GASTROENTEROLOGY 2003;125:917–936
SPECIAL REPORTS AND REVIEWS Protective Strategies Against Ischemic Injury of the Liver NAZIA SELZNER, HANNES RUDIGER, ROLF GRAF, and PIERRE–ALAIN CLAVIEN Laboratory of Liver Transplantation and Hepatobiliary Surgery, Department of Surgery, University Hospital of Zurich, Zurich, Switzerland
This article summarizes strategies to protect the liver from injuries caused by ischemia and reperfusion. Three different sections (i.e., surgical and pharmacologic strategies and gene therapy) present approaches to enhance the survival and viability of the liver in various surgical procedures including liver transplantation. The first section reviews approaches using surgical interventions such as ischemic preconditioning and intermittent clamping. Their protective effects are discussed with respect to the mechanism of injury. In the second section, pharmacologic agents targeting microcirculation, oxidative stress, proteases, and inflammation are described. Mechanisms of injury and their suppression by a wide variety of drugs are discussed. The third section focuses on gene therapy. Potential target genes have been identified (e.g., superoxide dismutase or heme oxygenase). Animal experiments in which the liver injury is reduced successfully may pave the way to novel strategies applied to different liver diseases in humans.
ew areas in medicine have enjoyed similar success as liver transplantation. As a result, the imbalance between organs available for transplantation and the number of patients awaiting an organ has grown dramatically over the past decade, triggering interest to maximize and optimize the use of potential organs. For example, marginal organs (i.e., organs not used previously or expected to be associated with increased risk for malfunction) and partial liver transplantation such as living-related and split-liver transplantations are used increasingly in most transplant centers.1,2 A common issue inherent to all strategies is the need to preserve the graft from the time of harvesting until implantation.3 From cooling of the graft, initiated in the 1950s, and the introduction of the University of Wisconsin (UW) cold-storage solution for cold preservation in the mid-1980s,4 many experimental studies have suggested novel protective strategies, although very few have yet reached clinical practice. Similarly, the volume of liver surgery as part of the transplant process (e.g., living-related liver transplantation) or for resection of tumors has increased dramatically over past years worldwide,5 and strategies to minimize the
F
negative effects of ischemia are now in the forefront of clinical and experimental studies related to liver resection. This article reviews established and promising protective strategies against ischemic injury of the liver.
Should We Differentiate Different Types of Ischemic Injury of the Liver? The liver can be subjected to 3 forms of ischemia, namely cold (or hypothermic), warm (or normothermic), and rewarming.3 Cold ischemia occurs almost exclusively in the transplant setting where it is applied intentionally to reduce metabolic activities of the graft while the organ awaits implantation. Warm ischemia occurs in a variety of situations including transplantation, trauma, shock, and liver surgery, when hepatic inflow occlusion (Pringle maneuver) or inflow and outflow (total vascular exclusion) are induced to minimize blood loss while dividing the liver parenchyma. Rewarming ischemia typically occurs during manipulation of the graft (e.g., ex situ split liver preparation) or during the period of implantation of the graft when the cold liver is subjected to room or body temperature while performing the vascular reconstruction. Of note, injury to the liver cells after any type of ischemia is detected mainly after reperfusion when oxygen supply and blood elements are restored. Morphologic studies in various animal models have shown major differences in the patterns of cold and warm injury. In the 1980s, it was shown that cold ischemia specifically caused injury to the sinusoidal endothelial cell (SEC),6 – 8 a finding supported by many subsequent studies.9 –12 The SEC detached, lost cytoplasmic processes, became rounded as a result of alteration of the extracellular matrix and cytoskeleton, and sloughed into the sinusoiAbbreviations used in this paper: Hsp, heat shock protein; IL, interleukin; MMP, matrix metalloprotease; OH, hydroxyl radical; SEC, sinusoidal endothelial cell; TNF-␣, tumor necrosis factor ␣; UW solution, University of Wisconsin cold-storage solution. © 2003 by the American Gastroenterological Association 0016-5085/03/$30.00 doi:10.1016/S0016-5085(03)01048-5
918
SELZNER ET AL.
dal lumen (Figure 1).7,9,13 Despite these structural changes, most SECs remain alive during the period of ischemia14,15 but rapidly die on reperfusion. Disruption of the endothelial wall leads to leukocyte16 –20 and platelet adhesion,21,22 which induces microcirculatory disturbances.23 The degree of endothelial cell detachment has
GASTROENTEROLOGY Vol. 125, No. 3
been shown to correlate with the duration of cold ischemia in numerous experimental models.9,13,19 The morphologic changes typically identified in the endothelial cells result from active processes involving the cytoskeleton and extracellular matrix.24,25 We have described biologic similarities between the injury of the SECs exposed to cold and the early stages of angiogenesis as seen in wound healing and tumor growth.26 In these cases, the endothelial cells undergo apoptosis on reperfusion.10 Adhesion of platelets to the sinusoid lining induces SEC apoptosis on reperfusion of the cold ischemic liver.22 Platelets are a rich source of transforming growth factor 27 and calpains.28 Nitric oxide production by platelets in combination with oxygen free radical synthesis on reoxygenation of the ischemic liver can lead to the formation of perioxynitrite, a highly reactive inducer of apoptosis in endothelial cells.29 Finally, models using the isolated perfused rat liver revealed that leukocytes and platelets synergistically exacerbate SEC injury by induction of apoptosis and that Kupffer cells are involved in the mechanism of injury mediated by these cells.30 In contrast to cold ischemia, warm ischemia is tolerated poorly and rapidly leads to the death of hepatocytes.31,32 This severe injury of the hepatocytes probably is preceded by massive death of endothelial cells33 (Figures 1 and 2). The role of Kupffer cells, the resident hepatic macrophages, and adherent leukocytes and platelets remains an area of active investigation in the warm ischemic liver.34 On reperfusion, Kupffer cells are activated.30,35,36 This is evidenced by structural changes,35 formation of oxygen free radicals,17,37 increased phagocytosis, and release of lysosomal enzymes10,38 and various cytokines including tumor necrosis factor ␣ (TNF-␣)38,39 (Figure 3). Further binding of these cytokines to their respective receptor or release of oxygen free radicals during early stages of reperfusion initiates the complex apoptotic machinery leading to the death of hepatocytes.38 Stress-activated protein kinase, especially c-jun Š Figure 1. Representative transmission electron micrographs of rat livers preserved in cold Euro-Collins solution for 16 hours of preservation (nonviable condition) and/or reperfused for 1 hour in the IPRL model. (A) Cold-preserved liver after 16 hours of cold ischemia. Note the typical cold preservation injury with rounding and detachment of endothelial cells and disruption of the sinusoidal wall. (B) Liver preserved for 16 hours and reperfused for 1 hour. An apoptotic endothelial cell is shown with typical shrinkage and chromatine condensation in the nucleus. Note the presence of intact mitochondria and numerous vacuoles (arrow). (C) In the same group are numerous activated Kupffer cells with phagolysosomes containing apoptotic bodies. S, sinusoid; EC, endothelial cell; K, Kupffer cell; L, lymphocyte; H, hepatocyte; R, red blood cell; A, apoptotic bodies. Reprinted with permission of Clavien et al.3 and Gao et al.10
September 2003
PROTECTION AGAINST LIVER ISCHEMIA
919
causes a T-cell–mediated type of injury in a model of warm ischemia in mice.49 Leukocyte recruitment into sinusoids during the early phase of reperfusion also is mediated through activation of the complement cascade. The complement chains activated rapidly by cellular proteins released during reperfusion up-regulate macrophage antigene 1 expression on neutrophils and their recruitment into sinusoids.50 Additionally, complement components can directly cause cell injury by assemblage and deposition on membranes.51 The impact of rewarming on the structural integrity of the liver and the mechanism of this type of injury is understood poorly. It probably reflects a combination of cold and warm injury.
How Can We Protect the Liver Against Ischemic Injury? Figure 2. Electron micrograph representing warm injury in the mouse liver. Endothelial cell swelling accompanied by hepatocyte necrosis (nH) is observed. Accumulation of polymorphonuclear leukocytes (PMN) and platelets (PLTs) are present in the sinusoids. R, red blood cell.
N terminal kinase, are activated by extracellular stimuli during hypoxia reoxygenation, leading to nuclear transcription factor activation and to the onset of hepatocyte apoptosis.40,41 After reperfusion, leukocytes rapidly adhere to the denuded sinusoids and contribute significantly to the injury.16,8-20,42 Both TNF-␣ and interleukin 1 (IL-1), released by activated Kupffer cells, can up-regulate CD11b expression on leukocytes and recruit these cells into sinusoids.43 The mechanism of injury involves the release of reactive oxygen intermediates by reduced nicotinamide adenosine dinucleotide phosphate– dependent oxidase systems expressed on the membrane surface of neutrophils. Other potential substances released by neutrophils include various proteases and hypochloric acid.44 There is a growing body of evidence suggesting that host T cells also participate in hepatic ischemic injury. For example, both cyclosporine A and Fk506, which are potent T-cell– deactivating agents, may decrease reperfusion injury after transplantation or warm ischemia.45,46 TNF-␣ and IL-1 also can recruit and activate CD4⫹ T lymphocytes in the liver during the early phase of reperfusion,47 which auto-amplify Kupffer cell activation and neutrophil recruitment into the liver. These phenomena are mediated through the release of several factors by CD4⫹ T lymphocytes, such as granulocyte colony stimulating factor and interferon ␥.47,48 The interaction between CD 154 expressed on mature activated CD4⫹ T lymphocytes and CD40 on antigen-presenting cells,
Many protective strategies have been proposed, which can be classified in different ways. For example, from a practical perspective, protective strategies can be divided into 3 different categories: (1) surgical interventions, (2) the use of pharmacologic agents, and (3) gene
Figure 3. Mechanisms of warm ischemic injury. Major pathways include TNF-␣–mediated apoptosis, dysregulation of ion distribution, and the generation of reactive oxygen intermediates (ROI). Intracellular sodium accumulation is caused by a combination of blocking sodium efflux driven by the Na⫹/K⫹ adenosine triphosphatase and activation of sodium influx by the Na⫹/H⫹ exchanger. In the cold, similar factors from the sinusoidal lumen cause injury on SECs. After reperfusion and rewarming, these cells rapidly undergo apoptosis.
920
SELZNER ET AL.
Figure 4. Continuous ischemia is used to prevent bleeding during liver resection. Three different surgical strategies are shown. Ischemic periods are drawn in black, and reperfusion is drawn in white.
therapy. Another possibility is to present the strategies based on the protective mechanisms: (1) strategies aimed at a preemptive induction of tolerance against ischemic injury, which can be covered by the concept of preconditioning; and (2) strategies aimed directly at interfering with the pathways of injury either by inhibiting deleterious molecules or enhancing protective pathways. This can be covered by the term direct protection. In this article, we use the first classification because it has more practical implications. Protective effects against cold and warm ischemic injury are discussed for each strategy. Although some protective strategies also may affect hepatic regeneration, we limit this review on ischemic and reperfusion injury. For an in-depth review on some specific aspects of hepatic injury, the reader is referred to recent reviews.52–55
Surgical Strategies Today, 2 powerful strategies are in clinical use, ischemic preconditioning and intermittent clamping (Figure 4). Other protocols that have shown protection in animal models include preconditioning by hyperthermia56 – 61 and application of a portosystemic shunt during the hepatic inflow occlusion,23 but these approaches never made the transition into clinical practice outside of case reports. Ischemic Preconditioning Ischemic preconditioning consists of a brief period of ischemia followed by a short interval of reperfusion before the actual surgical procedure, with a prolonged ischemic stress.62 During the surgery, hepatic
GASTROENTEROLOGY Vol. 125, No. 3
inflow is occluded by placing a vascular clamp or a loop around the portal triad (i.e., portal vein, hepatic artery, and bile duct), rendering the whole organ ischemic. After an ischemic interval of 10 –15 minutes, the clamp is removed and the liver is reperfused for 10 –15 minutes before the prolonged ischemic insult (Figure 4). Our current understanding of the underlying biologic principle is that cells primed by various kinds of subinjurious stress trigger defense mechanisms against subsequent lethal injury of the same or different type.54,63,64 The phenomenon initially was discovered in the myocardium by Murry et al.65 in 1986. Subsequently, beneficial effects were shown in various tissues including skeletal muscle,66 brain,67 spinal cord,68 kidney,69 retina,70 lung,71 intestine,72 and liver.73–76 Although the benefit of ischemic preconditioning in the liver already has been suggested in a clinical pilot study75 and a large randomized study,77 knowledge of the molecular mechanisms remains vague. Several mediators have been proposed to play a critical role in the protective pathways including adenosine,62 nitric oxide,74 oxidative stress, some heat shock proteins (Hsps) (e.g., Hsp 72 and heme oxygenase 1/Hsp32),78 and TNF-␣.79 In 1990, Colletti et al.39,80 provided evidence suggesting that prolonged ischemic intervals lead to a burst of various cytokines including TNF-␣. Other groups subsequently confirmed this finding.79,81,82 TNF-␣ initiates apoptosis (programmed cell death) in hepatocytes and SECs.38,82 Peralta et al.79 showed the protective effect of ischemic preconditioning in a rat model of warm ischemia through blockade of P selectin up-regulation induced by TNF-␣. Several proapoptotic proteins are activated during the reperfusion phase, including the proteases caspase-8 and caspase-3, and the release of cytochrome c from the mitochondria into the cytoplasm. This cascade finally leads to the destruction of nuclear DNA, resulting in cell death. However, a controversy emerged over the past year on whether necrotic or apoptotic cell death accounts for the severe parenchymal injury observed during reperfusion.31,83 Some investigators reported that the overwhelming part of parenchymal injury is caused by massive necrotic alterations.31 In contrast, others showed that specific inhibition of apoptosis significantly prevented parenchymal injury and improved animal survival after prolonged periods of ischemia.11,82,84-87 It is possible that both pathways are present after ischemic injury and that apoptosis and necrosis might overlap after reperfusion injury. This necroapoptosis theory was developed by Lemasters.88 It postulates that a process begins with a common death signal or toxic stress that culminates in either cell lysis (necrotic cell death) or programmed
September 2003
Figure 5. The necroapoptosis hypothesis proposed by Lemasters.88
cellular resorption (apoptosis), depending on other modifying factors such as the decline of cellular adenosine triphosphate levels or fat content (steatosis).89,90 In this perspective, it seems possible that the demarcation between apoptosis and necrosis might not be as clear-cut as was pointed out in numerous publications (Figure 5). Although the extent of apoptosis is still under debate, it is clear that ischemic preconditioning down-regulates ischemia reperfusion injury: the burst of TNF-␣ (see earlier) is inhibited and therefore activation of the apoptotic cascade is not observed.75,76 Furthermore, preconditioning preserved the morphology of the hepatic parenchyma and prevented a rapid increase of general markers of hepatocyte injury such as the serum transaminase levels. The protective effects of ischemic preconditioning could be mediated through various possible mechanisms. Peralta et al.91 identified in a model of warm ischemia a protective pathway initiated by ischemic preconditioning that involved the activation of nitric oxide synthase by adenosine. They found that blocking the specific adenosine receptor A2 prevented the beneficial effect of ischemic preconditioning (see Pharmacologic Strategies). An increase in nitric oxide production was detected immediately after hepatic preconditioning. It previously has been reported that nitric oxide, in a relatively narrow therapeutic window, can inhibit apoptosis by various mechanisms including the direct inhibition of caspases by nitrosylation of the active site.92 A second possible mechanism of protection is that ischemic preconditioning confers subinjurious stress to the liver leading to the development of natural defense mechanisms. Indeed, short intervals of ischemia as applied during ischemic preconditioning induce various types of stress including generation of oxidative stress. Our group recently has shown in a model of warm ischemia that a mild burst of oxidative stress generated during the process of ischemic preconditioning induces natural defense mechanisms against subsequent lethal
PROTECTION AGAINST LIVER ISCHEMIA
921
injury.93 Similar findings have been reported in cold ischemic liver.93,94 In addition to the extracellular mediators, studies in heart and liver indicate that the ischemic preconditioning process involves activation of intracellular messengers such as protein kinase C, adenosine monophosphate–activated protein kinase, p38 mitogen-activated protein kinase, Ik kinase, and signal transducers and activators of transcription-1.95–98 The downstream consequences of these pathways could be cytoprotective by abrogation of cell death pathways (such as activation of vacuolar adenosine triphosphatases, inhibition of intracellular Na accumulation, and cell swelling97,99), stimulating antioxidant and other cellular protective mechanisms, and by initiating entry into the cell cycle. In the steatotic liver subjected to warm ischemia, a predominance of necrosis as the primary form of cell death has been observed.100,101 Intracellular mediators of apoptosis were decreased in steatotic livers subjected to ischemia, indicating a failure to activate the apoptotic cascade. A further indication that apoptosis is not the predominant mechanism of injury was the lack of protection when caspase inhibitors were used; unlike in the control liver, treatment with these antiapoptotic agents was not effective in fatty livers.101 Thus, the question arose whether ischemic preconditioning also may prevent necrotic injury. In the murine steatotic liver, our group showed protective effects of ischemic preconditioning by reducing necrosis. These effects were associated with restoration of high adenosine triphosphate levels after reperfusion.102 Furthermore, clinical evidence suggests that patients suffering from hepatic steatosis (20%–50% steatosis) greatly benefit from ischemic preconditioning.75 One possible explanation could be the necroapoptotic theory.88 Based on this theory the lack of adenosine triphosphate and/or other changes in fatty livers might switch the type of cell death from apoptosis to necrosis. The protective effects of ischemic preconditioning on hepatic microcirculation have been studied recently.103,104 Improved microcirculation either might be the result of or the reason for the preserved parenchymal architecture.103 In addition, assessment of the hepatic microcirculation using intravital microscopy or laser Doppler technology is limited to the sinusoids in very close proximity to the capsule. These areas are supplied by passive oxygen diffusion via the peritoneum even during complete hepatic inflow occlusion and therefore are not representative for the whole organ. With intravital microscopy, we could show reduced Kupffer cell activation and improved microcirculation in a mouse model of
922
SELZNER ET AL.
warm ischemia and reperfusion (unpublished observations). Although a short cycle of ischemia and reperfusion triggers protective mechanisms, it also likewise is associated with injurious elements105 (e.g., generation of oxidative stress and nitric oxide). Such detrimental effects of ischemic preconditioning might outweigh in part the protective effects. From this point of view, nonspecific ischemic preconditioning might not be the most effective strategy. Further studies should focus on cellular mechanisms of ischemic preconditioning. In a second step, identification of pharmacologic agents should be attempted, which specifically interfere with the injurious effects of ischemic preconditioning. These agents might be used to specifically induce protection and, at the same time, circumvent the negative effects of the short cycle of ischemia and reperfusion.105 Intermittent Clamping The first attempts to minimize warm ischemic injury were undertaken by interrupting long ischemic periods with multiple short intervals of reperfusion (intermittent clamping).106 Although the protective mechanisms of this concept still remain elusive, intermittent clamping currently is used in practice by many centers. It is assumed that the protective mechanisms are similar to those described in ischemic preconditioning, mainly by reduction of apoptosis.82 In a prospective randomized study, Belghiti et al.107 showed that cycles of short intervals of ischemia (15 min) and reperfusion (5 min) provided a high degree of protection in patients undergoing major liver resection. We have compared this protocol with ischemic preconditioning82 and found that both strategies provide comparable protection for ischemic intervals of up to 75 minutes. For longer ischemic intervals, only intermittent clamping conferred significant protection. However, most resections required inflow occlusion of less than 60 minutes. We therefore concluded that ischemic preconditioning is preferable for most liver resections because each period of reperfusion in the intermittent clamping strategy may cause significant bleeding. Extracorporeal Machine Perfusion Systems Extracorporeal machine perfusion systems have been proposed as a tool to provide superior tissue preservation and viable non– heart beating donor organs. The aim of such systems is to stop the process of biodegradation.108,109 By continuously providing the graft with essential substrates (e.g., glucose, amino acids, nucleotides, oxygen) combined with permanent disposal of toxic metabolites,110 it is expected that organ viability
GASTROENTEROLOGY Vol. 125, No. 3
can be maintained better. The system is based on models of isolated liver perfusion that have been used widely to study the mechanisms of cold preservation injuries. Although this technique has been developed primarily as a tool for temporal extracorporeal liver support in patients with liver failure, it also has a potential application in organ preservation or resuscitation before transplantation.109 St Peter et al.111 and Imber and St Peter112 showed that warm oxygenated sanguineous machine perfusion recovers the liver function to a viable level after 24 hours of cold preservation whereas simple cold storage (UW solution) for 24 hours renders the liver nonviable. The perfusion techniques were improved by various modifications including oscillating pressure profiles imitating the intra-abdominal conditions113 or simultaneous dialysis of the recirculating perfusate removing water-soluble toxins and allowing regulation of the pH and electrolyte levels.114,115 Both warm110,111,114 –117 and hypothermic118 perfusion systems have been described. For example, using a warm extracorporeal perfusion system with a porcine liver, Butler et al.116 were able to maintain the liver in a viable condition for 72 hours. The introduction of extracorporeal perfusion systems into the clinical routine mainly will depend on the practicability of these still very complicated machines. The demanding and sophisticated handling of such perfusion systems may complicate the logistics and significantly increase the costs. Hyperthermic Preconditioning A number of animal studies indicate that the liver can be preconditioned by temporary exposure of the organ or the whole body to hyperthermia.57– 61,119 The heat stress response is associated with the induction of an intracellular stress protein (Hsps). Experiments covering many organs and species showed that Hsps are induced under a variety of conditions of stress,120,121 including oxidative stress (ischemia) and various pharmacologic reagents. They belong to a class of proteins called chaperones that are involved in protein folding122 during synthesis and represent cellular mechanisms of protection from protein degradation. In particular, Hsp70123,124 and heme oxygenase 1 (or Hsp32)78,125 contribute to the protective mechanism of hyperthermic preconditioning, based on the finding that overexpression of these 2 molecules increased the resistance of the liver and other organs to ischemic injury. Although it is impractical to perform whole-body hyperthermia in humans, these studies identified several protective pathways, which might be induced by more practical means.
September 2003
Pharmacologic Strategies A large number of pharmacologic agents were shown to confer protection against ischemic injury in the liver. They either blocked the injurious pathways directly or they subjected the liver to preconditioning (i.e., they induced a low level of stress to the liver cells that initiated cellular defense mechanisms against a subsequent stronger insult). A nonexhaustive list of all these agents is presented in Table 1. A number of these agents mentioned later have been identified during studies on ischemic preconditioning. Antioxidants There is growing evidence that the resident macrophages of the liver (Kupffer cells) can cause liver damage in a number of disease processes including cold35,36 and warm17,18,126 ischemic injury. Ischemia activates Kupffer cells, which are the main source of vascular reactive oxygen species during the reperfusion period.127,128 Previous studies had shown that newly recruited monocytes and leukocytes are partially responsible for the ischemic injury.18,20,129 Based on their ability to kill and digest cells, macrophages also play an important role in removing apoptotic bodies10,33 and in the synthesis of reactive oxygen species17 (e.g., superoxide and hydrogen peroxides). In hepatocytes, pro-inflammatory cytokines can induce the formation of reactive oxygen species, for example, TNF-␣, IL-1, or interferon-␥.130 Moreover, ischemic cell damage can lead to an intracellular oxidant stress during reoxygenation.131 Mitochondria are recognized as the major intracellular source of reactive oxygen species, which are generated as a product of cellular respiration.132 The most damaging form of reactive oxygen species generated in mitochondria is the hydroxyl radical (OH䡠). One of the major and most sensitive targets of OH䡠 is the mitochondrial DNA.133–135 OH䡠 attacks deoxyribose and causes the release of nucleic acids of mitochondrial DNA, resulting in strand breaks. OH䡠 also directly can attack bases, leading to modifications with a loss of DNA integrity and hence lead to impaired transcription. Although the role of reactive oxygen species in a number of liver diseases is generally accepted, the detailed mechanisms of reactive oxygen involvement are under debate. The most convincing hypothesis of reactive oxygen– induced cell injury is the destruction of cellular membranes through peroxidation of lipids.136 The parallel increase of glutathione, myeloperoxidase, and products of lipid peroxidation are strongly in favor of this degenerative process. In addition, all mitochondrial constituents, proteins, lipids, and mitochondrial DNA137 are potential
PROTECTION AGAINST LIVER ISCHEMIA
923
targets for reactive oxygen species–mediated damage. Through such damage, a gradual impairment of defenses in mitochondria will enhance the effect of further oxidative stress. In the liver, the involvement of reactive oxygen has been suggested in apoptotic cell death of hepatocytes and endothelial cells.138,139 One possible explanation is that oxidant stress can induce the mitochondrial membrane permeability transition, a central event preceding cell death.140 Another potential target could be the caspases, a family of cysteine proteases that is important for the initiation and progression of apoptosis.141 Caspases can be activated by low concentrations of hydrogen peroxide whereas higher levels inhibit enzymatic activity, presumably owing to oxidation of critical sulfhydryl groups. Thus, reactive oxygen species may induce or inhibit apoptosis depending on the severity of oxidative stress. The OH䡠 concentration-dependent activation or inhibition of caspases may provide an explanation why apoptosis and/or necrosis may both be the result of oxidative stress. Because of the central role of oxidative stress in the setting of ischemia reperfusion, a large number of studies (Table 1) attempted to identify methods to either prevent or neutralize oxidative stress. It furthermore has been shown that strategies aimed at overexpressing antioxidant proteins (e.g., superoxide dismutase142–144) may confer protection against extended ischemic injury. However, none of these strategies have found the way into routine clinical practice, with the exception of some antioxidant ingredients that were introduced into preservation solutions. Adenosine Agonists and Nitric Oxide Donors In various animal models of ischemic preconditioning in the heart, adenosine accumulates in the myocardia during ischemia and reperfusion, and confers strong protection against ischemic injuries.145,146 Investigating the protective mechanisms of ischemic preconditioning in the rat liver, Peralta et al.62,91,147,148 identified a similar protective pathway after prolonged periods of warm ischemia and reperfusion. In a series of experiments, they blocked adenosine receptors with specific antagonists or metabolized endogenous adenosine with adenosine deaminase. In both approaches, the protective effects of ischemic preconditioning were abolished.91 Peralta et al.91 further showed that the mechanisms by which adenosines confer protection involves the induction of the enzyme nitric oxide synthase in the ischemic liver. This leads to increased levels of nitric oxide, which, at moderate concentrations, prevents damage of hepatocytes and endothelial cells.149
924
SELZNER ET AL.
GASTROENTEROLOGY Vol. 125, No. 3
Table 1. Pharmacological Agents Reported to Confer Protection Against Ischemic Reperfusion Injury in the Liver Drug Oxidative stress Albumin Allopurinol Atrial natriuretic peptide Bucillamine Cyclosporin, ibuprofen combined treatment Ebselen FK506 ␥-glutamylcysteine ethyl ester Glycyrrhizin Green tea extract Melatonin Nicaravan S-nitroso-␣(1)-protease inhibitor (S-nitric oxide-␣(1)-PI) PGI2, superoxide dismutase, catalase, combination Picroliv Propyl gallate ␣-tocopherol Trimetazidine Trolox Energy depletion mitochondria Niacinamide Ozone S-15176, a potent trimetazidine derivative Tauroursodeoxy-cholate Microcirculation 2-aminoethyl-isothiourea Furosemide and bumetanide Misoprostol OP-41483 (prostacyclin) analog Sodium ozagrel OKY-046
Type of injury
Species
Reference
Antioxidant
Rat
216
Oxidative stress, scavenging
Rat
20,217
CGMP receptor–mediated oxidant stress Antioxidant
Rat Rat
218–220 221
Lipid peroxidation, oxidative stress? Lipid peroxidation, oxidative stress Free radicals, suppressor of cytokine response Oxidative stress Oxidative stress Free radical scavenger Oxidative stress Oxidative stress
Rat Rat Rat Rat Rat Rat Rat Dog
222 223 224 225 226 227 228 229
Warm ischemia
Blood flow, heme oxygenase induction
Rat
230
Warm Warm Warm Warm Warm Warm
Oxidative stress Antioxidant Antioxidant Lipid peroxidation? Scavenger of oxygen radicals Peroxyl radical scavenger
Rat Rat Rat Rat Rat Rat
231 232 233 234,235 236 237
Warm ischemia Warm ischemia
Adenosine triphosphate Adenosine triphosphate
Rat
238 239
Warm ischemia In vivo pig liver transplantation Warm ischemia
Mitochondrial permeability transition
Rat
240
Membrane stabilization Antiapoptotic?
Pig Rat
241 242
Warm ischemia
Inducible nitric oxide synthase Na⫹-K⫹-2Cl-cotransporter antagonist (loop diuretics) Microvascular system Microcirculation
Rat
243
Rat Rat Rat
244 245 182
Thromboxane A2 inhibitor Microcirculation Endothelin receptor antagonist, microcirculation Angiotensin type II antagonist Ca-channel blocker
Pig Rat
246 247,248
Dog Rat Rat
249 250 251
Rat Rat, human Rat
32 252,253 254
Cold ischemia Warm and cold ischemia Cold and warm ischemia Cold ischemia Warm Warm Warm Warm Warm Warm Warm Warm
ischemia ischemia ischemia ischemia ischemia ischemia ischemia ischemia
ischemia ischemia ischemia ischemia ischemia ischemia
Warm ischemia Warm ischemia Warm ischemia Transplantation, cold ischemia Warm ischemia
Effector system
TAK-044 TCV-116 Verapamil flunarizine Protease inhibitor/antiapoptotic Cbz-Leu-Leu-Tyr-CHN2
Warm ischemia Warm ischemia Warm ischemia
Gabexate mesilate ONO-5046 Secretory leukocyte protease inhibitor Urinary trypsin inhibitor Z-Asp-cmk (Z-Asp-2,6dichlorobenzoyl-oxymethylketone) ZVAD-fmk Inflammatory and leukocyte adhesion Cox-2 inhibitor Cyclosporine/FK506 Cyclosporine
Warm ischemia Warm ischemia
Calpain inhibitor Protease inhibitor release of reactive oxygen species Granulocyte elastase
Warm ischemia Warm ischemia
Leukocyte protease Myeloperoxidase, neutrophil proteases
Mouse Rat
255 256
Warm ischemia Warm ischemia
Caspase inhibitor Caspase inhibitor
Rat Rat
84,257 165
Warm ischemia Warm ischemia Warm ischemia Cold ischemia reperfusion with blood Warm ischemia Warm ischemia Cold ischemia Warm ischemia Warm ischemia Warm ischemia
Anti-inflammatory Inhibition of neutrophil infiltration Adenine nucleotides, mitochondrial function
Dog Rat Rat
258 259 260
P-selectin antagonist Inhibition of neutrophil infiltration Anti-inflammatory Cytokine attenuation Inflammatory preconditioning? Leukocyte adhesion H(2) receptor antagonist, neutrophil activation
Rat Rat Mouse Mouse Rat Rat Rat
261 262,263 264 48 265 266 267
Inhibits neutrophil infiltration
Rat
268
P-selectin blocker
Rat
269
Desferriexochelin 772SM FTY20 IL-6 IL-10 Lipopolysaccharide Prostaglandin F1 Ranitidine Recombinant hepatocyte growth factor Soluble P-selectin glycoprotein ligand-1
Warm ischemia
Warm ischemia Warm and cold ischemia
September 2003
This protective pathway offers several possibilities for pharmacologic interventions, including adenosine-receptor (A2) agonists and nitric oxide precursors (nitric oxide donors). Several studies in animal models indicated that synthetic adenosine receptor agonists (e.g., CGS-21680) may confer protection to the liver against cold150 and warm151 ischemic insults. Alternatively, administration of nitric oxide donors such as L-arginine,149 NONOate,91 FK409,152 and others induced protection against warm ischemic hepatic insults in rat models. Pentoxifylline Pentoxifylline is a methylxanthine theobromine derivative that has been used for many years in the treatment of peripheral vascular disease. Pentoxifylline has several additional properties, which makes it an appealing candidate drug against reperfusion injury. A generally accepted mechanism of action is the inhibition of intracellular elevation of cyclic adenosine monophosphate phosphodiesterase levels, leading to increased intracellular levels of cyclic adenosine monophosphate.153,154 One of the interesting and important observations is that pentoxifylline reduces TNF-␣ synthesis and reduces secretion of TNF-␣ in several organs.155–157 We have shown that blockage of TNF-␣ release from Kupffer cells by pentoxifylline prevents up-regulation of TNF-␣ expression in a model of warm ischemic injury in the mouse liver.38 In this model, inhibition of TNF-␣ resulted in a significant decrease of liver injury and markers of apoptosis and improved animal survival.38 Protective effects of pentoxifylline against reperfusion injury by inhibition of Kupffer cell activation also has been reported in models of cold ischemia.158 –160 Other mechanisms of action of pentoxifylline include increased red blood cell flexibility, reduction of blood viscosity,161 and decreased potential of platelet aggregation.162 These basic actions of pentoxifylline may result in therapeutic benefits owing to improved microcirculation and tissue oxygenation. However, so far no clinical studies have been presented that would show a beneficial effect of pentoxifylline on ischemic liver injury. Protease Inhibitors and Antiapoptotic Molecules Increasing evidence points to apoptosis as a critical mechanism of hepatic reperfusion injury.22,33,163 Caspases belong to the family of cysteine proteinases. Specific isoforms are involved in the initiation and execution phases of apoptosis. Among these isoforms, caspase 8 is activated during the early phase and caspase 3 is activated during the late phase of apoptosis. Sup-
PROTECTION AGAINST LIVER ISCHEMIA
925
pression of their activation, or inhibition of their activity, reduces or completely abolishes apoptosis in cell culture models. In a rat model of warm ischemia, Cursio et al. showed maximal caspase activation 3 hours after reperfusion, which preceded morphologic indicators of apoptosis.164 Pretreatment of the animals with the caspase inhibitor Z-Asp-cmk 2 minutes before ischemia efficiently protected rats from lethal liver injury that normally occurred 24 – 48 hours after surgery.84,164 However, serum transaminase levels remained relatively high despite total inhibition of caspase activities and strong suppression of DNA fragmentation. These contradictory findings suggest that caspase inhibitors may completely block apoptosis but have little effect on necrosis. Other groups have confirmed the protective effect of caspase inhibition during hepatic ischemia and reperfusion in similar or other models of cold11 and warm ischemia.11,165 In contrast, no protection was observed in the steatotic liver101 where the predominant form of cell death is necrosis. The danger of a prolonged treatment with caspase inhibitors might include the prevention of apoptosis in tissue that is not affected by the ischemic insult. The physiologic turnover of cells including the removal of defective cells or possibly even of potential cancer cells might be severely disturbed. This might increase the carcinogenicity of such therapies. However, the consequences of antiapoptotic treatment in the setting of ischemic insults may not be significant beyond the therapeutic goal because this type of treatment is envisioned as a short-term therapy. Other proteases such as calpain have been reported as mediators166 of preservation-reperfusion injury through modulation of apoptosis167 and necrosis.168 Calpains are a group of nonlysosomal, cytoplasmic, calcium-dependent cysteine proteases involved in proteolysis of several cytoskeletal and membrane proteins.169 The protective effects of calpain inhibition has been reported in cold170,171 and warm32 ischemic injury. Inhibition of calpain resulted in decreased tissue injury in both endothelial cells12,171 and hepatocytes,170 ultimately resulting in increased animal survival after liver transplantation or prolonged ischemia. Prostaglandins Prostaglandins are released mainly by activated Kupffer cells during the reperfusion phase.172 Animal studies proved that prostaglandins are effective in the treatment of ischemic liver injury owing to their ability to increase liver perfusion, inhibit platelet aggregation, and also direct cytoprotection in a model of isolated perfused cat liver.173 The protective action of prostaglan-
926
SELZNER ET AL.
din I2 and prostaglandin E1 may be related to their ability to reduce both the release of proteases and the generation of oxygen free radicals from activated leukocytes.174,175 In addition, because of the synergistic role of platelets and leukocytes and the interaction of these cells with the SECs during the reperfusion phase,30 it is conceivable that effects of prostaglandin I2 and prostaglandin E1 on leukocyte adherence may account for their favorable action.176 In clinical liver transplantation, preflush of the graft with albumin and prostaglandins before reperfusion improves early graft function.177 Greig et al.178 found that, after reperfusion and progression to primary dysfunction, liver function could be restored by treatment with a prostaglandin E1 analog. However, 2 randomized studies failed to prove a beneficial effect of a prophylactic treatment with a prostaglandin E1 analog; the incidence of primary liver failure after transplantation was not significantly different in the prostaglandin E1 analog group compared with the placebo group, although a benefit for renal function was observed.179,180 The use of pharmacologic doses of natural prostaglandins in clinical settings is limited because of drug-related side effects.181 Synthetic prostaglandin analogs (e.g., misoprostol181 or OP-41483181,182) were associated with milder side effects and a longer half-life. Several of these analogs improved animal survival and prevented parenchymal injury after prolonged periods of warm hepatic inflow occlusion.181–183 A study establishing a benefit after transplantation and a reduction of side effects of these new drugs has not been presented yet. Inhibitors of Matrix Metalloproteinase Inhibitors Matrix metalloproteinases (MMPs) belong to the family of zinc-dependent metalloproteinases that are involved in the degradation of extracellular matrix components. Arthur et al.184 showed that several cell types including Kupffer cells and probably SECs have the capacity to secrete MMPs. A comparative study reported a role for MMPs in cold preservation injury of the liver in humans and rats.24 The structural changes induced by cold preservation (i.e., rounding and detachment of SECs) most probably involve alterations in the cellular cytoskeleton and in the connections between cell and matrix.24 Changes in the extracellular MMP activities in the early phases of reperfusion are associated with morphologic changes. MMPs therefore represent an appealing target for pharmacologic interventions using MMP inhibitors. Interestingly, protection of the preserved liver by preservation solution can be explained partially by the
GASTROENTEROLOGY Vol. 125, No. 3
MMP-inhibiting activity of some of the ingredients (e.g., lactobionic acid in the UW solution).24,185 Consistently, in a rat model of prolonged warm ischemia and reperfusion, Cursio et al.186 showed that MMPs and their natural inhibitor (i.e., tissue inhibitors of MMPs) genes are induced in a specific time-dependent manner after ischemia and reperfusion, suggesting that MMPs and tissue inhibitors of MMPs could play both deleterious and beneficial roles. Pretreatment of the animals with the phosphinic MMP inhibitor RXPO3 significantly reduced markers of parenchymal injury and apoptosis. Unfortunately, clinical trials with several MMP inhibitors for the treatment of cancer or chronic inflammatory diseases have led to disappointment, including unfavorable side effects, suggesting that chronic pharmacologic application of such molecules is not advisable at this point.187,188 However, because protective strategies against ischemia reperfusion injuries will not require chronic application of MMP inhibitors, such drugs may be valuable in the treatment of acute processes such as cold or warm hepatic ischemia. Cooling of the Organ and Preservation Solutions Reduction of metabolic activities by cooling of the organ to 1°C to 4°C was among the first strategies designed to protect against ischemic injury.4 This strategy may safely preserve the liver for transplantation for up to 8 hours, whereas livers kept at room temperature tolerate only about 1 hour of ischemia before reperfusion. Cooling requires the application of a perfusion/preservation solution. Thus, efforts were directed at designing an effective solution that would extend the period for safe preservation. The breakthrough in this field came in the mid-1980s as a result of the lifelong work of Belzer and Southard.4 They empirically designed a solution based on the known and speculated negative effects of hypothermia. These negative effects include (1) cell swelling caused by inhibition of the membrane pump Na⫹/K⫹ adenosine triphosphatase regulating cell volume189; (2) intracellular acidosis caused by anaerobic metabolism and lactate accumulation190; (3) disturbances in the homeostasis of cytosolic Ca⫹⫹191; and (4) free radical generation.192,193 Belzer and Southard4 included several agents in their UW solution, each of them presumed to counteract the potential negative effects of hypothermia. Although this solution represents one of the most significant progresses in the field of liver transplantation by extending safe preservation beyond 24 hours (compared with 8 hours with Euro Collins solution),8,194 the mechanisms involved in protection remain largely unknown.
September 2003
The UW solution was shown to protect mainly against SEC injury.13,195 Therefore, it was speculated that the solution exerted its protective effect by inhibition of MMP activity owing to one of the ingredients (lactobionic acid), a potent MMP inhibitor.185 Recently, Bretschneider’s196 solution, also known as histidine/tryptophan/ketoglutarate solution, has been shown to be equally effective as the UW solution at the usual periods of cold preservation used in human transplantation.197,198 This is a surprising outcome because the compositions of these 2 solutions are very different, and, seemingly, the only property shared by these solutions is buffering capacity and MMP inhibition. However, buffering capacity alone cannot explain their effectiveness because solutions with excellent buffering capacity, such as Krebs-Henseleit solution and Euro-Collins solution, are poor preservation solutions. These results underline the lack of understanding of protective mechanisms of preservation solutions. Strategies aimed at improvement of currently used preservation solutions by adjunction of better pharmacologic agents should be a goal of future investigations but are not the main focus of this article. Gene Therapy Recent advances in genome manipulation provide new tools to reduce or suppress liver injury by gene therapy. Genome manipulation can be achieved by: (1) manipulation on the germ line (e.g., oocyte injections), (2) stem cell transformation and reintroduction into embryos, and (3) targeting specific cells or organs with vectors or viruses (e.g., adenovirus) carrying a gene of interest. The first 2 approaches may include germ-line alterations and are neither feasible nor accepted by society. The third approach, representing somatic therapy, would lend itself to the treatment of individual patients with either acquired or congenital diseases. In the past decade, efforts toward construction of cell or organspecific virus with a high infectivity rate,199 transgene expression,200 and replication deficiency have resulted in a battery of tools with which gene therapy may be envisioned.201 Modified recombinant adenovirus now allow specific targeting to the liver without affecting other organs. With respect to liver transplantation, pretreatment of donors with such vectors might help to suppress the reperfusion injuries of the liver. However, liver transplantations are in most cases an emergency procedure (cadaveric livers), leaving very little time to pretreat the donor with genetic approaches. In the situation of livingrelated donors, the elective nature of the procedure may allow a pretreatment protocol with such vectors. Again, the ethical aspect has to be taken into consideration: a
PROTECTION AGAINST LIVER ISCHEMIA
927
healthy subject (the donor) has to be treated before surgery. Some of the target genes that have been introduced successfully in animal experiments with the goal of suppressing ischemia reperfusion injury are discussed later. Antiapoptotic Strategies: Bcl-2 and Bag-1 Members of the family of proteins related to the oncogene Bcl-2 are potent regulators of the programmed cell death and either can prevent (e.g., Bcl-2 or Bcl-xL) or promote (e.g., Bid, Bad, or Bax) the apoptotic pathways.202–207 The antiapoptotic mitochondrial protein Bcl-2 is the best characterized member of the Bcl-2 family and has been studied in the setting of hepatic ischemia.85,86 In mouse models of prolonged hepatic ischemia, overexpression of this protein (either by using transgenic mice85 or by adenoviral transfection86) was associated with strong protection against reperfusion injury. Biochemical and morphologic evaluation of cell death revealed that the number of apoptotic hepatocytes was reduced significantly. In addition, increased expression of Bcl-2 prevented mortality of animals after prolonged ischemic insults. Although in the liver no involvement of this protein in the mechanisms of ischemic preconditioning was found,76 Bcl-2 appears to play a role in protecting the preconditioned heart against myocardial infarction.208 The Hsp70/Hsc70 chaperone regulator Bag-1209 also has been shown to be a powerful antiapoptotic protein that appears to interact with members of the Bcl-2 oncogene family. Bag-1 is a Bcl-2 binding protein resulting in a prolonged and stabilized antiapoptotic activity.210 In addition, Bag-1 appears to exert an indirect silencing effect on TNF receptor R1 and hence suppresses the death receptor signal. Sawitzki et al.211 used a model of cold ischemia and orthotopic liver transplantation in the rat to test the effect of adenoviral-mediated transfer of the Bag-1 gene. In contrast to an unrelated gene, -galactosidase, the adenoviral Bag-1 construct had a beneficial effect on the histopathology of the grafts, particularly on the extent of necrosis. Indicators of inflammation, TNF-␣, CD25, IL-2, and interferon ␥, were reduced on the messenger RNA level. Finally, survival of the recipient animals was increased from 50% to 100%. These results are highly encouraging to develop such a powerful technique. Antioxidant Therapy Ischemia and reperfusion injuries are characterized by a burst of oxygen radicals leading to increased
928
SELZNER ET AL.
apoptosis of hepatocytes (for a detailed description of oxidative stress–induced injury see Pharmacologic Strategies). To suppress the burst of reactive oxygen species or its effect on hepatocellular activation of nuclear factor kappa B, several groups have tried to introduce inhibitory proteins in the stress response pathway. The first protein, mitochondrial superoxide dismutase, was transfected in mice by adenoviral gene therapy.142 Using a model of partial hepatic ischemia and reperfusion injuries a beneficial effect of the treatment was shown. Subsequent studies by others showed that the introduction of genes coding for cytosolic as well as mitochondrial superoxide dismutase successfully reduced warm ischemia reperfusion injury.144 Surprisingly, intravenous application of an adenovirus coding for extracellular superoxide dismutase only was effective at a high rate of infection. Another protein, heme oxygenase 1, also known as Hsp32, appears to be induced by various conditions such as hypoxia212, 213 and hyperthermia.214 In several experimental approaches it was shown that heme oxygenase 1 exhibits a cytoprotective effect after hyperoxia or after ischemia and reperfusion. In an effort to develop strategies to expand the donor pool, Coito et al.125,215 used the gene therapy approach in Zucker rats. In contrast to control vectors, pretreatment of donor rats with adenoviral vectors carrying the heme oxygenase 1 gene were able to significantly improve several parameters after warm ischemia and orthotopic liver transplantation. Survival of the recipient was increased from 40% to 80%, whereas necrosis, edema, and macrophage infiltration were decreased markedly by the treatment. Although the mechanism of protection remains unclear, proteins with known antiapoptotic activities (e.g., Bag-1 and Bcl-2) were increased whereas inducible nitric oxide synthetase was reduced.125 The latter has been associated with ischemic injury because increases in enzymatic activity have been found in liver injuries induced by oxidative stress. Although gene therapy approaches have been used predominantly in experimental studies they appear to provide an elegant alternative strategy to induce protective mechanisms. If similar protocols as used in experimental settings would be applied to the clinical situation the donor would have to be treated with the virus 24 hours before transplantation. This temporal limitation thus restricts this potential therapy to a subgroup of donors (i.e., living-related donors). However, treatment of a healthy donor with adenoviral vectors with its potential negative side effects is currently ethically unacceptable. Efforts to reduce the time between gene therapy and transplantation might open new venues for preventative gene therapy.
GASTROENTEROLOGY Vol. 125, No. 3
Outlook on Possible Practical Applications In our view, among the various pharmacologic approaches mentioned earlier, only a few drugs are currently at the point of clinical application. Pentoxifylline could be one potential agent in the near future. This drug currently is used safely in clinical setting of various pathologies such as inflammatory bowel disease. Other drugs such as prostaglandins and MMP inhibitors have no proven beneficial effects in clinical trials despite encouraging results in animal models. Finally, caspase inhibitors present a potential danger of cancerogenesis by inhibition of apoptosis. Even though chances are slim that these inhibitors promote the development of cancerous growth in other tissues, their eventual use in the clinic should be monitored carefully. In the same setting, the gene therapy strategies are still far beyond introduction into clinical practice. Currently, only surgical strategies could be considered as protective strategies against ischemic reperfusion injury routinely used in clinical practice. All other modalities need to be better evaluated in phase I studies before routine use. In conclusion, the complex mechanisms of hepatic injuries encountered in various clinical situations have spawned a battery of different approaches to develop protective strategies. The most promising strategies to date are intermittent clamping and ischemic preconditioning, which are used during surgery. In addition, pharmacologic strategies with the goal to prevent injuries have led to the identification of dozens of promising drugs, most of which have not reached a clinical application. Finally, targeting the liver by gene therapy is a promising new tool that requires ethical discussion before reaching the clinical level.
References 1. Marcos A. Right lobe living donor liver transplantation: a review. Liver Transpl 2000;6:3–20. 2. Loinaz C, Gonzalez E. Marginal donors in liver transplantation. Hepatogastroenterology 2000;47:256 –263. 3. Clavien PA, Harvey PR, Strasberg SM. Preservation and reperfusion injuries in liver allografts. An overview and synthesis of current studies. Transplantation 1992;53:957–978. 4. Belzer F, Southard J. Principles of solid organ preservation by cold storage. Transplantation 1988;45:673. 5. Broelsch C, Testa G, Alexandrou A, Malago M. Living related liver transplantation: medical and social aspects of a controversial therapy. Gut 2002;5:143–145. 6. Otto G, Woll H, David H. Preservation damage in liver transplantation: electron microscopic findings. Transplant Proc 1984;46: 178. 7. McKeown C, Edwards C, Phillips M, Harvey P, Petrunka C, Strasberg S. Sinusoidal lining cell damage: the critical injury in cold preservation of liver allografts in the rat. Transplantation 1988;46:178 –191. 8. Momii S, Koga A. Time-related morphological changes in cold-
September 2003
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
stored rat livers: a comparison of Euro-Collins solution with UW solution. Transplantation 1990;50:745. Cladwell-Kenkel J, Thurman R, Lemasters JJ. Selective loss of non parenchymal cell viability after cold ischemic storage of rat livers. Transplantation 1988;45:834. Gao W, Bentley RC, Madden JF, Clavien PA. Apoptosis of sinusoidal endothelial cells is a critical mechanism of injury in rat liver transplantation. Hepatology 1998;27:1652–1660. Natori S, Selzner M, Valentino KL, Fritz LC, Srinivasan A, Clavien PA, Gores GJ. Apoptosis of sinusoidal endothelial cells occurs during liver preservation injury by a caspase-dependent mechanism. Transplantation 1999;68:89 –96. Sindram D, Kohli V, Madden J, Clavien P-A. Calpain inhibition prevents sinusoidal endothelial cell apoptosis in the cold ischemic rat liver. Transplantation 1999;68:136 –140. Holloway C, Harwey P, Strasberg S. Viability of sinusoidal lining cells in cold-preserved rat liver allografts. Transplantation 1990; 49:225–229. Imamura H, Brault A, Huet PM. Effects of extended cold preservation and transplantation on the rat liver microcirculation. Hepatology 1997;25:664 – 671. Miyagawa Y, Imamura H, Soeda J, Matsunaga K, Mochida S, Fujiwara K, Matsuyama Y, Kawasaki S. Fate of hepatocyte and sinusoidal lining cell function and kinetics after extended cold preservation and transplantation of the rat liver. Liver Transpl 2002;8:370 –381. Takei Y, Marzi I, Gao W, Gores G, Lemasters J, Thurman R. Leukocyte adhesion and cell death following orthotopic liver transplantation in the rat. Transplantation 1991;51:959 –965. Jaeschke H, Farhood A. Neutrophil and Kupffer cell-induced oxidant stress and ischemia-reperfusion injury in rat liver in vivo. Am J Physiol 1991;260:G355–G362. Jaeschke H, Farhood A, Smith C. Neutrophils contribute to ischemia/reperfusion injury in rat liver in vivo. FASEB J 1990; 4:3355–3359. Clavien PA, Morgan GR, Sanabria JR, Petrunka C, Levy GA, Robert P, Harvey C, Strasberg SM. Effect of cold preservation on lymphocyte adherence in the perfused rat liver. Transplantation 1991;52:412– 417. Clavien PA, Harvey PRC, Sanabria JR, Cywes R, Levy GA, Strasberg SM. Lymphocyte adherence in the reperfused rat liver allograft. Mechanisms and effects. Hepatology 1993;17:131– 142. Cywes R, Packham MA, Tietze L, Sanabria JR, Harvey P, Phillips MJ, Strasberg SM. Role of platelets in hepatic allograft preservation injury in the rat. Hepatology 1993;18:635– 647. Sindram D, Porte RJ, Hoffman MR, Bentley RC, Clavien PA. Platelets induce sinusoidal endothelial cell apoptosis upon reperfusion of the cold ischemic rat liver. Gastroenterology 2000;118:183–191. Marzi I, Knee J, Menger M, Harbauer G, Buhren V. Hepatic microcirculatory disturbances due to portal vein clamping in the orthotopic rat liver transplantation model. Transplantation 1991;52:432– 436. Upadhya A, Harvey R, Howard T, Lowell J, Shenoy S, Strasberg S. Evidence of a role for matrix metalloproteinases in cold preservation injury of the liver in humans and in the rat. Hepatology 1997;26:922–928. Upadhya G, Strasberg S. Evidence that actin disassembly is a requirement for matrix metalloproteinase secretion by sinusoidal endothelial cells during cold preservation in the rat. Hepatology 1999;30:169 –176. Gao W, Washington MK, Bentley RC, Clavien PA. Antiangiogenic agents protect liver sinusoidal lining cells from cold preservation injury in rat liver transplantation. Gastroenterology 1997; 113:1692–1700. Fukuda K, Kojiro M, Chiu JF. Induction of apoptosis by trans-
PROTECTION AGAINST LIVER ISCHEMIA
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42. 43.
44. 45.
46.
929
forming growth factor– beta 1 in the rat hepatoma cell line McA-RH7777: a possible association with tissue transglutaminase expression. Hepatology 1993;18:945–953. Schoenwaelder S, Yuan Y, Cooray P, Salem H, Jackson S. Calpain cleavage of focal adhesion proteins regulates the cytoskeletal attachment of integrin alphaIIbeta3 (platelet glycoprotein IIb/IIIa) and the cellular retraction of fibrin clots. J Biol Chem 1997;272:1694 –1702. Gow A, Thom S, Ischiropoulos H. Nitric oxide and peroxynitritemediated pulmonary cell death. Am J Physiol 1998;274:L112– L118. Sindram D, Porte RJ, Hoffmann MR, Bentley RC, Clavien PA. Synergism between platelets and leukocytes in inducing endothelial cell apoptosis in the cold ischemic rat liver: a Kupffer cell-mediated injury. FASEB J 2001;15:1230 –1232. Gujral J, Bucci T, Farhood A, Jaeschke H. Mechanism of cell death during warm hepatic ischemia-reperfusion in rats: apoptosis or necrosis? Hepatology 2001;33:397– 405. Kohli V, Madden JF, Bentley RC, Clavien PA. Calpain mediates ischemic injury of the liver through modulation of apoptosis and necrosis. Gastroenterology 1999;116:168 –178. Kohli V, Selzner M, Madden JF, Bentley RC, Clavien PA. Endothelial cell and hepatocyte deaths occur by apoptosis after ischemia-reperfusion injury in the rat liver. Transplantation 1999;67:1099 –1105. Yadav SS, Howell DN, Steeber DA, Harland RC, Tedder TF, Clavien PA. P-Selectin mediates reperfusion injury through neutrophil and platelet sequestration in the warm ischemic mouse liver. Hepatology 1999;29:1494 –1502. Thurman R, Cowper K, Marzi I, Currin R, Lemasters J. Functional activation of Kupffer cells by cold ischemic storage of rat liver in Euro-Collins solution. Hepatology 1988;8:1824 –1827. Caldwell-Kenkel J, Currin R, Tanaka Y, Thurman R, Lemasters J. Kupffer cell activation and endothelial cell damage after storage of rat livers: effects of reperfusion. Hepatology 1991;13:83–95. Jaeschke H. Kupffer cell-induced oxidant stress during hepatic ischemia-reperfusion: does the controversy continue? Hepatology 1999;30:1527–1528. Rudiger HA, Clavien PA. Tumor necrosis factor-alpha, but not Fas, mediates hepatocellular apoptosis in the murine ischemic liver. Gastroenterology 2002;122:202–210. Colletti LM, Remick DG, Burtch GD, Kunkel SL, Strieter RM, Campbell DA. Role of tumor necrosis factor-alpha in the pathophysiologic alterations after hepatic ischemia/reperfusion injury in the rat. J Clin Invest 1990;85:1936 –1943. Onishi I, Shimizu K, Tani T, Hashimoto T, Miwa K. JNK activation and apoptosis during ischemia-reperfusion. Transplant Proc 1999;31:1077–1079. Crenesse D, Gugenheim J, Hornoy J, Tornieri K, Laurens M, Cambien B, Lenegrate G, Curiso R, De Souza G, Auberger P, Heuteux C, Rossi B, Schmid-Alliana A. Protein kinase activation by warm and cold hypoxia-reoxygenation in primary-cultured rat hepatocytes—JNK/SPAK involvement in apoptosis. Hepatology 2000;32:1029 –1036. Jaeschke H, Smith C. Mechanisms of neutrophil-induced parenchymal cell injury. J Leukoc Biol 1997;61:647– 653. Bajt M, Farhood A, Jaeschke H. Effects of CXC chemokines on neutrophil activation and sequestration in the hepatic vasculature. Am J Physiol 2001;281:G1188 –G1195. Weiss SJ. Tissue destruction by neutrophils. N Engl J Med 1989;320:365–376. Suzuki S, Toledo-Pereyra L, Rodriguez F, Cejalvo D. Neutrophil infiltration as an important factor in liver ischemia reperfusion injury: modulating effects of Fk506 and cyclosporine. Transplantation 1993;55:1265. Kawano K, Kim Y, Ono M, Goto S, Kai T, Kobayashi M. Evidence
930
47.
48.
49.
50.
51.
52.
53. 54.
55.
56.
57.
58.
59.
60.
61.
62.
63.
SELZNER ET AL.
that both cyclosporine and azathioprine prevent warm ischemia reperfusion injury to the rat liver. Transpl Int 1993;6:330. Zwacka R, Zhang Y, Halldorson J, Sclossberg H, Dudus L, Engelhardt JF. CD4 (⫹) T-lymphocytes mediate ischemia reperfusion induced inflammatory response in mouse liver. J Clin Invest 1997;100:279 –288. Le Moine O, Louis H, Demols A, Desalle F, Demoor F, Quertinmont E, Goldman M, Deviere J. Cold liver ischemia-reperfusion injury critically depends on liver T cells and is improved by donor pretreatment with interleukin-10 in mice. Hepatology 2000;31: 1266 –1274. Shen X, Ke B, Zhai Y, Amersi F, Gao F, Anselmo D, Busuttil R, Kupiec-Weglinski J. CD154-Cd40 T cell costimulation pathway is required in the mechanism of hepatic ischemia/reperfusion injury, and its blockade facilitates and depends on heme oxygenase-1 mediated cytokines. Transplantation 2002;15:315– 319. Jaeschke H, Farhood A. Complement activates Kupffer cells and neutrophils during reperfusion after hepatic ischemia. Am J Physiol 1993;264:G801–G809. Chavez-Cartaya R, De Sola G, Wright L, Jamieson N, White D. Regulation of the complement cascade by soluble complement receptor type 1. Protective effect on experimental liver ischemia and reperfusion. Transplantation 1995;59:1047–1052. Lentsch A, Kato A, Yoshidome H, McMasters K, Edwards M. Inflammatory mechanisms and therapeutic strategies for warm hepatic ischemia/reperfusion injury. Hepatology 2000;32:169 – 173. Bilzer M, Gerbes A. Preservation injury of the liver: mechanisms and novel therapeutic strategies. J Hepatol 2000;32:508 –515. Jaeschke H. Molecular mechanisms of hepatic ischemia-reperfusion injury and preconditioning. Am J Physiol 2003;284:G15– G26. Vajdova´ K, Graf R, Clavien PA. ATP-supplies in the cold-preserved liver: a long-neglected factor of organ viability. Hepatology 2002;36:1543–1553. Marber M, Walker J, Latchman D, Yellon D. Myocardial protection after whole body heat stress in the rabbit is dependent on metabolic substrate and is related to the amount of inducible 70-kD heat stress protein. J Clin Invest 1994;93:1087–1094. Wagner A, Weber H, Jonas L, Nizze H, Strowski M, Fiedler F, Printz H. Hyperthermia induces heat shock protein expression and protection against cerulein-induced pancreatitis in rats. Gastroenterology 1996;111:1333–1342. Ishikawa Y, Yamamoto Y, Kume M, Yamagami K, Yamamoto H, Kimoto S, Sakai Y, Yamamoto M, Yamaoka Y. Heat shock preconditioning on mitochondria during warm ischemia in rat livers. J Surg Res 1999;87:178 –184. Terajima H, Enders G, Thiaener A, Hammer C, Kondo T, Thiery J, Yamamoto Y, Yamaoka Y, Messmer K. Impact of hyperthermic preconditioning on postischemic hepatic microcirculatory disturbances in an isolated perfusion model of the rat liver. Hepatology 2000;31:407– 415. Yonezawa K, Yamamoto Y, Yamamoto H, Ishikawa Y, Uchinami H, Taura K, Nakajima A, Yamaoka Y. Suppression of tumor necrosis factor-alpha production and neutrophil infiltration during ischemia-reperfusion injury of the liver after heat shock preconditioning. J Hepatol 2001;35:619 – 627. Matsumoto K, Honda K, Kobayashi N. Protective effect of heat preconditioning of rat liver graft resulting in improved transplant survival. Transplantation 2001;71:862– 868. Peralta C, Hotter G, Closa D, Gelpi E, Bulbena O, RoselloCatafau J. Protective effect of preconditioning on the injury associated to hepatic ischemia-reperfusion in the rat: role of nitric oxide and adenosine. Hepatology 1997;25:934 –937. Cavalieri B, Perrelli MG, Aragno M, Mastrocola R, Corvetti G, Durazzo M, Poli G, Cutrin JC. Ischemic preconditioning attenu-
GASTROENTEROLOGY Vol. 125, No. 3
64.
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
76.
77.
78.
79.
80.
81.
ates the oxidant-dependent mechanisms of reperfusion cell damage and death in rat liver. Liver Transpl 2002;8:990 –999. Schauer RJ, Gerbes AL, Vonier D, Op Den Winkel M, Fraunberger P, Bilzer M. Induction of cellular resistance against Kupffer cell-derived oxidant stress: a novel concept of hepatoprotection by ischemic preconditioning. Hepatology 2003;37:286 –295. Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation 1986;74:1124 –1136. Pang CY, Yang RZ, Zhong A, Xu N, Boyd B, Forrest CR. Acute ischemic preconditioning protects against skeletal muscle infarction in the pig. Cardiovasc Res 1995;29:782–788. Glazier SS, O’Rourke DM, Graham DI, Welsh FA. Induction of ischemic tolerance following brief focal ischemia in rat brain. J Cereb Blood Flow Metab 1994;14:545–553. Sakurai M, Hayashi T, Abe K, Aoki M, Sadahiro M, Tabayashi K. Enhancement of heat shock protein expression after transient ischemia in the preconditioned spinal cord of rabbits. J Vasc Surg 1998;27:720 –725. Turman MA, Bates CM. Susceptibility of human proximal tubular cells to hypoxia: effect of hypoxic preconditioning and comparison to glomerular cells. Ren Fail 1997;19:47– 60. Roth S, Li B, Rosenbaum PS, Gupta H, Goldstein IM, Maxwell KM, Gidday JM. Preconditioning provides complete protection against retinal ischemic injury in rats. Invest Ophthalmol Vis Sci 1998;39:777–785. Du ZY, Hicks M, Winlaw D, Spratt P, Macdonald P. Ischemic preconditioning enhances donor lung preservation in the rat. J Heart Lung Transplant 1996;15:1258 –1267. Hotter G, Closa D, Prados M, Fernandez-Cruz L, Prats N, Gelpi E, Rosello-Catafau J. Intestinal preconditioning is mediated by a transient increase in nitric oxide. Biochem Biophys Res Commun 1996;222:27–32. Hardy KJ, McClure DN, Subwongcharoen S. Ischaemic preconditioning of the liver: a preliminary study. Aust N Z J Surg 1996;66:707–710. Peralta C, Closa D, Hotter G, Gelpi E, Prats N, Rosello-Catafau J. Liver ischemic preconditioning is mediated by the inhibitory action of nitric oxide on endothelin. Biochem Biophys Res Commun 1996;229:264 –270. Clavien PA, Yadav S, Sindram D, Bentley RC. Protective effects of ischemic preconditioning for liver resection performed under inflow occlusion in humans. Ann Surg 2000;232:155–162. Yadav SS, Sindram D, Perry DK, Clavien PA. Ischemic preconditioning protects the mouse liver by inhibition of apoptosis through a caspase-dependent pathway. Hepatology 1999;30: 1223–1231. Ru¨ diger H, Selzner M, Dindo D, Selzner N, Kadry Z, Clavien PA. Prospective randomized study in 100 consecutive patients undergoing major liver resection with vs without ischemic preconditioning (abstr). Hepatology 2002;36:A28. Redaelli CA, Tian YH, Schaffner T, Ledermann M, Baer HU, Dufour JF. Extended preservation of rat liver graft by induction of heme oxygenase-1. Hepatology 2002;35:1082–1092. Peralta C, Fernandez L, Panes J, Prats N, Sans M, Pique J, Gelpi E, Rosello-Catafau J. Preconditioning protects against systemic disorders associated with hepatic ischemia-reperfusion through blockade of tumor necrosis factor-induced P-selectin up-regulation in the rat. Hepatology 2001;33:100 –113. Colletti LM, Burtch GD, Remick DG, Kunkel SL, Strieter RM, Guice KS, Oldham KT, Campbell DA. The production of tumor necrosis factor alpha and the development of a pulmonary capillary injury following hepatic ischemia/reperfusion. Transplantation 1990;49:268 –272. Wanner G, Muller P, Ertel W, Bauer M, Menger M, Messmer K. Differential effect of anti-TNF-alpha antibody on proinflammatory
September 2003
82.
83.
84.
85.
86.
87.
88.
89.
90.
91.
92.
93.
94.
95.
96.
97.
98.
cytokine release by Kupffer cells following liver ischemia and reperfusion. Shock 1999;11:391–395. Rudiger HA, Kang KJ, Sindram D, Riehle HM, Clavien PA. Comparison of ischemic preconditioning, intermittent and continuous inflow occlusion in the murine liver. Ann Surg 2002;235: 400 – 407. Clavien PA, Rudiger HA, Selzner M. Mechanisms of hepatocyte death after ischemia: apoptosis versus necrosis. Hepatology 2001;33:1555–1556. Cursio R, Guggenheim J, Ricci JE, Crenesse D, Rostagno P, Maulon L, Saint-Paul MC, Ferrua B, Auberger P. A caspase inhibitor fully protects rats against lethal normothermic liver ischemia by inhibition of liver apoptosis. FASEB J 1999;13: 253–261. Selzner M, Rudiger HA, Selzner N, Thomas DW, Sindram D, Clavien PA. Bcl-2 overexpression in transgenic mice protects against ischemia & reperfusion injury of the liver. J Hepatol 2002;36:218 –225. Bilbao G, Contreras J, Eckhoff D, Mikheeva G, Krasnykh V, Douglas J, Thomas F, Thomas J, Curiel D. Reduction of ischemia-reperfusion injury of the liver by in vivo adenovirus-mediated gene transfer of the antiapoptotic Bcl-2 gene. Ann Surg 1999;230:185–192. Bilbao G, Contreras J, Gomez-Navarro J, Eckhoff D, Mikheeva G, Krasnykh V, Hynes T, Thomas F, Thomas J, Curiel D. Genetic modification of liver grafts with an adenoviral vector encoding the Bcl-2 gene improves organ preservation. Transplantation 1999;67:775–783. Lemasters JJ. Necrapoptosis and the mitochondrial permeability transition: shared pathways to necrosis and apoptosis. Am J Physiol 1999;276:G1–G6. Cursio R, Gugenheim J, Panaia Ferrari P, Lasfar A, Tovey M, Chastanet S, Saint Paul MC, Ferre C, Mouiel J. Improvement of normothermic rat liver ischemia/reperfusion by muramyl dipeptide. J Surg Res 1998;80:339 –344. Selzner M, Rudiger HA, Sindram D, Madden J, Clavien PA. Mechanisms of ischemic injury are different in the steatotic and normal rat liver. Hepatology 2000;32:1280 –1288. Peralta C, Hotter G, Closa D, Prats N, Xaus C, Gelpi E, RoselloCatafau J. The protective role of adenosine in inducing nitric oxide synthesis in rat liver ischemia preconditioning is mediated by activation of adenosine A2 receptors. Hepatology 1999;29: 126 –132. Mannick JB, Hausladen A, Liu L, Hess DT, Zeng M, Miao QX, Kane LS, Gow AJ, Stamler JS. Fas-induced caspase denitrosylation. Science 1999;284:651– 654. Ru¨ diger HA, Selzner N, Selzner M, Clavien PA. Sublethal oxidative stress protects against ischemic injury in the mouse liver: a new mechanism of ischemic preconditioning (abstr). Hepatology 2001;34:421A. Sindram D, Rudiger HA, Upadhya AG, Strasberg SM, Clavien PA. Ischemic preconditioning protects against cold ischemic injury through an oxidative stress dependent mechanism. J Hepatol 2002;36:78 – 84. Riccardi R, Meyers W, Schaffer B, Kim R, Shah S, Wheeler S, Donohue S, Sheth K, Callery M, Chari R. Protein kinase C inhibition abrogates hepatic ischemic preconditioning responses. J Surg Res 2001;97:144 –149. Carini R, De Cesaris M, Splendore R, Bagnati M, Albano E. Ischemic preconditioning reduces Na(⫹) accumulation and cell killing in isolated rat hepatocytes exposed to hypoxia. Hepatology 2000;31:166 –172. Carini R, De Cesaris M, Splendore R, Vay D, Domenicotti C, Nitti M, Paola D, Pronzato M, Albano E. Signal pathway involved in the development of hypoxic preconditioning in rat hepatocytes. Hepatology 2001;33:131–139. Toeh N, Dela Pena A, Farrell G. Hepatic ischemic precondition-
PROTECTION AGAINST LIVER ISCHEMIA
99.
100.
101.
102.
103.
104.
105. 106.
107.
108.
109.
110.
111.
112.
113.
114.
115. 116.
117.
931
ing in mice is associated with activation of NF-kappaB, p38 kinase, and cell cycle entry. Hepatology 2002;36:94 –102. Carini R, Autelli R, Bellomo G, Albano E. Sodium-mediated swelling is associated with irreversible damage of isolated hepatocytes exposed to hypoxia or mitochondrial toxins. Biochem Biophys Res Commun 1994;202:360 –366. Koneru B, Reddy M, Torre A, Patel D, Ippolito T, Ferrante R. Studies of hepatic warm ischemia in the obese Zucker rat. Transplantation 1995;59:942–946. Selzner M, Rudiger HA, Sindram D, Madden J, Clavien PA. Mechanisms of ischemic injury are different in the steatotic and normal rat liver. Hepatology 2000;32:1280 –1288. Selzner N, Selzner M, Jochum W, Clavien PA. Ischemic preconditioning protects the steatotic mouse liver against reperfusion injury. An ATP dependent mechanism. J Hepatol 2003;39:55– 61. Zapletal C, Maksan S, Lehmann T, Guenther L, Fallsehr C, Mehrabi A, Weiss G, Golling M, Gebhard M, Herfarth C, Klar E. Ischemic preconditioning improves liver microcirculation after ischemia/reperfusion. Transplant Proc 1999;31:3260 –3262. Sawaya DJ, Brown M, Minardi A, Bilton B, Burney D, Granger D, McDonald J, Zibari G. The role of ischemic preconditioning in the recruitment of rolling and adherent leukocytes in hepatic venules after ischemia/reperfusion. J Surg Res 1999;85:163– 170. Rudiger HA, Clavien PA. A cytotoxic drug against reperfusion injury? Hepatology 2000;31:533–535. Makuuchi M, Mori T, Gunven P, Yamazaki S, Hasegawa H. Safety of hemihepatic vascular occlusion during resection of the liver. Surg Gynecol Obstet 1987;164:155–158. Belghiti J, Noun R, Malafosse R, Jagot P, Sauvanet A, Pierangeli F, Marty J, Farges O. Continuous versus intermittent portal triad clamping for liver resection: a controlled study. Ann Surg 1999; 229:369 –375. Adham M, Peyrol S, Chevallier M, Ducerf C, Vernet M, Barakat C, De La Roche E, Taibi A, Bizollon T, Rigal D, Pouyet M, Baulieux J. The isolated perfused porcine liver: assessment of viability during and after six hours of perfusion. Transpl Int 1997;10:299 –311. Butler AJ, Rees MA, Wight DGD, Casey ND, Alexander G, White DJG, Friend PJ. Successful extracorporeal porcine liver perfusion for 72 hr. Transplantation 2002;73:1212–1218. Schon M, Kollmar O, Wolf S, Schrem H, Matthes M, Akkoc N, Schnoy N, Neuhaus P. Liver transplantation after organ preservation with normothermic extracorporeal perfusion. Ann Surg 2001;233:114 –123. St Peter S, Imber C, Lopez I, Hughes D, Friend P. Extended preservation of non-heart-beating donor livers with normothermic machine perfusion. Br J Surg 2002;89:609 – 616. Imber C, St Peter S, Lopez de Cenarruzabeitia I, Pidott D, James T, Taylor R, McGuire J, Hughes D, Butler A, Rees M, Friend P. Advantages of normothermic perfusion over cold storage in liver preservation. Transplantation 2002;73:701–709. Neuhaus R, Blumhardt G. Extracorporeal liver perfusion: applications of an improved model for experimental studies of the liver. Int J Artif Organs 1993;16:729 –739. Schon M, Puhl G, Frank J, Neuhaus P. Hemodialysis improves results of pig liver perfusion after warm ischemic injury. Tranplant Proc 1993;25:3239 –3243. Schon M, Puhl G, Gerlach J. Hepatocyte isolation from pig livers after warm ischaemic injury. Transplant Int 1994;7:159 –162. Butler A, Rees M, Wight D, Casey N, Alexander G, White D, Friend P. Successful extracorporeal porcine liver perfusion for 72 hr. Transplantation 2002;73:1212–1218. Rees M, Butler A, Davies H, Bolton E, Wight D, Skepper J, White D, Friend P. Porcine livers perfused with human blood mount a
932
118.
119.
120. 121. 122. 123.
124.
125.
126.
127.
128. 129.
130.
131.
132.
133.
134.
135.
136.
137.
SELZNER ET AL.
graft-versus-“host” reaction. Transplantation 2002;73:1460 – 1467. Lee C, Zhang J, deSilva H, Coger R, Clemens M. Heterogeneous flow patterns during hypothermic machine perfusion preservation of livers. Transplantation 2000;70:1797–1811. Marber MS, Latchman DS, Walker JM, Yellon DM. Cardiac stress protein elevation 24 hours after brief ischemia or heat stress is associated with resistance to myocardial infarction. Circulation 1993;88:1264 –1272. Lindquist S, Craig EA. The heat-shock proteins. Annu Rev Genet 1988;22:631– 677. Schlesinger M. Heat shock proteins. J Biol Chem 1990;265: 12111. Gething M, Sambrook J. Protein folding in the cell. Nature 1992;355:33. Saad S, Kanai M, Awane M. Protective effect of heat shock pretreatment with heat shock protein induction before hepatic warm ischemia injury caused by Pringel’s maneuver. Surgery 1995;118:510. Kume M, Yamamoto Y, Saad S, Gomi T, Kimoto S, Shimabukoro T, Yagi Y, Nakagami M, Takada Y, Yamaoka Y. Ischemic preconditioning of the liver in rats. Implication of heat shock protein induction to increase tolerance of ischemia reperfusion injury. J Lab Clin Med 1996;128:251–258. Coito AJ, Buelow R, Shen XD, Amersi F, Moore C, Volk HD, Busuttil RW, Kupiec-Weglinski JW. Heme-oxygenase-1 gene transfer inhibits inducible nitric oxide synthase expression and protects genetically fat Zucker rat livers from ischemia reperfusion injury. Transplantation 2002;74:96 –102. Mochida S, Arai M, Ohnon A, Masaki N, Ogata L, Fujiwara K. Oxidative stress in hepatocytes and stimulatory state of Kupffer cells after reperfusion differ between warm and cold ischemia. Liver 1994;14:234 –240. Rymsa B, Wang J, de Groot H. O2 release by activated Kupffer cells upon hypoxia-reoxygenation. Am J Physiol 1991;206: G602–G607. Jaeschke H. Mechanism of reperfusion injury after warm ischemia of the liver. J Hepatol 1998;21:402– 408. Lasnier E, Blanc MC, Housset C, Rey C, Roch Arveiller M, Vaubourdolle M. Cytotoxic response of sinusoidal endothelial cells to polymorphonuclear leukocytes and its potential implication in hypoxia-reoxygenation injury. Liver 2002;22:495–500. Adamson G, Billings R. Tumor necrosis factor induced oxidative stress in isolated mouse hepatocytes. Arch Biochem Biophys 1992;294:223–229. Jaeschke H, Mitchell J. Mitochondria and xanthine oxidase both generate reactive oxygen species after hypoxic damage in isolated perfused rat liver. Biochem Biophys Res Commun 1989; 160:140 –147. Boveris A, Chance B. The mitochondrial generation of hydrogen peroxide. General properties and effect of hyperbaric oxygen. Biochem J 1973;134:707–716. Richter C, Park JW, Ames BN. Normal oxidative damage to mitochondrial and nuclear DNA is extensive. Proc Natl Acad Sci U S A 1988;85:6465– 6467. Adachi S, Zeisig M, Mo¨ ller L. Improvements in the analytical method for 8-hydroxydeoxyguanosine in nuclear DNA. Carcinogenesis 1995;16:253–258. Richter C, Gogvadze V, Laffranchi R, Schlapbach R, Schweizer M, Suter M, Walter P, Yaffee M. Oxidants in mitochondria: from physiology to diseases. Biochim Biophys Acta 1995;1271:67– 74. Omar R, Nomikos I, Piccorelli G, Savino J, Agarwal N. Prevention of postischaemic lipid peroxidation and liver cell injury by iron chelation. Gut 1989;30:510 –514. Freeman BA, Crapo JD. Biology of disease: free radicals and tissue injury. Lab Invest 1982;47:412– 426.
GASTROENTEROLOGY Vol. 125, No. 3
138. Rauen U, Polzar B, Stephan H, Mannherz G, De Groot H. Coldinduced apoptosis in cultured hepatocytes and liver endothelial cells: mediation by active oxygen species. FASEB J 1999;13: 155–168. 139. Czaja MJ. Induction and regulation of hepatocyte apoptosis by oxidative stress. Antioxid Redox Signal 2002;4:759 –767. 140. Lemasters J, Nieminen A, Qian T, Trost L, Elmore S, Nishimura Y, Crowe R, Cascio W, Bradham C, Brenner D, Herman B. The mitochondrial permeability transition in cell death: a common mechanism in necrosis, apoptosis and autophagy. Biochim Biophys Acta 1998;1366:177–196. 141. Cohen GM. Caspases: the executioners of apoptosis. Biochem J 1997;326:1–16. 142. Zwacka RM, Zhou W, Zhang Y, Darby CJ, Dudus L, Halldorson J, Engelhardt JF. Redox gene therapy for ischemia/reperfusion injury of the liver reduces AP1 and NF-kappaB activation. Nat Med 1998;4:698 –704. 143. Yabe Y, Kobayashi N, Nishihashi T, Takahashi R, Nishikawa M, Takukura Y, Hashida M. Prevention of neutrophil-mediated hepatic ischemia/reperfusion injury by superoxide dismutase and catalase derivates. J Pharmacol Exp Ther 2001;298:894 – 899. 144. Wheeler M, Katuna M, Smutney O, Froh M, Dikalova A, Mason R, Samulski R, Thurman R. Comparison of the effect of adenoviral delivery of three superoxide dismutase genes against hepatic ischemia-reperfusion injury. Hum Gene Ther 2001;12: 2167–2177. 145. Olsson R. Changes in content of purine nucleoside in canine myocardium during coronary occlusion. Circ Res 1970;26:301– 306. 146. Parrat J. Protection of the heart by ischemic preconditioning: mechanisms and possibilities for pharmacological exploitation. TIPS 1994;15:19 –25. 147. Peralta C, Hotter G, Closa D, Pi F, Badosa F, Gelpi E, RoselloCatafau J. Nitric oxide enhances endothelin production in pancreas transplantation. Pancreas 1997;14:369 –372. 148. Peralta C, Closa D, Xaus C, Gelpi E, Rosello-Catafau J, Hotter G. Hepatic preconditioning in rats is defined by a balance of adenosine and xanthine. Hepatology 1998;28:768 –773. 149. Cottart C, Do L, Blanc M, Vaubourdolle M, Descamps G, Durand D, Galen F, Clot J. Hepatoprotective effect of endogenous nitric oxide during ischemia-reperfusion in the rat. Hepatology 1999; 29:809 – 813. 150. Arai M, Thurman R, Lemasters J. Contribution of adenosine A(2) receptors and cyclic adenosine monophosphate to protective ischemic preconditioning of sinusoidal endothelial cells against storage/reperfusion injury in rat livers. Hepatology 2000;32: 297–302. 151. Nakayama H, Yamamoto Y, Kume M, Yamagami K, Yamamoto H, Kimoto S, Ishikawa Y, Ozaki N, Shimahara Y, Yamaoka Y. Pharmacologic stimulation of adenosine A2 receptor supplants ischemic preconditioning in providing ischemic tolerance in rat livers. Surgery 1999;126:945–954. 152. Dhar DK, Yamanoi A, Ohmori H, Nakashima Y, Yamamoto A, Osama NE, Kubota H, Kohno H, Nagasue N. Modulation of endothelin and nitric oxide: a rational approach to improve canine hepatic microcirculation. Hepatology 1998;28: 782–788. 153. Semmler J, Gebert U, Eisenhut T, Moeller J, Schonharting MM, Allera A, Endres S. Xantine derivates: comparison between suppression of tumor necrosis factor-alpha production and inhibition of cAMP phosphodiesterase activity. Immunology 1993; 78:520 –525. 154. Sinha B, Semmler J, Eisenhut T, Eigler A, Endres S. Enhanced tumor necrosis factor suppression and cyclic adenosine monophosphate accumulation by combination of phosphodiesterase inhibitors and prostanoids. Eur J Immunol 1995;25:147–153. 155. Strieter R, Remick DG, Ward PA, Spengler RN, Lynch JP, Larick
September 2003
156.
157.
158.
159.
160.
161.
162.
163.
164.
165.
166.
167. 168.
169.
170.
171.
172. 173.
174. 175.
J, Kunkel SL. Cellular and molecular regulation of tumor necrosis factor alpha production by pentoxifylline. Biochem Biophys Res Commun 1988;155:1230 –1236. Doherty GM, Jensen JC, Alexander NR, Buresh CM, Nortan JA. Pentoxifylline suppression of tumor necrosis factor gene transcription. Surgery 1991;110:192–198. Zabel P, Schade FU, Schlaak M. Inhibition of endogenous TNF formation by pentoxifyilline. Immunobiology 1993;187: 447– 463. Lemasters JJ, Peng XX, Bachmann S, Currin RT, Gao W, Thurman RG. Dual role of Kupffer cell activation and endothelial cell damage in reperfusion injury to livers stored for transplantation surgery. J Gastroenterol Hepatol 1995;10(suppl 1):S84 –S87. Nishizawa H, Egawa H, Inomata Y, Uemoto S, Asonuma K, Kiuchi T, Yamaoka Y, Tanaka K. Efficiency of pentoxifylline in donor pretreatment in rat liver transplantation. J Surg Res 1997;72:170 –176. Vajdova K, Smrekova R, Kukan M, Jakubovsky J, van Rooijen N, Horecky J, Lutterova M, Wsolova L. Endotoxin-induced aggravation of preservation-reperfusion injury of rat liver and its modulation. J Hepatol 2000;32:112–120. Ward A, Clissold S. Pentoxifylline: a review of its pharmocodynamic and pharmacokinetic properties, and its therapeutic efficacy. Drugs 1987;34:50 –97. Gastpar H, Ambrus JL, Ambrus CM, Spavento P, Weber FJ, Thurber L. Study of platelet aggregation in vivo. Effects of pentoxifylline. J Med 1977;8:191–197. Borghi-Scoazec G, Scoazec JY, Durand F, Bernuau J, Belghiti J, Feldmann G, Henin D, Degott C. Apoptosis after ischemiareperfusion in human liver allografts. Liver Transpl Surg 1997; 3:407– 415. Cursio R, Gugenheim J, Ricci J, Crenesse D, Rostagno P, Maulon L, Saint-Paul M, Ferrua B, Mouiel J, Auberger P. Caspase inhibition protects from liver injury following ischemia and reperfusion in rats. Transpl Int 2000;13:S568 –S572. Kobayashi A, Imamura H, Isobe M, Matsuyama Y, Soeda J, Matsunaga K, Kawasaki S. Mac-1 (CD11b/CD18) and intercellular adhesion molecule-1 in ischemia-reperfusion injury of rat liver. Am J Physiol 2001;281:G577–G585. Wang M, Sakon M, Umeshita K, Okuyama M, Shiozaki K, Nagano H, Dohno K, Nakamori S, Monden M. Prednisolone suppresses ischemia-reperfusion injury of the rat liver by reducing cytokine production and calpain mu activation. J Hepatol 2001; 34:278 –283. Squier M, Miller A, Malkinson A, Cohen J. Calpain activation in apoptosis. J Cell Physiol 1994;159:229 –237. Arora A, Groen P, Emori Y, Gores G. A cascade of degradative hydrolase activity contributes to hepatocyte necrosis during anoxia. Am J Physiol 1996;33:G238 –G245. Croall D, DeMartino G. Calcium-activated neutral protease (calpain) system: structure, function, and regulation. Physiol Rev 1991;71:813. Kohli V, Gao W, Camargo CA Jr, Clavien PA. Calpain is a mediator of preservation-reperfusion injury in rat liver transplantation. Proc Natl Acad Sci U S A 1997;94:9354 –9359. Upadhya G, Topp SA, Hotchkiss RS, Anagli J, Strasberg SM. Effect of cold preservation on intracellular calcium and calpain activity in rat sinusoidal endothelial cells. Hepatology 2003;37: 313–323. Decker K. Biologically active products of stimulated liver macrophages (Kupffer cells). Eur J Biochem 1990;192:245–261. Araki H, Lefer A. Cytoprotective actions of prostacyclin during hypoxia in the isolated perfused cat liver. Am J Physiol 1980; 238:176. Quiroga J, Prieto J. Liver cytoprotection by prostaglandins. Pharmacol Ther 1993;58:67. Fantone J, Kinnes D. Prostaglandin E1 and prostaglandin I2
PROTECTION AGAINST LIVER ISCHEMIA
176.
177.
178.
179.
180.
181.
182.
183.
184.
185.
186.
187.
188. 189.
190. 191.
192.
193.
194.
933
modulation of superoxide production by human neutrophils. Biochem Biophys Res Commun 1983;113:506. Granger D, Kvietys P, Perry M. Leukocyte-endothelial cell adhesion induced by ischemia and reperfusion. Can J Physiol Pharmacol 1993;71:67. Gaber AO, Thistlethwaite JJ, Busse HS. Improved results of preservation of hepatic grafts preflushed with albumin and prostaglandins. Transplant Proc 1988;20:992. Greig P, Woolf G, Abecassis M. Prostaglandin E1 for primary nonfunction following liver transplantation. Transplant Proc 1989;21:3360 –3361. Henley KS, Lucey MR, Normolle DP, Merion RM, McLaren ID, Crider BA, Mackie DS, Shieck VL, Nostrant TT, Brown KA. A double-blind, randomized, placebo-controlled trial of prostaglandin E1 in liver transplantation. Hepatology 1995;21:366 –372. Klein AS, Cofer JB, Pruett TL, Thuluvath PJ, McGory R, Uber L, Stevenson WC, Baliga P, Burdick JF. Prostaglandin E1 administration following orthotopic liver transplantation: a randomized prospective multicenter trial. Gastroenterology 1996;111:710 – 715. Totsuka E, Todo S, Zhu Y, Ishizaki N, Kawashima Y, Jin M, Urakami A, Shimamura T, Starzl T. Attenuation of ischemic liver injury by prostaglandin E1 analogue, misoprostol, and prostaglandin I2 analogue, OP-41483. J Am Coll Surg 1998;187:276 – 286. Abe T, Lynch S, Balderson G, Pillay P, Akiyama T, Inuzuka S, Matsunami H, Strong R. The effects of prostacyclin analog OP-41483 on normothermic liver ischemia and reperfusion injury in rats. Prostaglandins Leukot Essent Fatty Acids 1993;48: 417– 422. Suzuki S, Nakamura S, Koizumi T, Sakaguchi S, Baba S, Muro H, Fujise Y. The beneficial effect of a prostaglandin I2 analog on ischemic rat liver. Transplantation 1991;52:979 –983. Arthur M, Friedman S, Roll F, Bissell D. Lipocytes from normal rat liver release a neutral metalloproteinase that degrades basement membrane (type 4) collagen. J Clin Invest 1989;84: 1076 –1085. Upadhya A, Strasberg S. Glutathione, lactobionate, and histidine: cryptic inhibitors of matrix metalloproteinases contained in University of Wisconsin and histidine/tryptophane/ketoglutarate liver preservation solutions. Hepatology 2000;31:1115– 1122. Cursio R, Mari B, Louis K, Rostagno P, Saint-Paul MC, Giudicelli J, Bottero V, Anglard P, Yiotakis A, Dive V, Gugenheim J, Auberger P. Rat liver injury after normothermic ischemia is prevented by a phosphinic matrix metalloproteinase inhibitor. FASEB J 2002;16:93–95. Nelson A, Fingelton B, Rothenberg M, Matisian L. Matrix metalloproteinases: biologic activity and clinical implications. J Clin Oncol 2000;18:1135–1149. Brown P. Ongoing trials with matrix metalloproteinase inhibitors. Expert Opin Investig Drugs 2000;9:2167–2177. Raison J. The influence of temperature-induced phase changes on the kinetics of respiratory and other membrane-associated enzymes systems. Bioenergetics 1973;4:285. Woods H, Krebs H. Lactate production in the perfused rat liver. Biochem J 1971;125:129. Marsh D, Belzer F, Southard J. Hypothermic preservation of hepatocytes. II. Importance of Ca and amino acids. Cryobiology 1990;27:1– 8. Adkinson D, Hollwarth M, Benoit J, Parks D, McCord J, Granger D. Role of free radical in ischemia-reperfusion injury to the liver. Acta Physiol Scand 1986;548(suppl):101. Brass C, Narciso J, Gollan J. Enhanced activity of the free radical producing enzyme xanthine oxidase in hypoxic rat liver. J Clin Invest 1991;87:424 – 431. Todo S, Nevy J, Yanaga K, Podesta L, Gordon R, Starzl T.
934
195.
196. 197.
198.
199.
200.
201.
202.
203.
204.
205.
206.
207. 208.
209.
210.
211.
212.
SELZNER ET AL.
Extended preservation of human liver grafts with UW solution. JAMA 1989;261:711. Holloway CM, Harvey PR, Mullen JB, Strasberg SM. Evidence that cold preservation-induced microcirculatory injury in liver allografts is not mediated by oxygen-free radicals or cell swelling in the rat. Transplantation 1989;48:179 –188. Bretschneider H. Myocardial protection. Thorac Cardiovasc Surg 1980;28:295–302. Hatano E, Kiuchi T, Tanaka A, Shinohara H, Bretschneider HJ, Gebhard MM, Eigler FW. Hepatic preservation with histidinetryptophan-ketoglutarate solution in living-related and cadaveric liver transplantation. Clin Sci 1997;93:81– 88. Erhard J, Lange R, Scherer R, Kox WJ, Bretschneider HJ, Gebhard MM, Eigler FW. Comparison of histidine-tryptophan-ketoglutarate (HTK) solution versus University of Wisconsin (UW) solution for organ preservation in human liver transplantation. A prospective, randomized study. Transpl Int 1994;7:177–181. Drazan KE, Csete ME, Da Shen X, Bullington D, Cottle G, Busuttil RW, Shaked A. Hepatic function is preserved following liver-directed, adenovirus-mediated gene transfer. J Surg Res 1995;59:299 –304. Engelhardt JF, Ye X, Doranz B, Wilson JM. Ablation of E2A in recombinant adenoviruses improves transgene persistence and decreases inflammatory response in mouse liver. Proc Natl Acad Sci U S A 1994;91:6196 – 6200. Gao GP, Yang Y, Wilson JM. Biology of adenovirus vectors with E1 and E4 deletions for liver-directed gene therapy. J Virol 1996;70:8934 – 8943. Kroemer G. The proto-oncogene Bcl-2 and its role in regulating apoptosis. [published erratum appears in Nat Med 1997;3: 934] Ann Surg 1999;230:185–193. Shimizu S, Eguchi Y, Kamiike W, Itochy Y, Hasegawa J, Yamabe K, Tsuijimoto Y. Induction of apoptosis as well as necrosis by hypoxia and predominant prevention of apoptosis by Bcl-2 and Bcl-xL. Cancer Res 1996;56:2161–2166. Shimizu S, Eguchi Y, Kosaka H, Kamike W, Matsucla H, Tsuijimoto J. Prevention of hypoxia-induced cell death by Bcl-2 and Bcl-xL. Nature 1995;374:811– 813. Yamabe K, Shimizu S, Kamiike W, Waguri S, Eguchi Y, Hasegawa J, Okuno S, Yoshioka Y, Ito T, Sawa Y, Uchiyama Y, Tsujimoto Y, Matsuda H. Prevention of hypoxic liver cell neoctosis by in vivo human bcl-2 gene transfection. Biochem Biophys Res Commun 1998;243:217–223. Reed J, Miyashita T, Takayama S, Wang H, Sato T, Krajewski S, Aime-Sempe C, Bodrug S, Kitada S, Hanada M. BCL-2 family proteins: regulators of cell death involved in the pathogenesis of cancer and resistance to therapy. J Cell Biochem 1996;60: 23–32. Chao D, Korsmeyer S. BCL-2 family: regulators of cell death. Annu Rev Immunol 1998;16:395– 419. Maulik N, Engelman RM, Rousou JA, Flack JE, Deaton D, Das DK. Ischemic preconditioning reduces apoptosis by upregulating anti-death gene Bcl-2. Circulation 1999;100:I1369 –I1375. Takayama S, Xie Z, Reed JC. An evolutionarily conserved family of Hsp70/Hsc70 molecular chaperone regulators. J Biol Chem 1999;274:781–786. Takayama S, Sato T, Krajewski S, Kochel K, Irie S, Millan JA, Reed JC. Cloning and functional analysis of BAG-1: a novel Bcl-2-binding protein with anti-cell death activity. Cell 1995;80: 279 –284. Sawitzki B, Amersi F, Ritter T, Fisser M, Shen XD, Ke B, Busuttil R, Volk HD, Kupiec Weglinski JW. Upregulation of bag-1 by ex vivo gene transfer protects rat livers from ischemia/reperfusion injury. Hum Gene Ther 2002;13:1495–1504. Murphy BJ, Laderoute KR, Short SM, Sutherland RM. The identification of heme oxygenase as a major hypoxic stress protein in Chinese hamster ovary cells. Br J Cancer 1991;64:69 –73.
GASTROENTEROLOGY Vol. 125, No. 3
213. Bonkovsky HL, Lincoln B, Healey JF, Ou LC, Sinclair PR, Muller Eberhard U. Hepatic heme and drug metabolism in rats with chronic mountain sickness. Am J Physiol 1986;251:G467– G474. 214. Raju VS, Maines MD. Coordinated expression and mechanism of induction of HSP32 (heme oxygenase-1) mRNA by hyperthermia in rat organs. Biochim Biophys Acta 1994;1217:273–280. 215. Amersi F, Buelow R, Kato H, Ke B, Coito AJ, Shen XD, Zhao D, Zaky J, Melinek J, Lassman CR, Kolls JK, Alam J, Ritter T, Volk HD, Farmer DG, Ghobrial RM, Busuttil RW, Kupiec Weglinski JW. Upregulation of heme oxygenase-1 protects genetically fat Zucker rat livers from ischemia/reperfusion injury. J Clin Invest 1999;104:1631–1639. 216. Strubelt O, Younes M, Li Y. Protection by albumin against ischaemia- and hypoxia-induced hepatic injury. Pharmacol Toxicol 1994;75:280 –284. 217. Karwinski W, Soreide O. Allopurinol improves scavenging ability of the liver after ischemia/reperfusion injury. Liver 1997;17: 139 –143. 218. Gerbes AL, Vollmar AM, Kiemer AK, Bilzer M. The guanylate cyclase-coupled natriuretic peptide receptor: a new target for prevention of cold ischemia-reperfusion damage of the rat liver. Hepatology 1998;28:1309 –1317. 219. Bilzer M, Jaeschke H, Vollmar AM, Paumgartner G, Gerbes AL. Prevention of Kupffer cell-induced oxidant injury in rat liver by atrial natriuretic peptide. Am J Physiol 1999;276:G1137– G1144. 220. Kiemer AK, Gerbes AL, Bilzer M, Vollmar AM. The atrial natriuretic peptide and cGMP: novel activators of the heat shock response in rat livers. Hepatology 2002;35:88 –94. 221. Amersi F, Nelson SK, Shen XD, Kato H, Melinek J, KupiecWeglinski JW, Horwitz LD, Busuttil RW, Horwitz MA. Bucillamine, a thiol antioxidant, prevents transplantation-associated reperfusion injury. Proc Natl Acad Sci U S A 2002;99:8915– 8920. 222. Konukoglu D, Tasci I, Cetinkale O. Effects of cyclosporin A and ibuprofen on liver ischemia-reperfusion injury in the rat. Clin Chim Acta 1998;275:1– 8. 223. Ozaki M, Nakamura M, Teraoka S, Ota K. Ebselen, a novel anti-oxidant compound, protects the rat liver from ischemiareperfusion injury. Transpl Int 1997;10:96 –102. 224. Garcia Criado FJ, Palma Vargas JM, Valdunciel Garcia JJ, Toledo AH, Misawa K, Gomez Alonso A, Toledo Pereyra LH. Tacrolimus (FK506) down-regulates free radical tissue levels, serum cytokines, and neutrophil infiltration after severe liver ischemia. Transplantation 1997;64:594 –598. 225. Kobayashi H, Kurokawa T, Kitahara S, Nonami T, Harada A, Nakao A, Sugiyama S, Ozawa T, Takagi H. The effects of gamma-glutamylcysteine ethyl ester, a prodrug of glutathione, on ischemia-reperfusion-induced liver injury in rats. Transplantation 1992;54:414 – 418. 226. Nagai T, Egashira T, Kudo Y, Yamanaka Y, Shimada T. Attenuation of dysfunction in the ischemia-reperfused liver by glycyrrhizin. Jpn J Pharmacol 1992;58:209 –218. 227. Zhong Z, Froh M, Connor HD, Li X, Conzelmann LO, Mason RP, Lemasters JJ, Thurman RG. Prevention of hepatic ischemiareperfusion injury by green tea extract. Am J Physiol 2002;283: G957–G964. 228. Sewerynek E, Reiter RJ, Melchiorri D, Ortiz GG, Lewinski A. Oxidative damage in the liver induced by ischemia-reperfusion: protection by melatonin. Hepatogastroenterology 1996;43: 898 –905. 229. Yokota R, Fukai M, Shimamura T, Suzuki T, Watanabe Y, Nagashima K, Kishida A, Furukawa H, Hayashi T, Todo S. A novel hydroxyl radical scavenger, nicaraven, protects the liver from warm ischemia and reperfusion injury. Surgery 2000;127:661– 669. 230. Ikebe N, Akaike T, Miyamoto Y, Hayashida K, Yoshitake J,
September 2003
231.
232.
233.
234.
235.
236.
237.
238.
239.
240.
241.
242.
243.
244.
245.
246.
Ogawa M, Maeda H. Protective effect of S-nitrosylated alpha(1)protease inhibitor on hepatic ischemia-reperfusion injury. J Pharmacol Exp Ther 2000;295:904 –911. Tanaka J, Malchesky PS, Omokawa S, Goldcamp JB, Harasaki H, Vogt DP, Broughan TA, Nose Y. Effects of prostaglandin I2, superoxide dismutase, and catalase on ischemia-reperfusion injury in liver transplantation. ASAIO Trans 1990;36:M600 – M603. Singh AK, Mani H, Seth P, Gaddipati JP, Kumari R, Banuadha KK, Sharma SC, Kulshreshtha DK, Maheshwari RK. Picroliv preconditioning protects the rat liver against ischemia-reperfusion injury. Eur J Pharmacol 2000;395:229 –239. Wu TW, Fung KP, Zeng LH, Wu J, Nakamura H. Propyl gallate as a hepatoprotector in vitro and in vivo. Biochem Pharmacol 1994;48:419 – 422. Giakoustidis D, Papageorgiou G, Iliadis S, Kontos N, Kostopoulou E, Papachrestou A, Tsantilas D, Spyridis C, Takoudas D, Botsoglou N, Dimitriadou A, Giakoustidis E. Intramuscular administration of very high dose of alpha-tocopherol protects liver from severe ischemia/reperfusion injury. World J Surg 2002; 26:872– 877. Soltys K, Dikdan G, Koneru B. Oxidative stress in fatty livers of obese Zucker rats: rapid amelioration and improved tolerance to warm ischemia with tocopherol. Hepatology 2001;34:13–18. Tsimoyiannis EC, Moutesidou KJ, Moschos CM, Karayianni M, Karkabounas S, Kotoulas OB. Trimetazidine for prevention of hepatic injury induced by ischaemia and reperfusion in rats. Eur J Surg 1993;159:89 –93. Wu TW, Hashimoto N, Au JX, Wu J, Mickle DA, Carey D. Trolox protects rat hepatocytes against oxyradical damage and the ischemic rat liver from reperfusion injury. Hepatology 1991;13: 575–580. Chen CF, Wang D, Hwang CP, Liu HW, Wei J, Lee RP, Chen HI. The protective effect of niacinamide on ischemia-reperfusioninduced liver injury. J Biomed Sci 2001;8:446 – 452. Peralta C, Xaus C, Bartrons R, Leon OS, Gelpi E, Rosello Catafau J. Effect of ozone treatment on reactive oxygen species and adenosine production during hepatic ischemia-reperfusion. Free Radic Res 2000;33:595– 605. Elimadi A, Sapena R, Settaf A, Le Louet H, Tillement J, Morin D. Attenuation of liver normothermic ischemia-reperfusion injury by preservation of mitochondrial functions with S-15176, a potent trimetazidine derivative. Biochem Pharmacol 2001;62: 509 –516. Hertl M, Hertl MC, Malago M, Broelsch CE. In vivo protection of the pig liver against ischemia/reperfusion injury by tauroursodeoxycholate. Langenbecks Arch Surg 1999;384:461– 466. Ishigami F, Naka S, Takeshita K, Kurumi Y, Hanasawa K, Tani T. Bile salt tauroursodeoxycholic acid modulation of Bax translocation to mitochondria protects the liver from warm ischemiareperfusion injury in the rat. Transplantation 2001;72:1803– 1807. Wang Y, Lawson JA, Jaeschke H. Differential effect of 2-aminoethyl-isothiourea, an inhibitor of the inducible nitric oxide synthase, on microvascular blood flow and organ injury in models of hepatic ischemia-reperfusion and endotoxemia. Shock 1998; 10:20 –25. Fiegen RJ, Rauen U, Hartmann M, Decking UK, de Groot H. Decrease of ischemic injury to the isolated perfused rat liver by loop diuretics. Hepatology 1997;25:1425–1431. Lim SP, Andrews FJ, Christophi C, O’Brien PE. Microvascular changes in liver after ischemia-reperfusion injury. Protection with misoprostol. Dig Dis Sci 1994;39:1683–1690. Yokoyama I, Kobayashi T, Negita M, Hayashi S, Yasutomi M, Katayama A, Uchida K, Takagi H. Liberation of vasoactive substances and its prevention with thromboxane A2 synthase inhibitor in pig liver transplantation. Transpl Int 1996;9:76 – 81.
PROTECTION AGAINST LIVER ISCHEMIA
935
247. Sugawara Y, Harihara Y, Takayama T, Makuuchi M. Suppression of cytokine production by thromboxane A2 inhibitor in liver ischemia. Hepatogastroenterology 1998;45:1781–1786. 248. Kobayashi T, Esato K, Morita N, Noshima NS. Effects of thromboxane A2 synthesis inhibitor (OKY-046) on total liver ischemia in rats. Int Surg 1996;81:115–118. 249. Kitayama Y, Yamanaka N, Kawamura E, Kuroda N, Okamoto E. Hepatoprotective effect of the endothelin receptor antagonist TAK-044 against ischemia-reperfusion injury in the canine liver. Hepatology 1997;25:938 –942. 250. Araya J, Tsuruma T, Hirata K, Yagihashi A, Watanabe N. TCV116, an angiotensin II type 1 receptor antagonist, reduces hepatic ischemia-reperfusion injury in rats. Transplantation 2002;73:529 –534. 251. Konrad T, Bloechle C, Haller G, Broelsch CE, Usadel KH, Kusterer K. Verapamil and flunarizine protect the isolated perfused rat liver against warm ischemia and reperfusion injury. Res Exp Med (Berl) 1995;195:61– 68. 252. Kim YI, Hwang YJ, Song KE, Yun YK, Lee JW, Chun BY. Hepatocyte protection by a protease inhibitor against ischemia/ reperfusion injury of human liver. J Am Coll Surg 2002;195:41– 50. 253. Jung SE, Yun IJ, Youn YK, Lee JE, Ha J, Noh DY, Kim SJ, Oh SK, Choe KJ. Effect of protease inhibitor on ischemia-reperfusion injury to rat liver. World J Surg 1999;23:1027–1031. 254. Kushimoto S, Okajima K, Uchiba M, Murakami K, Harada N, Okabe H, Takatsuki K. Role of granulocyte elastase in ischemia/reperfusion injury of rat liver. Crit Care Med 1996;24: 1908 –1912. 255. Lentsch A, Yoshidome H, Warner R, Ward P, Edwards M. Secretory leukocyte protease inhibitor in mice regulates local and remote organ inflammatory injury induced by hepatic ischemia/ reperfusion. Gastroenterology 1999;117:953–961. 256. Li XK, Matin AF, Suzuki H, Uno T, Yamaguchi T, Harada Y. Effect of protease inhibitor on ischemia/reperfusion injury of the rat liver. Transplantation 1993;56:1331–1336. 257. Cursio R, Gugenheim J, Ricci JE, Crenesse D, Rostagno P, Maulon L, Saint Paul MC, Ferrua B, Mouiel J, Auberger P. Caspase inhibition protects from liver injury following ischemia and reperfusion in rats. Transpl Int 2000;13(suppl 1):S568 – S572. 258. Takeyoshi I, Sunose Y, Iwazaki S, Tsutsumi H, Aiba M, Kasahara M, Ohwada S, Matsumoto K, Morishita Y. The effect of a selective cyclooxygenase-2 inhibitor in extended liver resection with ischemia in dogs. J Surg Res 2001;100:25–31. 259. Suzuki S, Toledo Pereyra LH, Rodriguez FJ, Cejalvo D. Neutrophil infiltration as an important factor in liver ischemia and reperfusion injury. Modulating effects of FK506 and cyclosporine. Transplantation 1993;55:1265–1272. 260. Kurokawa T, Kobayashi H, Nonami T, Harada A, Nakao A, Sugiyama S, Ozawa T, Takagi H. Beneficial effects of cyclosporine on postischemic liver injury in rats. Transplantation 1992;53: 308 –311. 261. Amersi F, Dulkanchainun T, Nelson SK, Farmer DG, Kato H, Zaky J, Melinek J, Shaw GD, Kupiec Weglinski JW, Horwitz LD, Horwitz MA, Busuttil RW. A novel iron chelator in combination with a P-selectin antagonist prevents ischemia/reperfusion injury in a rat liver model. Transplantation 2001;71:112–118. 262. Mizuta K, Ohmori M, Miyashita F, Kitoh Y, Fujimura A, Mori M, Kanno T, Hashizume K, Kobayashi E. Effect of pretreatment with FTY720 and cyclosporin on ischaemia-reperfusion injury of the liver in rats. J Pharm Pharmacol 1999;51:1423–1428. 263. Anselmo DM, Amersi FF, Shen XD, Gao F, Katori M, Lassman C, Ke B, Coito AJ, Ma J, Brinkmann V, Busuttil RW, Kupiec Weglinski JW, Farmer DG. FTY720 pretreatment reduces warm hepatic ischemia reperfusion injury through inhibition of T-lymphocyte infiltration. Am J Transplant 2002;2:843– 849.
936
SELZNER ET AL.
264. Camargo CA Jr, Madden JF, Gao W, Selvan RS, Clavien PA. Interleukin-6 protects liver against warm ischemia/reperfusion injury and promotes hepatocyte proliferation in the rodent. Hepatology 1997;26:1513–1520. 265. Colletti LM, Remick DG, Campbell DA Jr. LPS pretreatment protects from hepatic ischemia/reperfusion. J Surg Res 1994; 57:337–343. 266. Natori S, Fujii Y, Kurosawa H, Nakano A, Shimada H. Prostaglandin E1 protects against ischemia-reperfusion injury of the liver by inhibition of neutrophil adherence to endothelial cells. Transplantation 1997;64:1514 –1520. 267. Okajima K, Harada N, Uchiba M. Ranitidine reduces ischemia/ reperfusion-induced liver injury in rats by inhibiting neutrophil activation. J Pharmacol Exp Ther 2002;301:1157–1165. 268. Sakakura Y, Kaibori M, Oda M, Okumura T, Kwon AH, Kamiyama Y. Recombinant human hepatocyte growth factor protects the liver against hepatic ischemia and reperfusion injury in rats. J Surg Res 2000;92:261–266.
GASTROENTEROLOGY Vol. 125, No. 3
269. Dulkanchainun TS, Goss JA, Imagawa DK, Shaw GD, Anselmo DM, Kaldas F, Wang T, Zhao D, Busuttil AA, Kato H, Murray NG, Kupiec Weglinski JW, Busuttil RW. Reduction of hepatic ischemia/reperfusion injury by a soluble P-selectin glycoprotein ligand-1. Ann Surg 1998;227:832– 840.
Received December 23, 2002. Accepted May 22, 2003. Address requests for reprints to: Pierre-Alain Clavien, M.D., Ph.D., Department of Visceral Surgery and Transplantation, University Hospital of Zurich, Ramistrasse 100, 8091 Zurich, Switzerland. e-mail:
[email protected]; fax: (41) 1-255-44-49. Supported by a grant from the National Institutes of Health (DK54048) and from the Swiss National Science Foundation (SNF3200-061411) (to P.-A.C.). N.S. and H.R. contributed equally to this work.