI3K, phosphoinositide 3-kinase; PKA, protein kinase A; PPAR, peroxisome-proliferator-activated receptor; RPC, remote pre-conditioning;
SIRT1, sirtuin 1; SOD, superoxide dismutase; STAT, signal transducer and activator of transcription; TLR, Toll-like receptor; TNFα, tumour necrosis factor α; Treg, regulatory T-cell.
(a) Pharmacological strategies
Drug Species Experimental model
War m
ischaemia Effect
15-deoxy-12,14-prostaglandin J2[76] Mouse Partial ischaemia 60 min Hepatic injury and inflammation; Nrf2-dependent antioxidant
response
Adiponectin [32] (adipocytokine) Rat Partial ischaemia 60 min Hepatic injury, inflammation and apoptosis; AMPK, eNOS
Ago-miR-146a [77] (a chemically modified
miR-146a)
Mouse Partial ischaemia 60 min Hepatic injury, TLR activation and apoptosis
Cold-inducible RNA-binding protein (CIRP) blockade
[78] (anti-inflammatory)
Mouse Partial ischaemia 60 min Hepatic injury, inflammatory response, apoptosis, nitrosative
stress; survival
Anti-CD25 monoclonal antibody [79] Mouse Partial ischaemia 60 min Hepatic injury; inflammatory response, CD4+T-lymphocytes
Atorvastatin [80] (vasoprotector) Rat Total ischaemia 60 min Hepatic injury and inflammation
Augmenter of liver regeneration [81] Mouse Partial ischaemia 90 min Hepatic injury and apoposis; recruitment of CD4+T-cells
Branched-chain amino acid (BCAA) [82] (Kupffer cell
inhibitor)
Rat Total ischaemia 30 min Hepatic injury, inflammation and microcirculatory disorders
Butyrate [28,83] (four-carbon short-chain fatty acid) Rat Partial ischaemia and partial
hepatectomy
30–60 min Hepatic injury and prevented acetylated histone H3 reduction,
inflammatory response, TLR4; inhibition of endotoxin
translocation; Hsp70
C1 esterase inhibitor [84] (complement inhibitor) Mouse Partial ischaemia and partial
hepatectomy
60 min Hepatic injury; liver regeneration and survival
Carbon monoxide [85] (gasotransmitter) Mouse Partial ischaemia 90 min Hepatic injury; phosphorylation of Akt, GSK3β
Carnosic acid [86] (rosemary derivative) Rat Partial ischaemia 45 min Hepatic injury and p66shc ;SOD and SIRT1
Carvacrol [87] (antimicrobial) Rat Total ischaemia 30 min Hepatic injury, apoptosis and oxidative stress; Akt
phosphorylation
Chloroquine [88] (anti-malaria and autophagy
inhibitor)
Rat Partial ischaemia 60, 90 min Early phase of reperfusion: hepatic injury, inflammation and
HMGB1. Late phase of reperfusion: hepatic injury and
apoptosis; autophagy
Cobalt protoporphyrin [89,90] (HO-1 inductor) Mouse Partial ischaemia 60–90 min Hepatic injury, inflammation and NF-κB, FoxO1 signalling; Nrf2,
HO-1, PI3K/Akt
CR2-CD59 [91] (complement inhibitor) Rat Total ischaemia 30 min Hepatic injury; regeneration and survival, TNFα, IL-6, STAT3,
Akt, ATP recovery
Dexmedetomidine [92] (sedative) Rat Par tial ischaemia 60 min Hepatic injury and oxidative stress
Diannexin [93] (microparticle inhibitor) Mouse Partial ischaemia 60 min Hepatic injury and inflammation; prevents formation of
pro-inflammatory and platelet-activating agents
Diazoxide [94,95] (potassium channel activator) Rat Partial ischaemia 30–60 min Hepatic injury, inflammation and TLR4, ihibition of endotoxin
translocation; protein kinase Cε
352 C
The Authors Journal compilation C
2015 Biochemical Society
Underlying mechanisms of hepatic I/R injury
Table 2 Continued
(a) Pharmacological strategies
Drug Species Experimental model
War m
ischaemia Effect
Dioscin [96] (antioxidant) Rat Partial ischaemia 60 min Hepatic injury, oxidative stress, inflammation and apoptosis;
survival
Dipyridamole [97] (equilibrative nucleoside
transporter inhibitor)
Mouse Partial ischaemia 45 min Hepatic injury; adenosine
Edaravone [98] (free radical scavenger) Rat Partial ischaemia 90 min Hepatic injury, oxidative stress and lung injury
Erythropoietin [99] (HO-1 inductor) Mouse Partial ischaemia 90 min Hepatic injury and apoptosis; HO-1
Ethyl pyruvate [100] (anti-inflammatory) Mouse Partial ischaemia 60 min Hepatic injury, apoptosis, autophagy and HMGB1–TLR4–NF-κB
axis
Exendin 4 [101] (glucagon-like peptide 1 analogue) Mouse Partial ischaemia (steatotic liver s) 20 min Hepatic injury and autophagy; peserves mitochondrial integrity
Fasting [102] Mouse Partial ischaemia 60 min Hepatic injury, inflammation and HMGB1; SIRT1, autophagy
Fibrin-derived peptide Bβ15-42 [103] (leucocyte
migration inhibitor)
Rat Partial ischaemia 60 min Hepatic injury, inflammation and HMGB1
Glucose or lipid emulsion [31] (nutritional
supplements)
Rat Partial ischaemia +partial
hepatectomy (steatotic and
non-steatotic livers)
60 min Hepatic injury; Liver regeneration, ATP level preservation
Helium [104] Mouse Partial ischaemia 90 min Hepatic injur y; survival, PI3K/Akt
Hydrogen sulfide [105,106] (gasotransmitter) Rat, mouse Partial ischaemia +partial
hepatectomy
75–90 min Hepatic injury and miR-34a;Nrf-2 signalling pathway,
pro-survival, anti-apoptotic and anti-inflammatory signals and
hepatic regeneration
Hydrolysed whey peptide [107,108] Rat Partial and total ischaemia (steatotic
and non-steatotic livers)
30 min Hepatic injury, apoptosis and inflammation; NF-κB
Hydroxytyrosol [109] (phenolic compound) Mouse Partial ischaemia 75 min Hepatic injury, apoptosis, inflammation and oxidative stress
Hyperbaric oxygen therapy [110] Rat Total ischaemia 30 min Hepatic injury; mitochondrial function
L-α-glycerylphosphorylcholine (GPC) [111]
(deacylated phospholipid)
Rat Partial ischaemia 60 min Hepatic injury, NADPH oxidase, MPO and HMGB1;
microcirculatory disorders
Levosimendan [112,113] (mitochondrial KATP
channel opener)
Rat Total and partial ischaemia 40 or 60 min Hepatic injury and apoptosis; cyclo-oxygenase-1, regulation of
oxidative stress, inflammation, NO, and KATP channel,
mitochondrial function; improved hepatic microcirculation
Limonin [114] (antioxidant) Rat Partial ischaemia 45 min Hepatic injury, oxidative stress, inflammation and TLR-signalling
pathway
Lithium [115] Rat Total and partial ischaemia 60–90 min Hepatic injur y; inflammation and HMGB1
Losartan [116] (angiotensin receptor antagonist) Mouse Partial ischaemia 60 min Hepatic injury, apoptosis and inflammation; PPARγ
Low-dose LPS [117] Mouse Partial ischaemia 90 min Hepatic injury, apoptosis, inflammation, and ATF4/CHOP pathway
Low-intensity laser therapy [118] Rat Partial ischaemia 45 min Hepatic injury, oxidative stress and TNF-α
Melatonin [119] (hormone) Rat Partial ischaemia (steatotic livers) 35 min Hepatic injury, apoptosis and oxidative stress; ATP
miR-370 inhibitor [120] Mouse Partial ischaemia 75 min Hepatic injury; inflammatory response
Minocycline [121] (antibiotic) Rat Partial ischaemia 2, 6 and 24 h Hepatic injur y, oxidative stress and inflammatory cytokines;
activation of Wnt/β-catenin signalling pathway
C
The Authors Journal compilation C
2015 Biochemical Society 353
J. Gracia-Sancho, A. Casillas-Ram´
ırez and C. Peralta
Table 2 Continued
(a) Pharmacological strategies
Drug Species Experimental model
War m
ischaemia Effect
N-Acetylcysteine [122] (antioxidant) Mice Partial ischaemia 40–90 min Hepatic injury, oxidative stress, ER stress and apoptosis;
improvement in structure of sinusoids; autophagy, apoptosis
and JNK phosphorylation
Platinum nanoparticles [123] Mouse Total ischaemia 15 min Hepatic injury
Propofol73 [124] (anaesthetic) Rat Partial ischaemia 30, 60, 90 min Hepatic injur y, apoptosis, GSK3β
Protease-activated receptor 4 antagonist [125]
(anti-platelet)
Mouse Partial ischaemia 90 min Hepatic injury, platelets, CD4+T-cell recruitment and apoptosis
Rapamycin [126] (mTOR inhibitor) Rat Partial ischaemia 60 min Hepatic injur y; autophagy, Akt
Reduced glutathione [127] (antioxidant) Rat Partial ischaemia (young and aged
rats)
90 min Hepatic injury, oxidative stress, TNFαand apoptosis
Resistin or anti-visfatin antibodies [35]
(adipocytokines)
Rat Partial ischaemia +partial
hepatectomy (steatotic and
non-steatotic livers)
60 min Hepatic injury and visfatin, NAD levels; liver regeneration
Rho-kinase inhibitor [128] Rat Total ischaemia (steatotic livers) 45 min Hepatic injury, portal perfusion pressure; survival
Riboflavin [129] (vitamin B2) Mice Partial ischaemia 60 min Hepatic injury and inflammation, oxidative stress, eNOS/iNOS
and NO levels
Rosmarinic acid [130] (antioxidant) Rat Partial ischaemia 60 min Hepatic injury; oxidative stress, inflammatory response, NF-κB
signalling pathway, iNOS, eNOS, NO
Sevofluorane [131] (anaesthetic) Swine Total ischaemia 40 min Hepatic injury
Sevofluorane [132] (anaesthetic) Rat Partial ischaemia 60 min Hepatic injury and oxidative stress
Sildenafil [133] (guanylate cyclase inhibitor) Rat Partial ischaemia 90 min Hepatic injury, apoptosis and inflammation
Simvastatin [39] (vasoprotector) Rat Partial ischaemia 60 min Early phase of reperfusion: hepatic injury, LSEC dysfunction. Late
phase of reperfusion: hepatic injury, macrophage and
neutrophil infiltration, apoptosis
Sivelestat sodium hydrate [134] (neutrophil
elastase inhibitor)
Rat Total ischaemia 30 min Hepatic injury and neutrophil accumulation
Thrombomodulin [135] (anti-coagulant) Rat Partial ischaemia +partial
hepatectomy
20 min Hepatic injury, apoptosis and macrophages infiltration; liver
regeneration
Vasoactive intestinal peptide neuropeptide [136] Mouse Partial ischaemia 90 min Hepatic injury, apoptosis and inflammation; cAMP/PKA
signalling
α7 Nicotinic acetylcholine receptor
activator/agonist [137,138]
Mouse Partial ischaemia 60 min Hepatic injury, oxidative stress, inflammation, TNFαand HMGB1;
HO-1, PI3K/Akt, Nrf2
ω3 Fatty acid formulation [139,140] Mouse Partial ischaemia 60 min Hepatic injury and inflammation; GPR120
354 C
The Authors Journal compilation C
2015 Biochemical Society
Underlying mechanisms of hepatic I/R injury
Table 2 Continued
(b) Gene and cell therapy strategies
Target gene/cell therapy strategy Species Experimental model Warm ischaemia Effect
Adipose-derived stem cells [141] Mouse Partial ischaemia 15, 20 min Hepatic injury; liver regeneration
ATF6 siRNA [142] (activating transcription factor 6) Mouse Partial ischaemia 90 min Hepatic injur y, ER stress, inflammation; macrophage TLR4
response, Akt activation
Bone-marrow-derived mesenchymal stem cells [143] Rat Partial ischaemia 60 min Hepatic injur y, oxidative stress and apoptosis
CD14/[144] Mouse Partial ischaemia 60 min Hepatic injur y, inflammation and apoptosis
ENT1/[97] (equilibrative nucleoside transporter 1) Mouse Partial ischaemia 45 min Hepatic injury; adenosine regulation of Adora2b receptor
Hepatic stellate cells [145] Mouse Partial ischaemia Not reported Hepatic injur y; Tre g s
Human adipose-derived mesenchymal stem cells [146] Mouse Partial ischaemia with or without partial
hepatectomy
60 min Hepatic injury; liver regeneration and survival
Interferon regulatory factor 9/[147] Mouse Partial ischaemia Not reported Hepatic injury, inflammation and apoptosis; SIRT1
Isolated viable mitochondria infusion [148] Rat Partial ischaemia 45 min Hepatic injury and apoptosis
Long non-coding RNA AK139328 siRNA [149] Rat Partial ischaemia 60 min Hepatic injury, apoptosis and inflammation; Akt, GSK3β
activation, eNOS
Mesenchymal stem cells [150] Rat Total ischaemia 30 min Hepatic injury and apoptosis
Mfn2 overexpression [151] (mitochondrial function
modulator)
Rat Partial ischaemia 90 min Hepatic injur y and apoptosis
MMP-9/[29] (matrix metalloproteinase 9) Mouse Partial ischaemia (steatotic livers) 60 min Hepatic injury and inflammation; liver regeneration
Myeloid PTEN deficiency [152] (phosphatase and
tensin homologue deleted on chromosome 10)
Mouse Partial ischaemia 90 min Hepatic injur y and inflammation; IL-10, macrophage
differentiation
Neogenin/[153] Mouse Partial ischaemia 30 min Hepatic injur y and inflammation
NLRP3/[154] (NOD-like receptor family, pyrin
domain-containing 3)
Mouse Partial ischaemia 60 min Hepatic injur y, inflammation, oxidative stress, apoptosis and
neutrophil infiltration
TIM-4/[155] (T-cell immunoglobulin and mucin 4) Mouse Partial ischaemia 90 min Hepatic injur y and inflammation; activation of
TLR2/4/9-dependent signalling
TLR4/[156] (Toll-like receptor 4) Mouse Partial ischaemia 60 min Hepatic injury, inflammation and HMGB1
Toll/interleukin-1 receptor blockade [157] Mouse Partial ischaemia 90 min Hepatic injury, oxidative stress, inflammation and TLR4
(c) Surgical strategies
Strategy Species Experimental model/clinical setting
Warm ischaemia
time Effect
IPC [158] Rat Partial ischaemia (young and aged rats) 60 min Hepatic injur y; autophagy, HO-1
IPC [86,159] Rat
Partial ischaemia (steatotic and
non-steatotic livers) 45–60 min Hepatic injury and p66shc ;SOD and SIRT1
IPC [131] Swine Total ischaemia 40 min Hepatic injury
IPC or GSK3 inhibitor [160] Rat Partial ischaemia (young and aged rats) 40 min Hepatic injury and oxidative stress; ATP
RPC [161] Rat Partial ischaemia 60 min Hepatic injur y; microcirculation and blood pressure
RPC [162] Rat Total ischaemia 30 min Hepatic injur y; survival and HO-1
RPC [163] Rat Partial ischaemia 45 min Hepatic injur y; HO-1/p38 MAPK-dependent and autophagy
RPC [164] Rat Partial ischaemia 60 min Hepatic injur y, inflammation and NADPH oxidase isoform 2
C
The Authors Journal compilation C
2015 Biochemical Society 355
J. Gracia-Sancho, A. Casillas-Ram´
ırez and C. Peralta
ACKNOWLEDGEMENTS
J.G.-S. and C.P. acknowledge the current and former members of
their laboratories for their valuable contributions.
FUNDING
Research from the groups of J.G.-S. and C.P. is supported by the
Instituto de Salud Carlos III [grant number FIS PI14/00029 to (J.G.-
S.)], the Spanish Ministry of Economy and Competitiveness [grant
number SAF2012-31238 to (C.P.)], and the European Funds FEDER.
CIBEREHD is funded by the Instituto de Salud Carlos III.
REFERENCES
1 de Rougemont, O., Lehmann, K. and Clavien, P.A. (2009)
Preconditioning, organ preservation, and postconditioning to
prevent ischemia–reperfusion injury to the liver. Liver Transpl.
15, 1172–1182 CrossRef PubMed
2 Montalvo-Jave, E.E., Escalante-Tattersfield, T., Ortega-Salgado,
J.A., Pina, E. and Geller, D.A. (2008) Factors in the
pathophysiology of the liver ischemia–reperfusion injury. J.
Surg. Res. 147, 153–159 CrossRef PubMed
3 Rauen, U., Polzar, B., Stephan, H., Mannherz, H.G. and de
Groot, H. (1999) Cold-induced apoptosis in cultured
hepatocytes and liver endothelial cells: mediation by reactive
oxygen species. FASEB J. 13, 155–168 PubMed
4 Russo, L., Gracia-Sancho, J., Garcia-Caldero, H., Marrone, G.,
Garcia-Pagan, J.C., Garcia-Cardena, G. and Bosch, J. (2012)
Addition of simvastatin to cold storage solution prevents
endothelial dysfunction in explanted rat livers. Hepatology 55,
921–930 CrossRef PubMed
5 Stolz, D.B., Ross, M.A., Ikeda, A., Tomiyama, K., Kaizu, T.,
Geller, D.A. and Murase, N. (2007) Sinusoidal endothelial cell
repopulation following ischemia/reperfusion injury in rat liver
transplantation. Hepatology 46, 1464–1475
CrossRef PubMed
6 Gracia-Sancho, J., Villarreal, Jr, G., Zhang, Y., Yu, J.X., Liu, Y.,
Tullius, S.G. and Garcia-Cardena, G. (2010) Flow cessation
triggers endothelial dysfunction during organ cold storage
conditions: strategies for pharmacologic intervention.
Transplantation 90, 142–149 CrossRef PubMed
7 Shen, X.D., Ke, B., Zhai, Y., Gao, F., Tsuchihashi, S., Lassman,
C.R., Busuttil, R.W. and Kupiec-Weglinski, J.W. (2007) Absence
of Toll-like receptor 4 (TLR4) signaling in the donor organ
reduces ischemia and reperfusion injury in a murine liver
transplantation model. Liver Transpl. 13, 1435–1443
CrossRef PubMed
8 Jimenez-Castro, M.B., Elias-Miro, M., Mendes-Braz, M.,
Lemoine, A., Rimola, A., Rodes, J., Casillas-Ram´
ırez, A. and
Peralta, C. (2012) Tauroursodeoxycholic acid affects PPARγ
and TLR4 in steatotic liver transplantation. Am. J. Transplant.
12, 3257–3271 CrossRef PubMed
9 Minor, T., Stegemann, J., Hirner, A. and Koetting, M. (2009)
Impaired autophagic clearance after cold preservation of fatty
livers correlates with tissue necrosis upon reperfusion and is
reversed by hypothermic reconditioning. Liver Transpl. 15,
798–805 CrossRef PubMed
10 Farid, W.R., Pan, Q., van der Meer, A.J., de Ruiter, P.E.,
Ramakrishnaiah, V., de Jonge, J., Kwekkeboom, J., Janssen,
H.L., Metselaar, H.J., Tilanus, H.W. et al. (2012)
Hepatocyte-derived microRNAs as serum biomarkers of hepatic
injury and rejection after liver transplantation. Liver Transpl. 18,
290–297 CrossRef PubMed
11 Kim, K.H., Dhupar, R., Ueki, S., Cardinal, J., Pan, P., Cao, Z.,
Cho, S.W., Murase, N., Tsung, A. and Geller, D.A. (2009) Donor
graft interferon regulatory factor-1 gene transfer worsens liver
transplant ischemia/reperfusion injury. Surgery 146, 181–189
CrossRef PubMed
12 Ueki, S., Dhupar, R., Cardinal, J., Tsung, A., Yoshida, J., Ozaki,
K.S., Klune, J.R., Murase, N. and Geller, D.A. (2010) Critical
role of interferon regulatory factor-1 in murine liver transplant
ischemia reperfusion injury. Hepatology 51, 1692–1701
CrossRef PubMed
13 Ueki, S., Castellaneta, A., Yoshida, O., Ozaki, K., Zhang, M.,
Kimura, S., Isse, K., Ross, M., Shao, L., Stolz, D.B., Thomson,
A.W., Demetris, A.J., Geller, D.A. and Murase, N. (2011)
Hepatic B7 homolog 1 expression is essential for controlling
cold ischemia/reperfusion injury after mouse liver
transplantation. Hepatology 54, 216–228
CrossRef PubMed
14 Van der Hoeven, J.A., Lindell, S., van, S.R., Molema, G., Ter
Horst, G.J., Southard, J.H. and Ploeg, R.J. (2001) Donor brain
death reduces survival after transplantation in rat livers
preserved for 20 hr. Transplantation. 72, 1632–1636
CrossRef PubMed
15 Jimenez-Castro, M.B., Merono, N., Mendes-Braz, M.,
Gracia-Sancho, J., Martinez-Carreres, L., Cornide-Petronio,
M.E., Casillas-Ram´
ırez, A., Rodes, J. and Peralta, C. (2014) The
effect of brain death in rat steatotic and non-steatotic liver
transplantation with previous ischemic preconditioning. J.
Hepatol. 62, 83–91 CrossRef PubMed
16 Vollmar, B., Glasz, J., Leiderer, R., Post, S. and Menger, M.D.
(1994) Hepatic microcirculatory perfusion failure is a
determinant of liver dysfunction in warm ischemia–reperfusion.
Am. J. Pathol. 145, 1421–1431 PubMed
17 Fondevila, C., Hessheimer, A.J., Maathuis, M.H., Munoz, J.,
Taura, P., Calatayud, D., Leuvenink, H., Rimola, A., Ploeg, R.J.
and Garcia-Valdecasas, J.C. (2011) Superior preservation of
DCD livers with continuous normothermic perfusion. Ann. Surg.
254, 1000–1007 CrossRef PubMed
18 Sutton, M.E., op den Dries, S., Karimian, N., Weeder, P.D., de
Boer, M.T., Wiersema-Buist, J., Gouw, A.S., Leuvenink, H.G.,
Lisman, T. and Por te, R.J. (2014) Criteria for viability
assessment of discarded human donor livers during ex vivo
normothermic machine perfusion. PLoS ONE 9, e110642
CrossRef PubMed
19 op den Dries, S., Karimian, N., Sutton, M.E., Westerkamp, A.C.,
Nijsten, M.W., Gouw, A.S., Wiersema-Buist, J., Lisman, T.,
Leuvenink, H.G. and Porte, R.J. (2013) Ex vivo normothermic
machine perfusion and viability testing of discarded human
donor livers. Am. J. Transplant. 13, 1327–1335
CrossRef PubMed
20 Barrou, B., Billault, C. and Nicolas-Robin, A. (2013) The use of
extracorporeal membranous oxygenation in donors after cardiac
death. Curr. Opin. Organ Transplant. 18, 148–153
CrossRef PubMed
21 Valero, R., Cabrer, C., Oppenheimer, F., Trias, E.,
Sanchez-Ibanez, J., De Cabo, F.M., Navarro, A., Paredes, D.,
Alcaraz, A., Gutierrez, R. and Manyalich, M. (2000)
Normothermic recirculation reduces primary graft dysfunction of
kidneys obtained from non-heart-beating donors. Transpl. Int.
13, 303–310 CrossRef PubMed
22 Fondevila, C., Hessheimer, A.J., Ruiz, A., Calatayud, D., Ferrer,
J., Charco, R., Fuster, J., Navasa, M., Rimola, A., Taura, P. et al.
(2007) Liver transplant using donors after unexpected cardiac
death: novel preservation protocol and acceptance criteria. Am.
J. Transplant. 7, 1849–1855 CrossRef PubMed
23 Butler, A.J., Randle, L.V. and Watson, C.J. (2014) Normothermic
regional perfusion for donation after circulatory death without
prior heparinization. Transplantation 97, 1272–1278
CrossRef PubMed
356 C
The Authors Journal compilation C
2015 Biochemical Society
Underlying mechanisms of hepatic I/R injury
24 Oniscu, G.C., Randle, L.V., Muiesan, P., Butler, A.J., Currie, I.S.,
Perera, M.T., Forsythe, J.L. and Watson, C.J. (2014) In situ
normothermic regional perfusion for controlled donation after
circulatory death: the United Kingdom experience. Am. J.
Transplant. 14, 2846–2854 CrossRef PubMed
25 Fontes, P., Lopez, R., van der Plaats, A., Vodovotz, Y., Minervini,
M., Scott, V., Soltys, K., Shiva, S., Paranjpe, S., Sadowsky, D.
et al. (2015) Liver preservation with machine perfusion and a
newly developed cell-free oxygen carrier solution under
subnormothermic conditions. Am. J. Transplant. 15, 381–394
CrossRef PubMed
26 Jaeschke, H. (1998) Mechanisms of reperfusion injury after
warm ischemia of the liver. J. Hepatobiliary Pancreat. Surg. 5,
402–408 CrossRef PubMed
27 Martens, J.C., Keilhoff, G., Halangk, W., Wartmann, T.,
Gardemann, A., Page, I. and Schild, L. (2015) Lipidomic
analysis of molecular cardiolipin species in livers exposed to
ischemia/reperfusion. Mol. Cell. Biochem. 400, 253–263
CrossRef PubMed
28 Sun, J., Wu, Q., Sun, H. and Qiao, Y. (2014) Inhibition of
histone deacetylase by butyrate protects rat liver from ischemic
reperfusion injury. Int. J. Mol. Sci. 15, 21069–21079
CrossRef PubMed
29 Kato, H., Kuriyama, N., Duarte, S., Clavien, P.A., Busuttil, R.W.
and Coito, A.J. (2014) MMP-9 deficiency shelters endothelial
PECAM-1 expression and enhances regeneration of steatotic
livers after ischemia and reperfusion injury. J. Hepatol. 60,
1032–1039 CrossRef PubMed
30 Garcia-Irigoyen, O., Carotti, S., Latasa, M.U., Uriarte, I.,
Fernandez-Barrena, M.G., Elizalde, M., Urtasun, R.,
Vespasiani-Gentilucci, U., Morini, S., Banales, J.M. et al.
(2014) Matrix metalloproteinase-10 expression is induced
during hepatic injury and plays a fundamental role in liver tissue
repair. Liver Int. 34, e257–e270 CrossRef PubMed
31 Mendes-Braz, M., Elias-Miro, M., Kleuser, B., Fayyaz, S.,
Jimenez-Castro, M.B., Massip-Salcedo, M., Gracia-Sancho, J.,
Ramalho, F.S., Rodes, J. and Peralta, C. (2014) The effects of
glucose and lipids in steatotic and non-steatotic livers in
conditions of partial hepatectomy under ischaemia–reperfusion.
Liver Int. 34, e271–e289 CrossRef PubMed
32 Zhang, C., Liao, Y., Li, Q., Chen, M., Zhao, Q., Deng, R., Wu, C.,
Yang, A., Guo, Z., Wang, D. and He, X. (2013) Recombinant
adiponectin ameliorates liver ischemia reperfusion injury via
activating the AMPK/eNOS pathway. PLoS ONE 8, e66382
CrossRef PubMed
33 Massip-Salcedo, M., Zaouali, M.A., Padrissa-Altes, S.,
Casillas-Ram´
ırez, A., Rodes, J., Rosello-Catafau, J. and Peralta,
C. (2008) Activation of peroxisome proliferator-activated
receptor-αinhibits the injurious effects of adiponectin in rat
steatotic liver undergoing ischemia–reperfusion. Hepatology
47, 461–472 CrossRef PubMed
34 Elias-Miro, M., Massip-Salcedo, M., Raila, J., Schweigert, F.,
Mendes-Braz, M., Ramalho, F., Jimenez-Castro, M.B.,
Casillas-Ram´
ırez, A., Bermudo, R., Rimola, A. et al. (2012)
Retinol binding protein 4 and retinol in steatotic and
nonsteatotic rat livers in the setting of partial hepatectomy
under ischemia/reperfusion. Liver Transpl. 18, 1198–1208
CrossRef PubMed
35 Elias-Miro, M., Mendes-Braz, M., Cereijo, R., Villarroya, F.,
Jimenez-Castro, M.B., Gracia-Sancho, J., Guixe-Muntet, S.,
Massip-Salcedo, M., Domingo, J.C., Bermudo, R. et al. (2014)
Resistin and visfatin in steatotic and non-steatotic livers in the
setting of partial hepatectomy under ischemia–reperfusion. J.
Hepatol. 60, 87–95 CrossRef PubMed
36 Mendes-Braz, M., Elias-Miro, M., Jimenez-Castro, M.B.,
Casillas-Ram´
ırez, A., Ramalho, F.S. and Peralta, C. (2012) The
current state of knowledge of hepatic ischemia–reperfusion
injury based on its study in experimental models. J. Biomed.
Biotechnol. 2012, 298657 CrossRef PubMed
37 Hide, D., Ortega-Ribera, M., Fernandez-Iglesias, A., Fondevila,
C., Salvado, M.J., Arola, L., Garcia-Pagan, J.C., Mancini, A.,
Bosch, J. and Gracia-Sancho, J. (2014) A novel form of the
human manganese superoxide dismutase protects rat and
human livers undergoing ischemia and reperfusion injuries.
Clin. Sci. 127, 527–537 CrossRef PubMed
38 Jaeschke, H., Bautista, A.P., Spolarics, Z. and Spitzer, J.J.
(1992) Superoxide generation by neutrophils and Kupffer cells
during in vivo reperfusion after hepatic ischemia in rats. J.
Leukoc. Biol. 52, 377–382 PubMed
39 Hide, D., Ortega-Ribera, M., Vila, S., Peralta, C., Garcia-Pagan,
J.C., Bosch, J. and Gracia-Sancho, J. (2014) Warm ischemia
and reperfusion causes liver microcirculatory injury and acute
endothelial dysfunction: simvastatin prevents these deleterious
events. Hepatology 60, 522A
40 Jimenez-Castro, M.B., Casillas-Ram´
ırez, A., Mendes-Braz, M.,
Massip-Salcedo, M., Gracia-Sancho, J., Elias-Miro, M., Rodes,
J. and Peralta, C. (2013) Adiponectin and resistin protect
steatotic livers undergoing transplantation. J. Hepatol. 59,
1208–1214 CrossRef PubMed
41 Chen, G., Zhang, L., Chen, L., Wang, H., Zhang, Y. and Bie, P.
(2013) Role of integrin αvβ6 in the pathogenesis of
ischemia-related biliary fibrosis after liver transplantation.
Transplantation 95, 1092–1099 CrossRef PubMed
42 Andria, B., Bracco, A., Attanasio, C., Castaldo, S., Cerrito,
M.G., Cozzolino, S., Di Napoli, D., Giovannoni, R., Mancini, A.,
Musumeci, A. et al. (2013) Biliverdin protects against liver
ischemia reperfusion injury in swine. PLoS ONE 8, e69972
CrossRef PubMed
43 Tiriveedhi, V., Upadhya, G.A., Busch, R.A., Gunter, K.L., Dines,
J.N., Knolhoff, B.L., Jia, J., Sarma, N.J., Ramachandran, S.,
Anderson, C.D. et al. (2014) Protective role of bortezomib in
steatotic liver ischemia/reperfusion injury through abrogation of
MMP activation and YKL-40 expression. Transpl. Immunol. 30,
93–98 CrossRef PubMed
44 Aguilar-Melero, P., Luque, A., Machuca, M.M., P´
erez de Obanos,
M.P., Navarrete, R., Rodr´
ıguez-Garc´
ıa, I.C., Briceno, J., Iniguez,
M., Ruiz, J., Prieto, J. et al. (2013) Cardiotrophin-1 reduces
ischemia/reperfusion injury during liver transplant. J. Surg.
Res. 181, e83–e91 CrossRef PubMed
45 Miyazawa, K., Miyagi, S., Maida, K., Murakami, K., Fujio, A.,
Kashiwadate, T., Nakanishi, W., Hara, Y., Nakanishi, C.,
Yamaya, H. et al. (2014) Edaravone, a free radical scavenger,
improves the graft viability on liver transplantation from
non-heart-beating donors in pigs. Transplant. Proc. 46,
1090–1094 CrossRef PubMed
46 Lehne, K. and Nobiling, R. (2013) Metabolic preconditioning
with fructose prior to organ recovery attenuates
ischemia–reperfusion injury in the isolated perfused rat liver.
Scand. J. Gastroenterol. 48, 218–224 CrossRef PubMed
47 Hori, T., Gardner, L.B., Hata, T., Chen, F., Baine, A.M., Uemoto,
S. and Nguyen, J.H. (2013) Pretreatment of liver grafts in vivo
by γ-aminobutyric acid receptor regulation reduces cold
ischemia/warm reperfusion injury in rat. Ann. Transplant. 18,
299–313 CrossRef PubMed
48 Chen, L., Chen, G., Guo, Y., Liu, L., Xiao, L., Fan, W., Shi, B. and
Qian, Y. (2014) Ketanserin, a serotonin 2A receptor antagonist,
alleviates ischemia-related biliary fibrosis following donation
after cardiac death liver transplantation in rats. Liver Transpl.
20, 1317–1326 CrossRef PubMed
49 Chung, H.S., Park, C.S., Hong, S.H., Lee, S., Cho, M.L., Her,
Y.M., Sa, G.J., Lee, J. and Choi, J.H. (2013) Effects of
magnesium pretreatment on the levels of T helper cytokines
and on the severity of reperfusion syndrome in patients
undergoing living donor liver transplantation. Magnes. Res. 26,
46–55 PubMed
50 Tang, Y., Zhang , W., Zhang, Y., Wang, W., Yao, F., Yan, J. and
Wan, C. (2014) 5-Methylthioadenosine attenuates ischemia
reperfusion injury after liver transplantation in rats.
Inflammation 37, 1366–1373 CrossRef PubMed
C
The Authors Journal compilation C
2015 Biochemical Society 357
J. Gracia-Sancho, A. Casillas-Ram´
ırez and C. Peralta
51 D’Amico, F., V itale, A., Piovan, D., Bertacco, A., Ramirez, M.R.,
Chiara, F.A., Bassi, D., Bonsignore, P., Gringeri, E., Valmasoni,
M. et al. (2013) Use of N-acetylcysteine during liver
procurement: a prospective randomized controlled study. Liver
Transpl. 19, 135–144 CrossRef PubMed
52 Bezinover, D., Ramamoorthy, S., Postula, M., Weller, G.,
Mahmoud, S., Mani, H., Kadry, Z., Uemura, T., Mets, B.,
Spiess, B. et al. (2014) Effect of cold perfusion and
perfluorocarbons on liver graft ischemia in a donation after
cardiac death model. J. Surg. Res. 188, 517–526
CrossRef PubMed
53 Zhang, Y., Ji, H., Shen, X., Cai, J., Gao, F., Koenig, K.M.,
Batikian, C.M., Busuttil, R.W. and Kupiec-Weglinski, J.W. (2013)
Targeting TIM-1 on CD4 T cells depresses macrophage
activation and overcomes ischemia–reperfusion injury in mouse
orthotopic liver transplantation. Am. J. Transplant. 13, 56–66
CrossRef PubMed
54 Gracia-Sancho, J., Garcia-Caldero, H., Hide, D., Marrone, G.,
Guixe-Muntet, S., Peralta, C., Garcia-Pagan, J.C., Abraldes, J.G.
and Bosch, J. (2013) Simvastatin maintains function and
viability of steatotic rat livers procured for transplantation. J.
Hepatol. 58, 1140–1146 CrossRef PubMed
55 Liu, Z.Y., Wang, W., Jin, B., Li, G.Z., Du, G., Zhang, Z.L., Han,
L.T., Huang, G.Z. and Tang, Z.Y. (2014) Protection against
ischemia–reperfusion injury in aged liver donor by the
induction of exogenous human telomerase reverse
transcriptase gene. Transplant. Proc. 46, 1567–1572
CrossRef PubMed
56 Yoshida, O., Kimura, S., Jackson, E.K., Robson, S.C., Geller,
D.A., Murase, N. and Thomson, A.W. (2013) CD39 expression
by hepatic myeloid dendritic cells attenuates inflammation in
liver transplant ischemia–reperfusion injury in mice. Hepatology
58, 2163–2175 CrossRef PubMed
57 Pommey, S., Lu, B., McRae, J., Stagg, J., Hill, P., Salvaris, E.,
Robson, S.C., d’Apice, A.J., Cowan, P.J. and Dwyer, K.M. (2013)
Liver grafts from CD39-overexpressing rodents are protected
from ischemia reperfusion injury due to reduced numbers of
resident CD4+T cells. Hepatology 57, 1597–1606
CrossRef PubMed
58 Ke, B., Shen, X.D., Zhang, Y., Ji, H., Gao, F., Yue, S., Kamo, N.,
Zhai, Y., Yamamoto, M., Busuttil, R.W. and Kupiec-Weglinski,
J.W. (2013) KEAP1–NRF2 complex in ischemia-induced
hepatocellular damage of mouse liver transplants. J. Hepatol.
59, 1200–1207 CrossRef PubMed
59 Du, Z., Wei, C., Cheng, K., Han, B., Yan, J., Zhang, M., Peng, C.
and Liu, Y. (2013) Mesenchymal stem cell-conditioned medium
reduces liver injury and enhances regeneration in reduced-size
rat liver transplantation. J. Surg. Res. 183, 907–915
CrossRef PubMed
60 Bejaoui, M., Zaouali, M.A., Folch-Puy, E., Pantazi, E.,
Bardag-Gorce, F., Carbonell, T., Oliva, J., Rimola, A., Abdennebi,
H.B. and Rosello-Catafau, J. (2014) Bortezomib enhances fatty
liver preservation in Institut George Lopez-1 solution through
adenosine monophosphate activated protein kinase and
Akt/mTOR pathways. J. Pharm. Pharmacol. 66, 62–72
CrossRef PubMed
61 Zaouali, M.A., Bardag-Gorce, F., Carbonell, T., Oliva, J., Pantazi,
E., Bejaoui, M., Ben, A.H., Rimola, A. and Rosello-Catafau, J.
(2013) Proteasome inhibitors protect the steatotic and
non-steatotic liver graft against cold ischemia reperfusion
injury. Exp. Mol. Pathol. 94, 352–359
CrossRef PubMed
62 Arthur, P.G., Niu, X.W., Huang, W.H., Deboer, B., Lai, C.T., Rossi,
E., Joseph, J. and Jeffrey, G.P. (2013) Desferrioxamine in warm
reperfusion media decreases liver injury aggravated by cold
storage. World J. Gastroenterol. 19, 673–681
CrossRef PubMed
63 Zaouali, M.A., Boncompagni, E., Reiter, R.J., Bejaoui, M.,
Freitas, I., Pantazi, E., Folch-Puy, E., Abdennebi, H.B.,
Garcia-Gil, F.A. and Rosello-Catafau, J. (2013) AMPK
involvement in endoplasmic reticulum stress and autophagy
modulation after fatty liver graft preservation: a role for
melatonin and trimetazidine cocktail. J. Pineal Res. 55, 65–78
CrossRef PubMed
64 Ishima, Y., Shinagawa, T., Yoneshige, S., Kragh-Hansen, U.,
Ohya, Y., Inomata, Y., Kai, T., Otagiri, M. and Maruyama, T.
(2013) UW solution improved with high anti-apoptotic activity by
S-nitrosated human serum albumin. Nitric Oxide 30, 36–42
CrossRef PubMed
65 Hara, Y., Akamatsu, Y., Maida, K., Kashiwadate, T., Kobayashi,
Y., Ohuchi, N. and Satomi, S. (2013) A new liver graft
preparation method for uncontrolled non-heart-beating donors,
combining short oxygenated warm perfusion and prostaglandin
E1. J. Surg. Res. 184, 1134–1142 CrossRef PubMed
66 Schlegel, A., Kron, P., Graf, R., Dutkowski, P. and Clavien, P.A.
(2014) Warm vs. cold perfusion techniques to rescue rodent
liver grafts. J. Hepatol. 61, 1267–1275 CrossRef PubMed
67 Schlegel, A., Graf, R., Clavien, P.A. and Dutkowski, P. (2013)
Hypothermic oxygenated perfusion (HOPE) protects from biliary
injury in a rodent model of DCD liver transplantation. J. Hepatol.
59, 984–991 CrossRef PubMed
68 Schlegel, A., Rougemont, O., Graf, R., Clavien, P.A. and
Dutkowski, P. (2013) Protective mechanisms of end-ischemic
cold machine perfusion in DCD liver grafts. J. Hepatol. 58,
278–286 CrossRef PubMed
69 Ren, Z., Cui, G., Lu, H., Chen, X., Jiang, J., Liu, H., He, Y., Ding,
S., Hu, Z., Wang, W. and Zheng, S. (2013) Liver ischemic
preconditioning (IPC) improves intestinal microbiota following
liver transplantation in rats through 16s rDNA-based analysis of
microbial structure shift. PLoS ONE 8, e75950
CrossRef PubMed
70 Izamis, M.L., Tolboom, H., Uygun, B., Berthiaume, F., Yarmush,
M.L. and Uygun, K. (2013) Resuscitation of ischemic donor
livers with normothermic machine perfusion: a metabolic flux
analysis of treatment in rats. PLoS ONE 8, e69758
CrossRef PubMed
71 Shigeta, T., Matsuno, N., Obara, H., Kanazawa, H., Tanaka, H.,
Fukuda, A., Sakamoto, S., Kasahara, M., Mizunuma, H. and
Enosawa, S. (2013) Impact of rewarming preservation by
continuous machine perfusion: improved post-transplant
recovery in pigs. Transplant. Proc. 45, 1684–1689
CrossRef PubMed
72 Kim, W.H., Lee, J.H., Ko, J.S., Min, J.J., Gwak, M.S., Kim, G.S.
and Lee, S.K. (2014) Effect of remote ischemic
postconditioning on patients undergoing living donor liver
transplantation. Liver Transpl. 20, 1383–1392
CrossRef PubMed
73 Nagai, K., Yagi, S., Afify, M., Bleilevens, C., Uemoto, S. and
Tolba, R.H. (2013) Impact of venous-systemic oxygen
persufflation with nitric oxide gas on steatotic grafts after
partial orthotopic liver transplantation in rats. Transplantation
95, 78–84 CrossRef PubMed
74 Yagi, S., Nagai, K., Kadaba, P., Afify, M., Teramukai, S., Uemoto,
S. and Tolba, R.H. (2013) A novel organ preservation for small
partial liver transplantations in rats: venous systemic oxygen
persufflation with nitric oxide gas. Am. J. Transplant. 13,
222–228 CrossRef PubMed
75 Kageyama, S., Yagi, S., Tanaka, H., Saito, S., Nagai, K., Hata,
K., Fujimoto, Y., Ogura, Y., Tolba, R. and Shinji, U. (2014) Graft
reconditioning with nitric oxide gas in rat liver transplantation
from cardiac death donors. Transplantation 97, 618–625
CrossRef PubMed
76 Kudoh, K., Uchinami, H., Yoshioka, M., Seki, E. and Yamamoto,
Y. (2014) Nrf2 activation protects the liver from
ischemia/reperfusion injury in mice. Ann. Surg. 260, 118–127
CrossRef PubMed
358 C
The Authors Journal compilation C
2015 Biochemical Society
Underlying mechanisms of hepatic I/R injury
77 Jiang, W., Kong, L., Ni, Q., Lu, Y., Ding, W., Liu, G., Pu, L., Tang ,
W. and Kong, L. (2014) miR-146a ameliorates liver
ischemia/reperfusion injury by suppressing IRAK1 and TRAF6.
PLoS ONE 9, e101530 CrossRef PubMed
78 Godwin, A., Yang, W.L., Sharma, A., Khader, A., Wang, Z.,
Zhang, F., Nicastro, J., Coppa, G.F. and Wang, P. (2015)
Blocking cold-inducible RNA-binding protein (CIRP) protects liver
from ischemia/reperfusion injury. Shock 43, 24–30
CrossRef PubMed
79 Yang, J., Wang, X., Song, S., Liu, F., Fu, Z. and Wang, Q. (2014)
Near-term anti-CD25 monoclonal antibody administration
protects murine liver from ischemia–reperfusion injury due to
reduced numbers of CD4+T cells. PLoS ONE 9, e106892
CrossRef PubMed
80 C´
amara-Lemarroy, C.R., Guzm´
an-de la Garza, F.J.,
Alarc´
on-Galv´
an, G., Cordero-P´
erez, P., Mu˜
noz-Espinosa, L.,
Torres-Gonz ´
alez, L. and Fern´
andez-Garza, N.E. (2014) Hepatic
ischemia/reperfusion injury is diminished by atorvastatin in
Wistar rats. Arch. Med. Res. 45, 210–216 CrossRef PubMed
81 Khandoga, A., Mende, K., Iskandarov, E., Rosentreter, D.,
Schelcher, C., Reifart, J., Jauch, K.W. and Thasler, W.E. (2014)
Augmenter of liver regeneration attenuates inflammatory
response in the postischemic mouse liver in vivo. J. Surg. Res.
192, 187–194 CrossRef PubMed
82 Kitagawa, T., Yokoyama, Y., Kokuryo, T. and Nagino, M. (2013)
Protective effects of branched-chain amino acids on hepatic
ischemia–reperfusion-induced liver injury in rats: a direct
attenuation of Kupffer cell activation. Am. J. Physiol.
Gastrointest. Liver Physiol. 304, G346–G355 CrossRef PubMed
83 Liu, B., Qian, J., Wang, Q., Wang, F., Ma, Z. and Qiao, Y. (2014)
Butyrate protects rat liver against total hepatic ischemia
reperfusion injury with bowel congestion. PLoS ONE 9,
e106184 CrossRef PubMed
84 Saidi, R.F., Rajeshkumar, B., Shariftabrizi, A., Dresser, K. and
Walter, O. (2014) Human C1 inhibitor attenuates liver
ischemia–reperfusion injury and promotes liver regeneration. J.
Surg. Res. 187, 660–666 CrossRef PubMed
85 Kim, H.J., Joe, Y., Kong, J.S., Jeong, S.O., Cho, G.J., Ryter, S.W.
and Chung, H.T. (2013) Carbon monoxide protects against
hepatic ischemia/reperfusion injury via ROS-dependent Akt
signaling and inhibition of glycogen synthase kinase 3β. Oxid.
Med. Cell Longev. 2013, 306421 PubMed
86 Yan, H., Jihong, Y., Feng, Z., Xiaomei, X., Xiaohan, Z., Guangzhi,
W., Zhenhai, M., Dongyan, G., Xiaochi, M., Qing, F. et al. (2014)
Sirtuin 1-mediated inhibition of p66shc expression alleviates
liver ischemia/reperfusion injury. Crit. Care Med. 42,
e373–e381 CrossRef PubMed
87 Suo, L., Kang, K., Wang, X., Cao, Y., Zhao, H., Sun, X., Tong, L.
and Zhang, F. (2014) Carvacrol alleviates ischemia reperfusion
injury by regulating the PI3K–Akt pathway in rats. PLoS ONE 9,
e104043 CrossRef PubMed
88 Fang, H., Liu, A., Dahmen, U. and Dirsch, O. (2013) Dual role of
chloroquine in liver ischemia reperfusion injury: reduction of
liver damage in early phase, but aggravation in late phase. Cell
Death Dis. 4, e694 CrossRef PubMed
89 Ben-Ari, Z., Issan, Y., Katz, Y., Sultan, M., Safran, M., Michal,
L.S., Nader, G.A., Kornowski, R., Grief, F., Pappo, O. and
Hochhauser, E. (2013) Induction of heme oxygenase-1 protects
mouse liver from apoptotic ischemia/reperfusion injury.
Apoptosis 18, 547–555 CrossRef PubMed
90 Huang, J., Yue, S., Ke, B., Zhu, J., Shen, X.D., Zhai, Y.,
Yamamoto, M., Busuttil, R.W. and Kupiec-Weglinski, J.W.
(2014) Nuclear factor erythroid 2-related factor 2 regulates
Toll-like receptor 4 innate responses in mouse liver
ischemia–reperfusion injury through Akt-forkhead box protein
O1 signaling network. Transplantation 98, 721–728
CrossRef PubMed
91 Marshall, K.M., He, S., Zhong, Z., Atkinson, C. and Tomlinson,
S. (2014) Dissecting the complement pathway in hepatic injury
and regeneration with a novel protective strategy. J. Exp. Med.
211, 1793–1805 CrossRef PubMed
92 Sahin, T., Begec, Z., Toprak, H.I., Polat, A., Vardi, N., Yucel, A.,
Durmus, M. and Ersoy, M.O. (2013) The effects of
dexmedetomidine on liver ischemia–reperfusion injury in rats. J.
Surg. Res. 183, 385–390 CrossRef PubMed
93 Teoh, N.C., Ajamieh, H., Wong, H.J., Croft, K., Mori, T., Allison,
A.C. and Farrell, G.C. (2014) Microparticles mediate hepatic
ischemia–reperfusion injury and are the targets of Diannexin
(ASP8597). PLoS ONE 9, e104376 CrossRef PubMed
94 Nogueira, M.A., Coelho, A.M., Sampietre, S.N., Patzina, R.A.,
Pinheiro da Silva, F., D’Albuquerque, L.A. and Machado, M.C.
(2014) Beneficial effects of adenosine triphosphate-sensitive
K+channel opener on liver ischemia/reperfusion injury. World
J. Gastroenterol. 20, 15319–15326 CrossRef PubMed
95 Tian, Y.S., Rong, T.Z., Hong, Y.L., Min, L. and Jian, P.G. (2013)
Pharmacological postconditioning with diazoxide attenuates
ischemia/reperfusion-induced injury in rat liver. Exp. Ther. Med.
5, 1169–1173 PubMed
96 Tao, X., Wan, X., Xu, Y., Xu, L., Qi, Y., Yin, L., Han, X., Lin, Y. and
Peng, J. (2014) Dioscin attenuates hepatic
ischemia–reperfusion injury in rats through inhibition of
oxidative–nitrative stress, inflammation and apoptosis.
Transplantation 98, 604–611 CrossRef PubMed
97 Zimmerman, M.A., Tak, E., Ehrentraut, S.F., Kaplan, M., Giebler,
A., Weng, T., Choi, D.S., Blackburn, M.R., Kam, I., Eltzschig,
H.K. and Grenz, A. (2013) Equilibrative nucleoside transporter
(ENT)-1-dependent elevation of extracellular adenosine protects
the liver during ischemia and reperfusion. Hepatology 58,
1766–1778 CrossRef PubMed
98 Uchiyama, M., Tojo, K., Yazawa, T., Ota, S., Goto, T. and
Kurahashi, K. (2015) Edaravone prevents lung injury induced by
hepatic ischemia–reperfusion. J. Surg. Res. 194, 551–7
CrossRef PubMed
99 Riehle, K.J., Hoagland, V., Benz, W., Campbell, J.S., Liggitt, D.H.
and Langdale, L.A. (2014) Hepatocellular heme oxygenase-1: a
potential mechanism of erythropoietin-mediated protection
after liver ischemia–reperfusion injury. Shock 42, 424–431
CrossRef PubMed
100 Shen, M., Lu, J., Dai, W., Wang , F., Xu, L., Chen, K., He, L.,
Cheng, P., Zhang, Y., Wang, C. et al. (2013) Ethyl pyruvate
ameliorates hepatic ischemia–reperfusion injury by inhibiting
intrinsic pathway of apoptosis and autophagy. Mediators
Inflamm. 2013, 461536 PubMed
101 Gupta, N.A., Kolachala, V.L., Jiang, R., Abramowsky, C., Shenoi,
A., Kosters, A., Pavuluri, H., Anania, F. and Kirk, A.D. (2014)
Mitigation of autophagy ameliorates hepatocellular damage
following ischemia–reperfusion injury in murine steatotic liver.
Am. J. Physiol. Gastrointest. Liver Physiol. 307, G1088–G1099
CrossRef PubMed
102 Rickenbacher, A., Jang , J.H., Limani, P., Ungethum, U.,
Lehmann, K., Oberkofler, C.E., Weber, A., Graf, R., Humar, B.
and Clavien, P.A. (2014) Fasting protects liver from ischemic
injury through Sirt1-mediated downregulation of circulating
HMGB1 in mice. J. Hepatol. 61, 301–308
CrossRef PubMed
103 Liu, A., Fang, H., Yang, Y., Sun, J., Fan, H., Liu, S., Dirsch, O.
and Dahmen, U. (2013) The fibrin-derived peptide bβ15–42
attenuates liver damage in a rat model of liver
ischemia/reperfusion injury. Shock 39, 397–403
CrossRef PubMed
104 Zhang, R., Zhang, L., Manaenko, A., Ye, Z., Liu, W. and Sun, X.
(2014) Helium preconditioning protects mouse liver against
ischemia and reperfusion injury through the PI3K/Akt pathway.
J. Hepatol. 61, 1048–1055 CrossRef PubMed
C
The Authors Journal compilation C
2015 Biochemical Society 359
J. Gracia-Sancho, A. Casillas-Ram´
ırez and C. Peralta
105 Huang, X., Gao, Y., Qin, J. and Lu, S. (2014) The role of
miR-34a in the hepatoprotective effect of hydrogen sulfide on
ischemia/reperfusion injury in young and old rats. PLoS ONE 9,
e113305 CrossRef PubMed
106 Shimada, S., Fukai, M., Wakayama, K., Ishikawa, T., Kobayashi,
N., Kimura, T., Yamashita, K., Kamiyama, T., Shimamura, T.,
Taketomi, A. and Todo, S. (2014) Hydrogen sulfide augments
survival signals in warm ischemia and reperfusion of the mouse
liver. Surg. Today, doi:10.1007/s00595-014-1064-4
107 Hanaoka, J., Shimada, M., Utsunomiya, T., Morine, Y., Imura,
S., Ikemoto, T., Mori, H., Sugimoto, K., Saito, Y., Yamada, S.
and Asanoma, M. (2014) Beneficial effects of enteral nutrition
containing with hydrolyzed whey peptide on warm
ischemia/reperfusion injury in the rat liver. Hepatol. Res. 44,
114–121 CrossRef PubMed
108 Nii, A., Utsunomiya, T., Shimada, M., Ikegami, T., Ishibashi, H.,
Imura, S., Morine, Y., Ikemoto, T., Sasaki, H. and Kawashima,
A. (2014) A hydrolyzed whey peptide-based diet ameliorates
hepatic ischemia–reperfusion injury in the rat nonalcoholic fatty
liver. Surg. Today 44, 2354–2360 CrossRef PubMed
109 Pan, S., Liu, L., Pan, H., Ma, Y., Wang, D., Kang, K., Wang, J.,
Sun, B., Sun, X. and Jiang, H. (2013) Protective effects of
hydroxytyrosol on liver ischemia/reperfusion injury in mice. Mol.
Nutr. Food Res. 57, 1218–1227 CrossRef PubMed
110 Losada, D.M., Chies, A.B., Feres, O., Chaib, E., D’Albuquerque,
L.A. and Castro-e-Silva, L.A. (2014) Effects of hyperbaric oxygen
therapy as hepatic preconditioning in rats submitted to hepatic
ischemia/reperfusion injury. Acta Cir. Bras. 29, 61–66
CrossRef PubMed
111 Hartmann, P., Fet, N., Garab, D., Szabo, A., Kaszaki, J.,
Srinivasan, P.K., Tolba, R.H. and Boros, M. (2014)
l-α-glycerylphosphorylcholine reduces the microcirculatory
dysfunction and nicotinamide adenine dinucleotide
phosphate-oxidase type 4 induction after partial hepatic
ischemia in rats. J. Surg. Res. 189, 32–40
CrossRef PubMed
112 Ibrahim, M.A., Abdel-Gaber, S.A., Amin, E.F., Ibrahim, S.A.,
Mohammed, R.K. and Abdelrahman, A.M. (2014) Molecular
mechanisms contributing to the protective effect of
levosimendan in liver ischemia–reperfusion injury. Eur. J.
Pharmacol. 741, 64–73 CrossRef PubMed
113 Onody, P., Stangl, R., Fulop, A., Rosero, O., Garbaisz, D., Turoczi,
Z., Lotz, G., Rakonczay, Jr, Z., Balla, Z., Hegedus, V. et al.
(2013) Levosimendan: a cardiovascular drug to prevent liver
ischemia–reperfusion injury? PLoS ONE 8, e73758
CrossRef PubMed
114 Mahmoud, M.F., Gamal, S. and El-Fayoumi, H.M. (2014)
Limonin attenuates hepatocellular injury following liver ischemia
and reperfusion in rats via Toll-like receptor dependent pathway.
Eur. J. Pharmacol. 740, 676–682 CrossRef PubMed
115 Liu, A., Fang, H., Dahmen, U. and Dirsch, O. (2013) Chronic
lithium treatment protects against liver ischemia/reperfusion
injury in rats. Liver Transpl. 19, 762–772
CrossRef PubMed
116 Koh, E.J., Yoon, S.J. and Lee, S.M. (2013) Losar tan protects
liver against ischaemia/reperfusion injury through PPAR-γ
activation and receptor for advanced glycation end-products
down-regulation. Br. J. Pharmacol. 169, 1404–1416
CrossRef PubMed
117 Rao, J., Qin, J., Qian, X., Lu, L., Wang, P., Wu, Z., Zhai, Y.,
Zhang, F., Li, G. and Wang, X. (2013) Lipopolysaccharide
preconditioning protects hepatocytes from
ischemia/reperfusion injury (IRI) through inhibiting ATF4–CHOP
pathway in mice. PLoS ONE 8, e65568 CrossRef PubMed
118 Takhtfooladi, M.A., Takhtfooladi, H.A. and Khansari, M. (2014)
The effects of low-intensity laser therapy on hepatic
ischemia–reperfusion injury in a rat model. Lasers Med. Sci.
29, 1887–1893 CrossRef PubMed
119 Kireev, R., Bitoun, S., Cuesta, S., Tejerina, A., Ibarrola, C.,
Moreno, E., Vara, E. and Tresguerres, J.A. (2013) Melatonin
treatment protects liver of Zucker rats after
ischemia/reperfusion by diminishing oxidative stress and
apoptosis. Eur. J. Pharmacol. 701, 185–193
CrossRef PubMed
120 Li, L., Li, G., Yu, C., Shen, Z., Xu, C., Feng, Z., Zhang, X. and Li,
Y. (2013) A role of microRNA-370 in hepatic
ischaemia–reperfusion injury by targeting transforming growth
factor-βreceptor II. Liver Int. 35, 1124–1132 CrossRef
121 Li, Y., Li, T., Qi, H. and Yuan, F. (2015) Minocycline protects
against hepatic ischemia/reperfusion injury in a rat model.
Biomed. Rep. 3, 19–24 PubMed
122 Sun, Y., Pu, L.Y., Lu, L., Wang, X.H., Zhang, F. and Rao, J.H.
(2014) N-acetylcysteine attenuates
reactive-oxygen-species-mediated endoplasmic reticulum stress
during liver ischemia–reperfusion injury. World J. Gastroenterol.
20, 15289–15298 CrossRef PubMed
123 Katsumi, H., Fukui, K., Sato, K., Maruyama, S., Yamashita, S.,
Mizumoto, E., Kusamori, K., Oyama, M., Sano, M., Sakane, T.
and Yamamoto, A. (2014) Pharmacokinetics and preventive
effects of platinum nanoparticles as reactive oxygen species
scavengers on hepatic ischemia/reperfusion injury in mice.
Metallomics 6, 1050–1056 CrossRef PubMed
124 Zhao, G., Ma, H., Shen, X., Xu, G.F., Zhu, Y.L., Chen, B., Tie, R.,
Qu, P., Lv, Y., Zhang, H. and Yu, J. (2013) Role of glycogen
synthase kinase 3βin protective effect of propofol against
hepatic ischemia–reperfusion injury. J. Surg. Res. 185,
388–398 CrossRef PubMed
125 Mende, K., Reifart, J., Rosentreter, D., Manukyan, D., Mayr, D.,
Krombach, F., Rentsch, M. and Khandoga, A. (2014) Targeting
platelet migration in the postischemic liver by blocking
protease-activated receptor 4. Transplantation 97, 154–160
CrossRef PubMed
126 Zhu, J., Lu, T., Yue, S., Shen, X., Gao, F., Busuttil, R.W.,
Kupiec-Weglinski, J.W., Xia, Q. and Zhai, Y. (2015) Rapamycin
protection of livers from ischemia and reperfusion injury is
dependent on both autophagy induction and mammalian target
of rapamycin complex 2–Akt activation. Transplantation 99,
48–55 CrossRef PubMed
127 Suyavaran, A., Ramamurthy, C., Mareeswaran, R., Subastri, A.,
Lokeswara, R.P. and Thirunavukkarasu, C. (2015) TNF-α
suppression by glutathione preconditioning attenuates hepatic
ischemia reperfusion injury in young and aged rats. Inflamm.
Res. 64, 71–81 CrossRef PubMed
128 Kuroda, S., Tashiro, H., Kimura, Y., Hirata, K., Tsutada, M.,
Mikuriya, Y., Kobayashi, T., Amano, H., Tanaka, Y. and Ohdan, H.
(2015) Rho-kinase inhibitor targeting liver prevents ischemic
reperfusion injury in steatotic liver without major systemic
adverse in rat. Liver Transpl. 21, 123–131 CrossRef PubMed
129 Sanches, S.C., Ramalho, L.N., Mendes-Braz, M., Terra, V.A.,
Cecchini, R., Augusto, M.J. and Ramalho, F.S. (2014) Riboflavin
(vitamin B-2) reduces hepatocellular injury following liver
ischaemia and reperfusion in mice. Food Chem. Toxicol. 67,
65–71 CrossRef PubMed
130 Ramalho, L.N., Pasta, A.A., Terra, V.A., Augusto, M.J., Sanches,
S.C., Souza-Neto, F.P., Cecchini, R., Gulin, F. and Ramalho, F.S.
(2014) Rosmarinic acid attenuates hepatic ischemia and
reperfusion injury in rats. Food Chem. Toxicol. 74C, 270–278
CrossRef
131 Balzan, S.M., Gava, V.G., Rieger, A., Pra, D., Trombini, L.,
Zenkner, F.F., Horta, J.A., Azambuja, G., Schopf, L. and de
Souza, P.L. (2014) Ischemic versus pharmacologic hepatic
preconditioning. J. Surg. Res. 191, 134–139
CrossRef PubMed
132 Zhou, S.P., Jiang, P., Liu, L. and Liu, H. (2013) Protective effect
of sevoflurane on hepatic ischaemia/reperfusion injury in the
rat: a dose–response study. Eur. J. Anaesthesiol. 30, 612–617
CrossRef
360 C
The Authors Journal compilation C
2015 Biochemical Society
Underlying mechanisms of hepatic I/R injury
133 Savvanis, S., Nastos, C., Tasoulis, M.K., Papoutsidakis, N.,
Demonakou, M., Karmaniolou, I., Arkadopoulos, N., Smyrniotis,
V. and Theodoraki, K. (2014) Sildenafil attenuates
hepatocellular injury after liver ischemia reperfusion in rats: a
preliminary study. Oxid. Med. Cell Longev. 2014, 161942
CrossRef PubMed
134 Sakai, S., Tajima, H., Miyashita, T., Nakanuma, S., Makino, I.,
Hayashi, H., Nakagawara, H., Kitagawa, H., Fushida, S.,
Fujimura, T. et al. (2014) Sivelestat sodium hydrate inhibits
neutrophil migration to the vessel wall and suppresses hepatic
ischemia–reperfusion injury. Dig. Dis. Sci. 59, 787–794
CrossRef PubMed
135 Tanemura, A., Kuriyama, N., Azumi, Y., Ohsawa, I., Kishiwada,
M., Mizuno, S., Usui, M., Sakurai, H., Tabata, M. and Isaji, S.
(2014) Thrombomodulin administration attenuates
ischemia–reperfusion injury of the remnant liver after 70 %
hepatectomy in rats: simulated model of small-for-size graft in
living donor liver transplantation. Transplant. Proc. 46,
1107–1111 CrossRef PubMed
136 Ji, H., Zhang, Y., Liu, Y., Shen, X.D., Gao, F., Nguyen, T.T.,
Busuttil, R.W., Waschek, J.A. and Kupiec-Weglinski, J.W. (2013)
Vasoactive intestinal peptide attenuates liver
ischemia/reperfusion injury in mice via the cyclic adenosine
monophosphate–protein kinase A pathway. Liver Transpl. 19,
945–956 CrossRef PubMed
137 Park, J., Kang, J.W. and Lee, S.M. (2013) Activation of the
cholinergic anti-inflammatory pathway by nicotine attenuates
hepatic ischemia/reperfusion injury via heme oxygenase-1
induction. Eur. J. Pharmacol. 707, 61–70
CrossRef PubMed
138 Li, F., Chen, Z., Pan, Q., Fu, S., Lin, F., Ren, H., Han, H., Billiar,
T.R., Sun, F. and Li, Q. (2013) The protective effect of
PNU-282987, a selective α7 nicotinic acetylcholine
receptor agonist, on the hepatic ischemia–reperfusion
injury is associated with the inhibition of high-mobility
group box 1 protein expression and nuclear factor κB
activation in mice. Shock 39, 197–203
CrossRef PubMed
139 Raptis, D.A., Limani, P., Jang, J.H., Ungethum, U., Tschuor, C.,
Graf, R., Humar, B. and Clavien, P.A. (2014) GPR120 on Kupffer
cells mediates hepatoprotective effects of ω3-fatty acids. J.
Hepatol. 60, 625–632 CrossRef PubMed
140 Kim, K., Jung, N., Lee, K., Choi, J., Kim, S., Jun, J., Kim, E. and
Kim, D. (2013) Dietary ω3 polyunsaturated fatty acids
attenuate hepatic ischemia/reperfusion injury in rats by
modulating Toll-like receptor recruitment into lipid rafts. Clin.
Nutr. 32, 855–862 CrossRef PubMed
141 Saito, Y., Shimada, M., Utsunomiya, T., Ikemoto, T., Yamada, S.,
Morine, Y., Imura, S., Mori, H., Sugimoto, K., Iwahashi, S. and
Asanoma, M. (2013) The protective effect of adipose-derived
stem cells against liver injury by trophic molecules. J. Surg.
Res. 180, 162–168 CrossRef PubMed
142 Rao, J., Yue, S., Fu, Y., Zhu, J., Wang, X., Busuttil, R.W.,
Kupiec-Weglinski, J.W., Lu, L. and Zhai, Y. (2014) ATF6
mediates a pro-inflammatory synergy between ER stress and
TLR activation in the pathogenesis of liver
ischemia–reperfusion injury. Am. J. Transplant. 14, 1552–1561
CrossRef PubMed
143 Jin, G., Qiu, G., Wu, D., Hu, Y., Qiao, P., Fan, C. and Gao, F.
(2013) Allogeneic bone marrow-derived mesenchymal stem
cells attenuate hepatic ischemia–reperfusion injury by
suppressing oxidative stress and inhibiting apoptosis in rats.
Int. J. Mol. Med. 31, 1395–1401 PubMed
144 Cai, C., Shi, X., Korff, S., Zhang, J., Loughran, P.A., Ruan, X.,
Zhang, Y., Liu, L. and Billiar, T.R. (2013) CD14 contributes to
warm hepatic ischemia–reperfusion injury in mice. Shock 40,
115–121 CrossRef PubMed
145 Feng, M., Wang, Q., Wang, H., Wang, M., Guan, W. and Lu, L.
(2014) Adoptive transfer of hepatic stellate cells ameliorates
liver ischemia reperfusion injury through enriching regulatory T
cells. Int. Immunopharmacol. 19, 267–274
CrossRef PubMed
146 Saidi, R.F., Rajeshkumar, B., Shariftabrizi, A., Bogdanov, A.A.,
Zheng, S., Dresser, K. and Walter, O. (2014) Human
adipose-derived mesenchymal stem cells attenuate liver
ischemia–reperfusion injury and promote liver regeneration.
Surgery 156, 1225–1231 CrossRef PubMed
147 Wang, P.X., Zhang, R., Huang, L., Zhu, L.H., Jiang, D.S., Chen,
H.Z., Zhang, Y., Tian, S., Zhang , X.F., Zhang, X.D. et al. (2015)
Interferon regulatory factor 9 is a key mediator of hepatic
ischemia/reperfusion injury. J. Hepatol. 62, 111–120
CrossRef PubMed
148 Lin, H.C., Liu, S.Y., Lai, H.S. and Lai, I.R. (2013) Isolated
mitochondria infusion mitigates ischemia–reperfusion
injury of the liver in rats. Shock 39, 304–310
CrossRef PubMed
149 Chen, Z., Jia, S., Li, D., Cai, J., Tu, J., Geng, B., Guan, Y., Cui,
Q. and Yang, J. (2013) Silencing of long noncoding RNA
AK139328 attenuates ischemia/reperfusion
injury in mouse livers. PLoS ONE 8, e80817
CrossRef PubMed
150 Fu, J., Zhang, H., Zhuang, Y., Liu, H., Shi, Q., Li, D. and Ju, X.
(2014) The role of N-acetyltransferase 8 in mesenchymal stem
cell-based therapy for liver ischemia/reperfusion injury in rats.
PLoS ONE 9, e103355 CrossRef PubMed
151 Li, J., Ke, W., Zhou, Q., Wu, Y., Luo, H., Zhou, H., Yang, B., Guo,
Y., Zheng, Q. and Zhang , Y. (2014) Tumour necrosis factor-α
promotes liver ischaemia–reperfusion injury through the
PGC-1α/Mfn2 pathway. J. Cell. Mol. Med. 18, 1863–1873
CrossRef PubMed
152 Yue, S., Rao, J., Zhu, J., Busuttil, R.W., Kupiec-Weglinski, J.W.,
Lu, L., Wang, X. and Zhai, Y. (2014) Myeloid PTEN deficiency
protects livers from ischemia reperfusion injury by facilitating
M2 macrophage differentiation. J. Immunol. 192, 5343–5353
CrossRef PubMed
153 Schlegel, M., Granja, T., Kaiser, S., Korner, A., Henes, J., Konig,
K., Straub, A., Rosenberger, P. and Mirakaj, V. (2014) Inhibition
of neogenin dampens hepatic ischemia–reperfusion injury. Crit.
Care Med. 42, e610–e619 CrossRef PubMed
154 Inoue, Y., Shirasuna, K., Kimura, H., Usui, F., Kawashima, A.,
Karasawa, T., Tago, K., Dezaki, K., Nishimura, S., Sagara, J.
et al. (2014) NLRP3 regulates neutrophil functions and
contributes to hepatic ischemia–reperfusion injury
independently of inflammasomes. J. Immunol. 192,
4342–4351 CrossRef PubMed
155 Ji, H., Liu, Y., Zhang, Y., Shen, X.D., Gao, F., Busuttil, R.W.,
Kuchroo, V.K. and Kupiec-Weglinski, J.W. (2014) T-cell
immunoglobulin and mucin domain 4 (TIM-4) signaling in innate
immune-mediated liver ischemia–reperfusion injury. Hepatology
60, 2052–2064 CrossRef PubMed
156 Nace, G.W., Huang, H., Klune, J.R., Eid, R.E., Rosborough, B.R.,
Korff, S., Li, S., Shapiro, R.A., Stolz, D.B., Sodhi, C.P. et al.
(2013) Cellular-specific role of Toll-like receptor 4 in hepatic
ischemia–reperfusion injury in mice. Hepatology 58, 374–387
CrossRef PubMed
157 Meimei, H., Dejin, M., Erzhen, C., Minmin, S., Songyao, J.,
Jianfang, L. and Hao, C. (2014) Inhibiting the Toll-like receptor
4 Toll/interleukin-1 receptor domain protects against hepatic
warm ischemia and reperfusion injury in mice. Crit. Care Med.
42, e123–e131 CrossRef PubMed
158 Liu, A., Fang, H., Wei, W., Dirsch, O. and Dahmen, U. (2014)
Ischemic preconditioning protects against liver
ischemia/reperfusion injury via heme oxygenase-1-mediated
autophagy. Crit. Care Med. 42, e762–e771
CrossRef PubMed
C
The Authors Journal compilation C
2015 Biochemical Society 361
J. Gracia-Sancho, A. Casillas-Ram´
ırez and C. Peralta
159 Pantazi, E., Zaouali, M.A., Bejaoui, M., Serafin, A., Folch-Puy, E.,
Petegnief, V., De Vera, N., Ben Abdennebi, H., Rimola, A. and
Rosello-Catafau, J. (2014) Silent information regulator 1
protects the liver against ischemia–reperfusion injury:
implications in steatotic liver ischemic preconditioning. Transpl.
Int. 27, 493–503 CrossRef PubMed
160 Fu, H., Xu, H., Chen, H., Li, Y., Li, W., Zhu, Q., Zhang, Q., Yuan,
H., Liu, F., Wang, Q., Miao, M. and Shi, X. (2014) Inhibition of
glycogen synthase kinase 3 ameliorates liver ischemia/
reperfusion injury via an energy-dependent mitochondrial
mechanism. J. Hepatol. 61, 816–824 CrossRef PubMed
161 Czigany, Z., Turoczi, Z., Kleiner, D., Lotz, G., Homeyer, A.,
Harsanyi, L. and Szijarto, A. (2015) Neural elements behind the
hepatoprotection of remote preconditioning. J. Surg. Res. 193,
642–651 CrossRef PubMed
162 Kageyama, S., Hata, K., Tanaka, H., Hirao, H., Kubota, T.,
Okamura, Y., Iwaisako, K., Takada, Y. and Uemoto, S. (2015)
Intestinal Ischemic preconditioning ameliorates hepatic
ischemia reperfusion injury in rats: role of heme oxygenase-1 in
the second-window of protection. Liver Transpl. 21, 112–122
CrossRef PubMed
163 Wang, Y., Shen, J., Xiong, X., Xu, Y., Zhang, H., Huang, C., Tian,
Y., Jiao, C., Wang, X. and Li, X. (2014) Remote ischemic
preconditioning protects against liver ischemia–reperfusion
injury via heme oxygenase-1-induced autophagy. PLoS ONE 9,
e98834 CrossRef PubMed
164 Garab, D., Fet, N., Szabo, A., Tolba, R.H., Boros, M. and
Hartmann, P. (2014) Remote ischemic preconditioning
differentially affects NADPH oxidase isoforms during hepatic
ischemia–reperfusion. Life Sci. 105, 14–21 CrossRef PubMed
Received 24 March 2015; accepted 15 April 2015
Published on the Internet 21 May 2015, doi: 10.1042/CS20150223
362 C
The Authors Journal compilation C
2015 Biochemical Society
... Cold ischemia occurs during organ retrieval, when the liver is cooled, perfused and then stored in a cold preservation solution. Rewarming ischemia is encountered during back-table graft preparation and also at the implementation stage while performing the vascular anastomoses before reperfusion (2). Despite this fact, the preservation method for organ transplantation has been left unchanged for many years and simple static cold storage still remains the gold standard worldwide. ...
... 1 patient characteristics: age, gender, height, weight, BMI, blood group, cause of cirrhosis, indication for transplantation, medical history (diabetes mellitus, arterial hypertension, transjugular intrahepatic portosystemic shunt) pretransplant status of residence (home, hospital ward or intensive care unit (ICU)). 2 Randomization will be performed after the harvesting team has macroscopically assessed the graft and con rmed that the graft will be harvested. After checking the inclusion and non-inclusion criteria an ...
Preprint
Full-text available
• Background: Given the scarce donor supply, an increasing number of so-called marginal or extended criteria donor (ECD) organs are used for liver transplantation. These ECD liver grafts are however known to be associated with a higher rate of early allograft dysfunction and primary non-function because of a greater vulnerability to ischemia-reperfusion injury. The end-ischemic Hypothermic Oxygenated Machine Perfusion (HOPE) technique may improve outcomes of liver transplantation with ECD grafts by decreasing reperfusion injury. • Methods: HOPExt trial is a comparative open-label, multicenter, national, prospective, randomized, controlled study, in two parallel groups, using static cold storage, the gold standard procedure, as control. The trial will enroll adult patients on the transplant waiting list for liver failure or liver cirrhosis and/or liver malignancy requiring liver transplantation and receiving an ECD liver graft from a brain-dead donor. In the experimental group, ECD liver grafts will first undergo a classical static cold (4°C) storage followed by a hypothermic oxygenated perfusion (HOPE) for a period of 1 to 4 hours. The control group will consist of the classic static cold storage which is the gold standard procedure in liver transplantation. The primary objective of this trial is to study the efficacy of HOPE used before transplantation of ECD liver grafts from brain-dead donors in reducing postoperative early allograft dysfunction within the first 7 postoperative days compared to simple cold static storage. • Discussion: We present in this protocol all study procedures in regard to the achievement of the HOPExt trial, to prevent biased analysis of trial outcomes and improve the transparency of the trial results. Enrollment of patients in the HOPExt trial has started on June 31, 2021, and is ongoing • Trial registration: ClinicalTrials.gov NCT03929523. Registered on April 29, 2019, before the start of inclusion.
... In recent years, precision medicine research is focused on unraveling the molecular pathways associated with specific liver graft pathologies [26]. A primary objective of this field of research is the investigation of the potential role of liver-specific biomarkers in the non-invasive accurate diagnosis. ...
Chapter
Full-text available
Donor-derived cell-free DNA (Dd-cfDNA) is a novel biomarker with many diagnostic applications in various areas of medicine and particularly transplantation. This biomarker is derived from donor cells that have undergone apoptosis or cell death and thus reflects possible graft damage. Regarding the field of liver transplantation, dd-cfDNA can contribute to the diagnosis of complications that include signs of rejection or other types of possible graft injury. Measurements of dd-cfDNA also depend on the graft’s size and origin; therefore, these data should be considered for the estimation and explanation of dd-cfDNA values. Despite the utility of this novel diagnostic technique, it comes with some limitations and application exclusions, such as cases where there is a blood relation between the donor and recipient. Combination of dd-cfDNA evaluation with the assessment of other currently used biomarkers, such as liver enzymes, or other novel biomarkers can result to high diagnostic value.
... SCS has yielded satisfactory outcomes following LT with relative ease and low cost, but its use has been limited in high-risk or marginal organs [4]. Marginal livers are especially vulnerable to ischemia-reperfusion injury (IRI), which leads to an increased risk of death [16,17]. ...
Article
Full-text available
Despite improvements in short-term and long-term outcomes of liver transplant patients, the discrepancy between the number of available livers and transplant candidates continues to increase. The use of expanded criteria donors is one strategy that can be used to address donor shortages. In recent years, preservation strategies such as normothermic machine perfusion (NMP) have been explored to improve the preservation of organs and test their viability before transplantation. We reviewed the recent literature and trials assessing the use of NMP in the setting of liver transplantation. Multiple feasibility trials have demonstrated the clinical prospect of NMP and proved its numerous advantages compared to conventional static cold storage. These advantages include preservation and viability assessment of high-risk donor allografts and grafts that would have otherwise been discarded. This review aims to address the topic of liver NMP in the setting of current and future applications in the setting of extended criteria donor grafts.
... The pathogenesis of HIRI involves many different mechanisms, mainly including oxidative stress, inflammation, apoptosis, and autophagy. These pathological processes ultimately lead to cell death [67]. As expected, NF-κB pathway is involved in HIRI pathological processes through multiple mechanisms [30] (Fig. 5). ...
Article
Full-text available
Hepatic ischemia–reperfusion injury (HIRI) is a major complication of liver trauma, resection, and transplantation that can lead to liver dysfunction and failure. Scholars have proposed a variety of liver protection methods aimed at reducing ischemia–reperfusion damage, but there is still a lack of effective treatment methods, which urgently needs to find new effective treatment methods for patients. Many studies have reported that signaling pathway plays a key role in HIRI pathological process and liver function recovery mechanism, among which nuclear transfer factor-κB (NF-κB) signaling pathway is one of the signal transduction closely related to disease. NF-κB pathway is closely related to HIRI pathologic process, and inhibition of this pathway can delay oxidative stress, inflammatory response, cell death, and mitochondrial dysfunction. In addition, NF-κB can also interact with PI3K/Akt, MAPK, and Nrf2 signaling pathways to participate in HIRI regulation. Based on the role of NF-κB pathway in HIRI, it may be a potential target pathway for HIRI. This review emphasizes the role of inhibiting the NF-κB signaling pathway in oxidative stress, inflammatory response, cell death, and mitochondrial dysfunction in HIRI, as well as the effects of related drugs or inhibitors targeting NF-κB on HIRI. The objective of this review is to elucidate the role and mechanism of NF-κB pathway in HIRI, emphasize the important role of NF-κB pathway in the prevention and treatment of HIRI, and provide a theoretical basis for the target NF-κB pathway as a therapy for HIRI.
... Cold ischemia occurs during organ retrieval, when the liver is cooled, perfused and then stored in a cold preservation solution. Rewarming ischemia is encountered during back-table graft preparation and also at the implementation stage while performing the vascular anastomoses before reperfusion [2]. Despite this fact, the preservation method for organ transplantation has been left unchanged for many years and simple static cold storage still remains the gold standard worldwide. ...
Article
Full-text available
Background Given the scarce donor supply, an increasing number of so-called marginal or extended criteria donor (ECD) organs are used for liver transplantation. These ECD liver grafts are however known to be associated with a higher rate of early allograft dysfunction and primary non-function because of a greater vulnerability to ischemia–reperfusion injury. The end-ischemic hypothermic oxygenated machine perfusion (HOPE) technique may improve outcomes of liver transplantation with ECD grafts by decreasing reperfusion injury. Methods HOPExt trial is a comparative open-label, multicenter, national, prospective, randomized, controlled study, in two parallel groups, using static cold storage, the gold standard procedure, as control. The trial will enroll adult patients on the transplant waiting list for liver failure or liver cirrhosis and/or liver malignancy requiring liver transplantation and receiving an ECD liver graft from a brain-dead donor. In the experimental group, ECD liver grafts will first undergo a classical static cold (4 °C) storage followed by a hypothermic oxygenated perfusion (HOPE) for a period of 1 to 4 h. The control group will consist of the classic static cold storage which is the gold standard procedure in liver transplantation. The primary objective of this trial is to study the efficacy of HOPE used before transplantation of ECD liver grafts from brain-dead donors in reducing postoperative early allograft dysfunction within the first 7 postoperative days compared to simple cold static storage. Discussion We present in this protocol all study procedures in regard to the achievement of the HOPExt trial, to prevent biased analysis of trial outcomes and improve the transparency of the trial results. Enrollment of patients in the HOPExt trial has started on September 10, 2019, and is ongoing. Trial registration ClinicalTrials.gov NCT03929523. Registered on April 29, 2019, before the start of inclusion.
... Hepatic ischemia-reperfusion injury (HIRI) is the result of temporary decrease in tissue blood supply due to vascular reconstruction. 59 In 2014, Angeli et al 60 proved that ferroptosis was a mechanism included in HIRI. Yamada et al 61 analyzed clinical data from pediatric liver transplantation patients and found that iron overload was closely associated with poor prognosis, suggesting the potential effect of ferroptosis in HIRI. ...
Article
Full-text available
Ferroptosis is a recently identified iron-dependent form of intracellular lipid peroxide accumulation-mediated cell death. Different from other types of cell death mechanisms, it exhibits distinct biological and morphological features characterized by the loss of lipid peroxidase repair activity caused by glutathione peroxidase 4, the presence of redox-active iron, and the oxidation of phospholipids-containing polyunsaturated fatty acids. In recent years, studies have shown that ferroptosis plays a key role in various liver diseases such as alcoholic liver injury, non-alcoholic steatohepatitis, liver cirrhosis, and liver cancer. However, the mechanism of ferroptosis and its regulation on chronic liver disease are controversial among different types of cells in the liver. Herein, we summarize the current studies on mechanism of ferroptosis in chronic liver disease, aiming to outline the blueprint of ferroptosis as an effective option for chronic liver disease therapy.
Article
Full-text available
Introduction Brain death (BD) and steatosis are both risk factors for organ dysfunction or failure in liver transplantation (LT) Material and methods Here, we examine the role of interleukin 6 (IL- 6) and IL-10 in LT of both non-steatotic and steatotic liver recovered from donors after brain death (DBDs), as well as the molecular signaling pathways underlying the effects of such cytokines. Results BD reduced IL-6 levels only in nonsteatotic grafts, and diminished IL-10 levels only in steatotic ones. In both graft types, BD increased IL-1β, which was associated with hepatic inflammation and damage. IL-6 administration reduced IL-1β only in non-steatotic grafts and protected them against damage and inflammation. Concordantly, IL-1β inhibition via treatment with an IL-1 receptor antagonist caused the same benefits in non-steatotic grafts. Treatment with IL-10 decreased IL-1β only in steatotic grafts and reduced injury and inflammation specifically in this graft type. Blockading the IL-1β effects also reduced damage and inflammation in steatotic grafts. Also, blockade of IL-1β action diminished hepatic cAMP in both types of livers, and this was associated with a reduction in liver injury and inflammation, then pointing to IL-1β regulating cAMP generation under LT and BD conditions. Additionally, the involvement of nitric oxide (NO) in the effects of interleukins was evaluated. Pharmacological inhibition of NO in LT from DBDs prompted even more evident reductions of IL-6 or IL-10 in non-steatotic and steatotic grafts, respectively. This exacerbated the already high levels of IL-1β seen in LT from DBDs, causing worse damage and inflammation in both graft types. The administration of NO donors to non-steatotic grafts potentiated the beneficial effects of endogenous NO, since it increased IL-6 levels, and reduced IL-1β, inflammation, and damage. However, treatment with NO donors in steatotic grafts did not modify IL-10 or IL-1β levels, but induced more injurious effects tan the induction of BD alone, characterized by increased nitrotyrosine, lipid peroxidation, inflammation, and hepatic damage. Conclusion Our study thus highlights the specificity of new signaling pathways in LT from DBDs: NO–IL-6–IL-1β in non-steatotic livers and NO–IL-10–IL-1β in steatotic ones. This opens up new therapeutic targets that could be useful in clinical LT.
Article
Hepatic ischemia-reperfusion (I/R) injury is a multifactorial process caused by transient tissue hypoxia and the following reoxygenation, commonly occurring in liver transplantation and hepatectomy. Hepatic I/R can induce a systemic inflammatory response, liver dysfunction, or even multiple organ failure. Although we have previously reported that taurine could attenuate acute liver injury after hepatic I/R, only a tiny proportion of the systemically injected taurine could reach the targeted organ and tissues. In this present study, we prepared taurine nanoparticles (Nano-taurine) by coating taurine with neutrophil membranes and investigated the protective effects of Nano-taurine against I/R-induced injury and the underlying mechanisms. Our results showed that Nano-taurine restored liver function by declining AST and ALT levels and reducing histology damage. Nano-taurine decreased inflammatory cytokines, including interleukin (IL)-6, tumor necrosis factor (TNF)-α, intercellular adhesion molecule (ICAM)-1, NLR pyrin domain containing 3 (NLRP3) and apoptosis-associated speck-like protein containing CARD (ASC) and oxidants including superoxide dismutase (SOD), malondialdehyde (MDA), glutathione (GSH), catalase (CAT) and reactive oxygen species (ROS), exhibiting anti-inflammatory and antioxidant properties. The expression of solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) was increased, while prostaglandin-endoperoxide synthase 2 (Ptgs2) was decreased upon administration of Nano-taurine, suggesting that inhibiting ferroptosis may be involved in the mechanism during hepatic I/R injury. These results suggest that Nano-taurine have a targeted therapeutic effect on hepatic I/R injury by inhibiting inflammation, oxidative stress, and ferroptosis.
Article
Full-text available
We describe a new preservation modality combining machine perfusion (MP) at subnormothermic conditions (21°C) with a new hemoglobin-based oxygen carrier (HBOC) solution. MP (n = 6) was compared to cold static preservation (CSP; n = 6) in porcine orthotopic liver transplants after 9 h of cold ischemia and 5-day follow-up. Recipients' peripheral blood, serial liver biopsies, preservation solutions and bile specimens were collected before, during and after liver preservation. Clinical laboratorial and histological analyses were performed in addition to mitochondrial functional assays, transcriptomic, metabolomic and inflammatory mediator analyses. Compared with CSP, MP animals had: (1) significantly higher survival (100% vs. 33%; p < 0.05); (2) superior graft function (p < 0.05); (3) eight times higher hepatic O2 delivery than O2 consumption (0.78 mL O2 /g/h vs. 0.096 mL O2 /g/h) during MP; and (4) significantly greater bile production (MP = 378.5 ± 179.7; CS = 151.6 ± 116.85). MP down-regulated interferon (IFN)-α and IFN-γ in liver tissue. MP allografts cleared lactate, produced urea, sustained gluconeogenesis and produced hydrophilic bile after reperfusion. Enhanced oxygenation under subnormothermic conditions triggers regenerative and cell protective responses resulting in improved allograft function. MP at 21°C with the HBOC solution significantly improves liver preservation compared to CSP. 2015 The Authors. American Journal of Transplantation Published by The American Society of Transplantation and the American Society of Transplant Surgeons.
Article
Full-text available
Aim: To investigate the effect of diazoxide administration on liver ischemia/reperfusion injury. Methods: Wistar male rats underwent partial liver ischemia performed by clamping the pedicle from the medium and left anterior lateral segments for 1 h under mechanical ventilation. They were divided into 3 groups: Control Group, rats submitted to liver manipulation, Saline Group, rats received saline, and Diazoxide Group, rats received intravenous injection diazoxide (3.5 mg/kg) 15 min before liver reperfusion. 4 h and 24 h after reperfusion, blood was collected for determination of aspartate transaminase (AST), alanine transaminase (ALT), tumor necrosis factor (TNF-α), interleukin-6 (IL-6), interleukin-10 (IL-10), nitrite/nitrate, creatinine and tumor growth factor-β1 (TGF-β1). Liver tissues were assembled for mitochondrial oxidation and phosphorylation, malondialdehyde (MDA) content, and histologic analysis. Pulmonary vascular permeability and myeloperoxidase (MPO) were also determined. Results: Four hours after reperfusion the diazoxide group presented with significant reduction of AST (2009 ± 257 U/L vs 3523 ± 424 U/L, P = 0.005); ALT (1794 ± 295 U/L vs 3316 ± 413 U/L, P = 0.005); TNF-α (17 ± 9 pg/mL vs 152 ± 43 pg/mL, P = 0.013; IL-6 (62 ± 18 pg/mL vs 281 ± 92 pg/mL); IL-10 (40 ± 9 pg/mL vs 78 ± 10 pg/mL P = 0.03), and nitrite/nitrate (3.8 ± 0.9 μmol/L vs 10.2 ± 2.4 μmol/L, P = 0.025) when compared to the saline group. A significant reduction in liver mitochondrial dysfunction was observed in the diazoxide group compared to the saline group (P < 0.05). No differences in liver MDA content, serum creatinine, pulmonary vascular permeability and MPO activity were observed between groups. Twenty four hours after reperfusion the diazoxide group showed a reduction of AST (495 ± 78 U/L vs 978 ± 192 U/L, P = 0.032); ALT (335 ± 59 U/L vs 742 ± 182 U/L, P = 0.048), and TGF-β1 (11 ± 1 ng/mL vs 17 ± 0.5 ng/mL, P = 0.004) serum levels when compared to the saline group. The control group did not present alterations when compared to the diazoxide and saline groups. Conclusion: Diazoxide maintains liver mitochondrial function, increases liver tolerance to ischemia/reperfusion injury, and reduces the systemic inflammatory response. These effects require further evaluation for using in a clinical setting.
Article
Full-text available
We showed previously that pretreatment of butyrate, which is an endogenous histone deacetylase (HDAC) inhibitor normally fermented from undigested fiber by intestinal microflora, seriously alleviated ischemia reperfusion (I/R)-induced liver injury by inhibiting the nuclear factor κB (NF-κB) pathway. The goal of this study was to investigate the effect of butyrate administrated at the onset of ischemia for HDAC inhibition in hepatic I/R injury. Sprague Dawley rats were subjected to warm ischemia for 60 min followed by 6 and 24 h of reperfusion. Butyrate was administrated at the onset of ischemia. Liver injury was evaluated by serum levels of aminotransferase, inflammatory factors, and histopathology. The levels of acetylated histone H3 and expression of heat shock protein (Hsp) 70 were measured by Western blot. After reperfusion, the levels of acetylated histone H3 significantly decreased. Butyrate treatment markedly prevented the reduction of acetylated histone H3 and upregulated the expression of Hsp70, thereby reducing liver injury. Our study demonstrated that I/R resulted in marked reduction of histone acetylation; butyrate exerted a great hepatoprotective effect through HDAC inhibition and Hsp70 induction.
Article
Full-text available
Hydrogen sulfide (H2S) can protect the liver against ischemia-reperfusion (I/R) injury. However, it is unknown whether H2S plays a role in the protection of hepatic I/R injury in both young and old patients. This study compared the protective effects of H2S in a rat model (young and old animals) of I/R injury and the mechanism underlying its effects. Young and old rats were assessed following an injection of NaHS. NaHS alone reduced hepatic I/R injury in the young rats by activating the nuclear erythroid-related factor 2 (Nrf2) signaling pathway, but it had little effect on the old rats. NaHS pretreatment decreased miR-34a expression in the hepatocytes of the young rats with hepatic I/R. Overexpresion of miR-34a decreased Nrf-2 and its downstream target expression, impairing the hepatoprotective effect of H2S on the young rats. More importantly, downregulation of miR-34a expression increased Nrf-2 and the expression of its downstream targets, enhancing the effect of H2S on hepatic I/R injury in the old rats. This study reveals the different effects of H2S on hepatic I/R injury in young and old rats and sheds light on the involvement of H2S in miR-34a modulation of the Nrf-2 pathway.
Article
Full-text available
Abstract Background and aim Hepatic ischemia reperfusion (I/R) stimulates Kupffer cells and initiates injury through tumor necrosis factor-α (TNF-α) upregulation. Aim of this study was to compare the variable effects of reduced glutathione (GSH) pre-treatment on I/R liver injury in young and aged rats. Methods Wistar male rats were sorted into young (groups I–III) and aged (groups IV–VI). All groups except sham (groups I and IV) were subjected to 90-min ischemia and 2-h reperfusion. The treatment groups received 200 mg/kg bwt (groups III and VI) of GSH, 30 min prior to I/R. Variable effects of GSH were studied by transaminase activities, thiobarbituric acid-reactive substances (TBARS), GSH level, GSH/oxidized GSH (GSSG) ratio, TNF-α level, apoptotic markers and confirmed by histopathological observations. Results Our findings revealed that I/R inflicted more liver damage in aged rats than young rats. The GSH treatment prior to surgery significantly lowered the serum transaminase activities, hepatic TBARS level and effectively restored the GSH/GSSG ratio in both young and aged rats more remarkably in the mitochondria. Western analysis depicted that the GSH treatment effectively suppressed TNF-α expression and apoptotic markers in both young and aged rats. These findings were further confirmed by terminal deoxynucleotide transferase dUTP nick end labeling assay and histopathological observations of liver sections of young and aged rats. Conclusion Restoration of GSH/GSSG ratio through GSH pre-conditioning inhibits TNF-α and apoptosis in hepatic I/R injury. Hence, GSH pre-conditioning may be utilized in both young and aged individuals during liver transplantation/surgery for better post-operative outcomes.
Article
Full-text available
Aim: To investigate the effects of N-acetylcysteine (NAC) on endoplasmic reticulum (ER) stress and tissue injury during liver ischemia reperfusion injury (IRI). Methods: Mice were injected with NAC (300 mg/kg) intraperitoneally 2 h before ischemia. Real-time polymerase chain reaction and western blotting determined ER stress molecules (GRP78, ATF4 and CHOP). To analyze the role of NAC in reactive oxygen species (ROS)-mediated ER stress and apoptosis, lactate dehydrogenase (LDH) was examined in cultured hepatocytes treated by H2O2 or thapsigargin (TG). Results: NAC treatment significantly reduced the level of ROS and attenuated ROS-induced liver injury after IRI, based on glutathione, malondialdehyde, serum alanine aminotransferase levels, and histopathology. ROS-mediated ER stress was significantly inhibited in NAC-treated mice. In addition, NAC treatment significantly reduced caspase-3 activity and apoptosis after reperfusion, which correlated with the protein expression of Bcl-2 and Bcl-xl. Similarly, NAC treatment significantly inhibited LDH release from hepatocytes treated by H2O2 or TG. Conclusion: This study provides new evidence for the protective effects of NAC treatment on hepatocytes during IRI. Through inhibition of ROS-mediated ER stress, NAC may be critical to inhibit the ER-stress-related apoptosis pathway.
Article
Hepatic ischemia/reperfusion (I/R) injury is a common clinical problem. The present study was conducted to investigate the protective effect and mechanism of minocycline (Mino), a tetracycline with anti-inflammatory and antioxidant properties, on I/R injury of liver in rats. In total, 54 male Sprague-Dawley rats were randomly divided into 3 groups with 18 rats in each: Sham-operated (control group), I/R model (I/R group) and Mino preconditioning groups (Mino group). The rats of the Mino group were administered Mino (45 mg/kg) by gastric irrigation at 36 h before surgery and were subsequently administered with 22.5 mg/kg every 12 h for the 36 h before surgery. The rats were sacrificed at 2, 6 and 24 h after reperfusion, and the serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST) and lactate dehydrogenase (LDH) were measured. Hematoxylin/eosin staining of liver tissues was performed to detect the rat liver histological changes and the grade of liver I/R injury (Suzuki's criteria); the levels of malondialdehyde (MDA) and myeloperoxidase (MPO) were determined by spectrophotometry; hepatic tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) mRNA were measured by quantitative polymerase chain reaction; the Dickkopf-1 (DKK-1) and β-catenin gene products of the liver were detected by western blot analysis. Mino treatment significantly ameliorated the I/R injury of the liver, as shown by decreased Suzuki scores and liver function (ALT, AST and LDH). After 2, 6 and 24 h reperfusion, compared to the I/R group the MDA and MPO levels of the Mino group decreased in the liver tissues and the levels of hepatic TNF-α and IL-1β mRNA were decreased too. The protein expression of hepatic DKK-1 decreased, whereas β-catenin increased, which indicates that the Wnt/β-catenin pathway has been activated. In conclusion, Mino protects the liver from I/R injury mainly through reducing oxidative stress and inhibiting the release of pro-inflammatory cytokines by activating the Wnt/β-catenin signaling pathway in the liver.
Article
Transient hepatic ischemia can cause significant liver injury. A central and early event in ischemia/reperfusion (I/R) injury is the impairment of mitochondria. The phospholipid cardiolipin (CL) is required for efficient mitochondrial function. The aim of this study was to analyze composition, content, and oxidation of CL in dependence of I/R stress. Therefore, we exposed rat livers to 20 min ischemia by interrupting the perfusion with Krebs-Ringer solution in situ. Tissue histology as well as increased activities of LDH, GLDH, and ASAT analysed in the efflux after 50 min reperfusion indicated impairment of the liver. For the analysis of local CL distribution the liver homogenate was separated according to density into 11 fractions. The fractions displayed different contents of CL and citrate synthase peaking at density of about 1.07 g/cm(3). Among the fractions, the distribution of molecular CL species significantly differed. I/R caused loss of about 30 % CL and 17 % citrate synthase activity. Further, I/R shifted the CL and citrate synthase activity profile toward lower densities. Oxidized CL was exclusively found in fractions with high CL and citrate synthase content after I/R stress. I/R treatment caused significant changes in the distribution of molecular CL species. Our data demonstrate that I/R causes significant decrease in CL content and increase of oxidized CL that may be of impact for impairment of mitochondrial function by I/R. These results lead to the suggestion that strategies supporting anti-oxidative defence and CL synthesis may be beneficial to reduce I/R injury of the liver.
Article
Objectives: Ischemic preconditioning exerts a protective effect in hepatic ischemia/reperfusion injury. The exact mechanism of ischemic preconditioning action remains largely unknown. Recent studies suggest that autophagy plays an important role in protecting against ischemia/reperfusion injury. However, the role of autophagy in ischemic preconditioning-afforded protection and its regulatory mechanisms in liver ischemia/reperfusion injury remain poorly understood. This study was designed to determine whether ischemic preconditioning could protect against liver ischemia/reperfusion injury via heme oxygenase-1-mediated autophagy. Design: Laboratory investigation. Setting: University animal research laboratory. Subjects: Male inbred Lewis rats and C57BL/6 mice. Interventions: Ischemic preconditioning was produced by 10 minutes of ischemia followed by 10 minutes of reperfusion prior to 60 minutes of ischemia. In a rat model of hepatic ischemia/reperfusion injury, rats were pretreated with wortmannin or rapamycin to evaluate the contribution of autophagy to the protective effects of ischemic preconditioning. Heme oxygenase-1 was inhibited with tin protoporphyrin IX. In a mouse model of hepatic ischemia/reperfusion injury, autophagy or heme oxygenase-1 was inhibited with vacuolar protein sorting 34 small interfering RNA or heme oxygenase-1 small interfering RNA, respectively. Measurements and main results: Ischemic preconditioning ameliorated liver ischemia/reperfusion injury, as indicated by lower serum aminotransferase levels, lower hepatic inflammatory cytokines, and less severe ischemia/reperfusion-associated histopathologic changes. Ischemic preconditioning treatment induced autophagy activation, as indicated by an increase of LC3-II, degradation of p62, and accumulation of autophagic vacuoles in response to ischemia/reperfusion injury. When ischemic preconditioning-induced autophagy was inhibited with wortmannin in rats or vacuolar protein sorting 34-specific small interfering RNA in mice, liver ischemia/reperfusion injury was worsened, whereas rapamycin treatment increased autophagy and mimicked the protective effects of ischemic preconditioning. Furthermore, ischemic preconditioning increased heme oxygenase-1 expression. The inhibition of heme oxygenase-1 with tin protoporphyrin IX in rats or heme oxygenase-1-specific small interfering RNA in mice decreased ischemic preconditioning-induced autophagy and diminished the protective effects of ischemic preconditioning against ischemia/reperfusion injury. Conclusions: Ischemic preconditioning protects against liver ischemia/reperfusion injury, at least in part, via heme oxygenase-1-mediated autophagy.