«H HIV-In. ». . $3. 4% (lat “‘3 ‘— 6&2 This is to certify that the thesis entitled THE ROLE OF P-SELECTIN AND ITRACELLULAR ADHESION MOLECULE-1 IN HEPATIC ISCHEMIA/REPERFUSION INJURY presented by Curtis Steven Young has been accepted towards fulfillment of the requirements for Masters Sur e degree in g ry W chr Elahé Crockett, Ph.D. Major professor Date . Gig/ZOO] 0-7639 MS U is an Affirmative Action/Equal Opportunity Institution LIBRARY Michigan State University PLACE IN RETURN Box to remove this checkout from your record. To AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 6/01 cJCIRC/DateDuthS‘pJS THE ROLE OF P-SELECTIN AND INTERCELLULAR ADHESION MOLECULE-1 IN HEPATIC lSCHEMIA/REPERFUSION INJURY BY Curtis Steven Young A THESIS Submitted to Michigan State University in partial fulfillment of the requirements for the degree of MASTERS OF SURGERY Department of Surgery 2001 ABSTRACT THE ROLE OF P-SELECTIN AND INTERCELLULAR ADHESION MOLECULE-1 IN HEPATIC lSCHEMIA/REPERFUSION INJURY By Curtis Steven Young Neutrophil recruitment and emigration represent one of the early cellular events that occur following hepatic ischemia/reperfusion (IR) injury. These two factors play a critical role in determining the extent of hepatocellular damage. Adhesion molecules on neutrophils and endothelial cells regulate the migration of neutrophils from the vasculature to the sites of injury. The goal of this research was to evaluate the role of the adhesion molecules P-selectin and intercellular adhesion molecule-1 (lCAM-1) in neutrophil-mediated hepatic IR injury utilizing P-selectin/ICAM-1 double knockout (P/l null) mice. Male wild-type C578L/6 and I’ll null mice underwent a midline Iaparotomy with 90 minutes of lobar hepatic ischemia followed by reperfusion at various time points. Liver injury was determined by changes of plasma alanine aminotransferase (ALT) levels and histopathology. Immunohistochemical staining of hepatic tissue sections was prepared to assess lCAM-1 expression and neutrophil recruitment. The results demonstrated that in both wild-type and PH null mice, reperfusion following ischemia induced significant hepatocellular injury. The injury was minimal in both sham operated mice and at 0 hours of reperfusion. lmmunohistochemistry of lCAM-1 confirmed the absence of expression in the PH null mice, as well as constitutive and inducible expression of lCAM-1 in wild-type mice. Histopathology and immunohistochemical staining revealed recruitment of neutrophils into the hepatic parenchyma after IR in both wild-type and P/l null mice. There was no significant difference in hepatocellular injury in response to IR between wild-type and P/l null mice. This investigation suggests that P- selectin and lCAM-1 do not play a critical role in neutrophil recruitment and hepatocellular injury following IR. Copyright by Curtis Steven Young 2001 I would like to dedicate this thesis to my daughters, Elizabeth and Amanda, who lovingly remind me of why I work so hard in the first place. ACKNOWLEDGMENTS First, I would like to express my deepest gratitude to Dr. Richard E. Dean, MD, Professor and Chairman of the Department of Surgery forthe opportunity to become a surgical resident here at Michigan State University and the privilege to pursue basic science research. His dedication to the residency program and research experience has provided a nurturing environment that has fostered my development as both a surgeon and scientist. I would also like to express my gratitude to my major professor, Dr. Elahe Crockett, Ph.D., Director of Basic Science Research Program and the Chair of my Research Thesis Committee, for her academic counseling, advice, encouragement, as well as assistance throughout the course of this study. I would especially like to thank Dr. Alan Davis, Ph.D., for his invaluable advice, support and assistance with this research project, and statistical analysis. I am particularly grateful for his insight and approach to research. I would like to acknowledge the other members of my graduate committee: Dr. Douglas F. Naylor, MD, for his advice and dedication to teaching; Dr. L. Rao Kareti, MD, for his advice and technical expertise; and Dr. Jack Harkema, D.V.M., Ph.D., for his teaching and assistance with histopathologic examination of tissues, as well as his critique of the manuscript. I owe a world of thanks to Crystal Remelius, 8.8., Research Assistant, and Karen Hess, M.S., Research Assistant for their technical assistance, support and patience. vi I also owe my deepest thanks to my friend and colleague, Dr. Juan Palma, MD, for all of his contributions to this project. He was an indispensable resource for his technical expertise in the animal model, as well as his insight into this area of research. To my dear friend and colleague, Dr. Ben Mosher, MD, I thank him for laying the foundation for this project. I am also thankful for his encouragement and for being the standard to which I compared myself, but could never achieve. Finally, I would like to thank my wife, Carmela, for her support and dedication. vii TABLE OF CONTENTS LIST OF TABLES .................................................................................. ix LIST OF FIGURES ................................................................................. x LIST OF ABBREVIATIONS ..................................................................... xi INTRODUCTION ................................................................................... 1 REVIEW OF THE LITERATURE ............................................................... 5 Hepatic lschemia/Reperfusion Injury ................................................. 5 Early Phase ........................................................................ 5 Late Phase ....................................................................... 1O lntercellular Adhesion Molecules .................................................... 1 1 Regulation of Hepatic Adhesion Molecule Expression ......................... 13 Adhesion Molecules and Neutrophil-Induced Liver Damage ................. 17 Adhesion Molecule Deficient Mice in Models of Ishemia/Reperfusion.....20 P-selectin/lCAM-I Double Mutant Mice in Models of Inflammation.........29 MATERIALS AND METHODS ................................................................ 31 RESULTS .......................................................................................... 40 DISCUSSION ...................................................................................... 52 REFERENCES .................................................................................... 65 viii LIST OF TABLES TABLE 1: Summary of Plasma ALT Levels in Wild-Type and P/l Null Mice After Sham Operation and IR ....................................... 44 FIGURE 1: FIGURE 2: FIGURE 3: FIGURE 4: FIGURE 5: LIST OF FIGURES Model of Partial Hepatic lschemia ....................................... 34 Verification of ICAM-1 Deficiency in P/I Null Mice .................... 41 Hepatocellular Injury After 90 Minutes of Hepatic lschemia With Various Reperfusion Times — Wild-Type vs. P/l Null Mice ......... 45 Demonstration of Hepatocellular Injury by Histopathology — Hematoxylin and Eosin Staining .......................................... 47 Demonstration of Neutrophil Recruitment by Immunohistochemical Staining ............................................ 50 LIST OF ABBREVIATIONS Adenosine Triphosphate .................................................................................. ATP Alanine Aminotransferase ................................................................................ ALT Aspartate Aminotransferase ............................................................................. AST Cytokine-lnduced Neutrophil Chemoattractant .............................................. CINC Epithelial Neutrophil Activating Protein-78 ................................................. ENA-78 lntercellular Adhesion Molecule-1 ............................................................... lCAM-1 Interleukin-1 ..................................................................................................... IL-1 Interleukin-6 ..................................................................................................... lL-6 Interleukin-8 ..................................................................................................... lL-8 lschemia/Reperfusion ......................................................................................... IR Lymphocyte Function-Associated Antigen-1 ................................................. LFA-1 Macrophage-1 Antigen .................................................................................. Mac-1 Macrophage Inflammatory Protein-2 ............................................................. MIP-2 Messenger Ribonucleic Acid ........................................................................ mRNA Monoclonal Antibody ....................................................................................... mAb Nitric Oxide ....................................................................................................... NO Nuclear Factor KB ........................................................................................... NFKB P-selectin/lCAM-1 Double Knockout ........................................................... P/I Null PIatelet-Endothelial Cell Adhesion Molecule-1 ........................................ PECAM-1 P-selectin Glycoprotein Ligand-1 ............................................................... PSGL-1 Reactive Oxygen Species ............................................................................... ROS Standard Error of Mean ................................................................................... SEM xi LIST OF ABBREVIATIONS Tumor Necrosis Factor on .............................................................................. TNFa Vascular Cell Adhesion Molecule-1 ........................................................... VCAM-1 Very Late Antigen ............................................................................................. VLA xii INTRODUCTION The functional consequences of depriving a tissue of its blood supply have been appreciated for many years. Recently, however, it has become evident that reperfusion, the restoration of blood flow after a period of ischemia, can place ischemic organs at risk of further cellular necrosis and thereby limit functional recovery (Carden and Granger, 2000). Much of the current understanding of ischemia/reperfusion (IR) injury is based on studies from many different organs, including brain, heart, lung, kidney, liver, intestine and skeletal muscle. Reperfusion of ischemic tissues results in an acute inflammatory response, which is characterized by an increased fluid filtration and steric hindrance of circulating blood cells in capillaries, with a concomitant recruitment of adherent and emigrating leukocytes. The results of several investigations suggest that there is a cause-effect relationship between the accumulation of inflammatory cells and the microvascular, as well as parenchymal cell, dysfunction that are elicited by IR (Oliver, 1991; Horie et al., 1997). Compromised blood flow to the liver, followed by resumption of normal blood flow causes hepatic damage known as IR injury. Hepatic IR injury is an important clinical problem that often follows circulatory shock with resuscitation, as well as many types of liver surgery. Total interruption of hepatic blood flow is sometimes necessary during repair of liver injury or during resection for tumors (Feliciano et al., 1986; Pachter and Spencer, 1979). Furthermore, the liver is subjected to both warm and cold ischemia during hepatic transplantation (Toledo-Pereyra, 1991 ). It is well established that the liver is quite sensitive to ischemia, and morbidity and mortality Increase rapidly with lengthening of the ischemic interval (Chien et al., 1977). Interruption of blood flow to the liver can result in cellular injury from the ischemia itself, but there is an additional component of injury that occurs during restoration of blood flow. Therefore, while reperfusion is essential to prevent the progression of metabolic and structural abnormalities during hepatic ischemia, paradoxically it can increase the degree of injury. The clinical consequences of hepatic IR injury include liver failure and multiple organ system failure, both of which carry high rates of morbidity and mortality (Faist et al., 1983; Keller et al., 1985). Although the sequelae of hepatic IR injury are well known, the exact mechanisms remain elusive. Several mechanisms have been proposed including enhanced production of proinflammatory mediators (Camargo et al., 1997; Clavien 1997; Jaeschke et al., 1991), recruitment of polymorphonuclear leukocytes (Jaeschke et al., 1990; Del Zoppo et al., 1991; Suzuki et al., 1993) and occlusion of flow (Ames et al., 1968; Koo et al., 1992). A few studies have described hepatic IR injury as being comprised of two phases (Jaeschke et al., 1990). The early phase develops during the first one to three hours of reperfusion. It is characterized by increased levels of liver transaminases, both alanine aminotransferase and aspartate aminotransferase. This phase is mediated through a reactive oxygen species (ROS) mechanism and correlates with the activation of Kupffer cells. The injury incurred during this phase is independent of intrahepatic neutrophils (Jaeschke and Farhood, 1991; Jaeschke et al., 1992). The late phase occurs after six to twenty-four hours of reperfusion. During this phase there is further increase in transaminases and massive emigration of neutrophils into the hepatic parenchyma (Jaeschke et al., 1993; Langdale et al., 1993; Suzuki et al., 1993). Neutrophil recruitment and emigration represent one of the early cellular events that occur following hepatic IR injury and play a critical role in determining the extent of hepatocellular damage. Adhesion molecules on neutrophils and endothelial cells regulate the migration of neutrophils from the vasculature to the sites of inflammation. The hypothesis of this study is that the cellular adhesion molecules P-selectin and intercellular adhesion molecule-1 (ICAM-1) play a pivotal role in neutrophil-mediated hepatic IR injury. The goal of this research was to evaluate the role of the adhesion molecules P-selectin and lCAM-1 in neutrophil-mediated hepatic IR injury utilizing P-selectin/lCAM-1 double knockout (P/l null) mice. Male wild-type C57BL/6 and PH null mice underwent a midline Iaparotomy with 90 minutes of lobar hepatic ischemia followed by reperfusion at various time points. Liver injury was determined by changes of plasma alanine aminotransferase (ALT) levels and histopathology. Immunohistochemical staining of hepatic tissue sections were prepared to assess lCAM-1 expression and neutrophil recruitment. Significant hepatocellular injury was induced in a model of warm hepatic ischemia/reperfusion injury in both wild-type and P/I null mice. Minimal injury was incurred in both sham operated mice and mice that underwent ischemia and 0 hours of reperfusion. lmmunohistochemistry for ICAM-1 confirmed the absence of expression in the PH null mice, as well as constitutive and inducible expression of lCAM-1 in wild-type mice. Significant neutrophil infiltration into the hepatic parenchyma after IR was identified on immunohistochemical staining in both wild-type and P/l null mice. However qualitatively, there was less neutrophil infiltration in the livers of PH null mice. Hepatocellular injury, as assessed by serum ALT levels, was not significantly different between wild-type and PH null mice. Collectively, the results of this investigation suggest that P- selectin and ICAM-1 do not play a critical role in neutrophil recruitment and hepatocellular injury following IR. REVIEW OF THE LITERATURE Hepatic lschemia/Reperfusion Injury: Early Phase As the cells become hypoxic, they shift from aerobic to anaerobic metabolism, which results in derangements in the intracellular milieu. Anaerobic glycolysis leads to increased lactic acid production and accumulation, with subsequent lowering of the intracellular pH. Hydrolysis of adenosine triphosphate (ATP) further exacerbates the decrease in pH. This naturally occurring acidosis protects against the onset of necrotic cell death in hepatocytes and other cells (Bonventure and Cheung, 1985; Gores et al., 1988; Gores et al., 1989). However, the recovery of normal a pH after reperfusion of ischemic cells then accelerates cell killing, a phenomenon known as the pH paradox (Gores et al., 1988; Currin et al., 1991; Bond et al., 1991; Bond et al., 1993; Harper et al., 1993; Zager et al., 1993; Kaplan et al., 1995; Currin et al., 1996; Qian et al., 1997; Nishimura et al., 1998; Lichtman and Lemasters, 1999). Treatments that delay the recovery of intracellular pH after reperfusion, such as Na‘lH” exchange inhibitors and Na+-free reperfusion, prevent pH dependent reperfusion injury (Lichtman and Lemasters1999). However, accelerated recovery of intracellular pH by substances such as monensin, hastens cell death after reperfusion (Bond et al., 1993; Harper et al., 1993; Zager et al., 1993; Kaplan et al., 1995; Currin et al., 1996; Qian et al., 1997; Nishimura et al., 1998; Lichtman and Lemasters 1999). Cytoprotection by an acidic intracellular pH appears to be partially mediated by suppression of degradative enzymes such as proteases and phospholipases that are suppressed by an acidic pH but are activated at normal pH during reperfusion (Harrison et al., 1991; Bronk and Gores, 1993; Arora et al., 1996; Harrison-Shostak et al.,1997). Another consequence of hepatic IR in the early phase of reperfusion injury is the activation of Kupffer cells (Jaeschke and Farhood, 1991; Lindert et al., 1992). Activated Kupffer cells release tumor necrosis factor a (TNFa), interleukin 1 (IL-1) and ROS, which aggravate and escalate the process of hepatic IR injury (Lichtman et al., 1994). Kupffer cells primed in the early phase worsen IR injury (Shiratori et al., 1994), whereas inactivation of Kupffer cells with methyl palmitate or gadolinium chloride protects against warm hepatic IR injury and decreases oxidative stress (Jaeschke and Farhood, 1991; Bremer et al., 1994). A significant consequence of Kupffer cell activation is the release of TNFa, which has local as well as systemic effects. Tumor necrosis factor or released after IR is derived from the liver and induces the release of epithelial neutrophil activating protein-78 (ENA-78), which is a powerful chemotactic agent for neutrophils (Muraoka et al., 1997). Overall, the evidence that TNFa plays a role in hepatic IR injury is substantial. The precise stimulus for its release by Kupffer cells remains poorly understood but may involve anoxia, perturbations in blood flow and ROS. Despite the experimental studies indicating a primary role for TNFa in IR injury, clinical studies have not supported these observations (Chazouilleres et al., 1992, Clavien et al., 1996). In addition to TNFa, lL-1 and lL-6 have been shown to be involved in the pathophysiology of hepatic IR injury. In vivo lL-6 release is delayed compared to TNFa and lL-1, which increase within minutes of IR injury (Wanner et al., 1996). Interleukin-1 release is elevated in rat Kupffer cells that have been isolated after reperfusion (Wanner et al., 1996). Interleukin-1 promotes ROS production (Shirasugi et al., 1997), whereas an lL-1 receptor antagonist attenuates hepatic IR injury in rat and lowers TNFa levels, suggesting that IL-1 promotes TNFa production (Shito et al., 1997). Treatment with lL-6 protects against warm hepatic IR injury in rats (Camargo et al., 1997). Yet other studies demonstrate that hypoxia induces the release of lL-6 from cultured endothelial cells and lymphocytes through activation of nuclear factor KB (NFKB), an effect not mediated by reperfusion (Muraoka et al., 1997). Epithelial neutrophil activating protein-78 is a member of a group of molecules known as chemotactic cytokines, or chemokines. These Chemoattractant proteins have a varying specificities and activities on different leukocytes. Other members of the chemokine family include IL-8, platelet factor 4, cytokine-induced neutrophil Chemoattractant (CINC), and macrophage inflammatory protein (MlP)—2 (Lichtman and Lemasters, 1999). Cytokine- induced neutrophil chemattractant is another important chemokine released from activated Kupffer cells. It is released within hours of IR injury and also acts as a potent Chemoattractant for neutrophils (Lichtman and Lemasters, 1999). Cytokine-induced neutrophil chemattractant links the early phase of hepatic IR injury mediated my Kupffer cells with the late phase of injury mediated by neutrophils (Lichtman and Lemasters, 1999). Macrophage inflammatory protein- 2 is another chemokine involved in hepatic IR injury. Its mRNA begins to increase within 3 hours of IR injury and continues to rise for at least 9 hours (Lentsch et al., 1998); both MlP—2 and CINC increase integrin expression on neutrophils, which further promotes transmigration from the microvasculature (Shanley et al., 1997). The hepatocyte is susceptible to injury by ROS. The mechanisms of oxygen-derived free radical formation include activation of phospholipase and the arachidonic acid cascade, as well as activation of Kupffer cells and neutrophils. These metabolites may also result from the breakdown products of ATP and the oxidative stress secondary to the reduction of glutathione during reperfusion (Atalla et al., 1985; Metzger and Lauterburg, 1988). Sources of ROS include endothelial cells, Kupffer cells, neutrophils and hepatocytes (Dahm et al., 1991; Drath and Karnovsky, 1975). Kupffer cells are a main source of ROS in the early phase of hepatic IR injury (Caldwell-Kenkel 1991; Jaeschke 1991; Jaeschke 1992), whereas neutrophils are the major source of ROS during the later phase of injury (Atalla et al., 1985; Marabayashi et al., 1986). These alterations disrupt enzyme systems that maintain normal cellular homeostasis. During this time, xanthine dehydrogenase is irreversibly converted to xanthine oxidase via a calcium-dependent protease (Batelli 1980; Della Corte and Stirpe, 1972; McCord 1985). As the ATP is depleted and hypoxanthine accumulates, upon reperfusion, hypoxanthine is converted to xanthine with generation of oxygen free radicals such as superoxide, hydroxyl radical and hydrogen peroxide by xanthine oxidase (McCord 1985). Recent studies have demonstrated that the microvascular reperfusion injury induced by clamping the portal triad (hepatic artery, portal vein and common bile duct) is triggered by superoxide radicals derived from the xanthine oxidase system (Muller et al., 1996). Liberation of these substances leads to denaturation of proteins and polyunsaturated fatty acids. When severe, enzyme dysfunction, lipid peroxidation and membrane disruption can occur. Superoxide, hydrogen peroxide and the hydroxyl radical are the major biologically active oxygen species (Reilly 1991). During physiologic homeostasis, these oxygen radicals are generated in low concentrations and protective biochemical mechanisms are in place to neutralize them. After a period of ischemia, followed by reperfusion, the rate of synthesis of these ROS far exceeds the capacity to convert them to inert molecules. Superoxide is thought to stimulate changes in the vascular endothelium that produce conditions that are conducive for the generation of the inflammatory response. In contrast, nitric oxide (NO) production, which is stimulated by IR injury, is protective. Nitric oxide attenuates sinusoidal perfusion failure, improves liver oxygenation, and may scavenge oxygen radicals (Lichtman and Lemasters, 1999). Nitric oxide synthesis can be rapid in endothelial cells in response to shear stress (Kuchan et al., 1994) and during hepatic hypoperfusion (Liu et al., 1998). Inhibition of NO synthase increases hepatocellular death and increases liver enzyme release after hepatic IR (Wang et al., 1995; Pannen et al., 1998). Nitric oxide scavenges ROS by reacting with superoxide to form peroxynitrite. With increased IR injury, NO synthase inhibition not only increases net superoxide production, but also increase the gene expression of P-selectin and lCAM-1, thereby promoting the adherence of neutrophils and the late phase of hepatic IR injury (Kobayashi et al., 1995; Liu et al., 1998). Hepatic lschemia/Reperfusion Injury: Late Phase Neutrophil infiltration mediates the late phase of hepatic IR injury. Once neutrophils reach the site of injury, they amplify and perpetuate the injury by releasing some of the same proinflammatory mediators that attracted them in the first place (Lichtman and Lemasters, 1999). In a study utilizing anti-neutrophil antibodies, hepatocyte necrosis decreased from 80% to 28% after 24 hours of IR injury (Jaeschke et al., 1990). Furthermore, direct infusion of neutrophils into isolated livers after warm IR increases hepatocellular damage, oxygen uptake and formation of the superoxide radical (Bilzer and Lauterburg, 1994). The mechanism of hepatocellular injury by neutrophils includes three steps. The sequence of events begins with activation of neutrophils and their sequestration in the hepatic vasculature. This process occurs predominantly in the sinusoids and appears more a passive mechanical trapping than an adhesion molecule mediated event (Jaeschke et al., 1997; Jaeschke and Smith, 1997). Next, neutrophils undergo transendothelial migration if the sinusoidal lining is intact. This process, which is dependent on [32 integrins/ICAM-1 and B1 integrinsNCAM-1, takes place more in the sinusoids and appears to be less important in postsinusoidal venules (Chosay et al., 1997). The final step is the adherence to the parenchymal cell, which can be dependent on macrophage-1 antigen (Mac-1)/unknown ligand and lymphocyte function-associated antigen-1 (LFA-1)/ICAM-1 (Jaeschke and Smith, 1997). Subsequent hepatocellular 10 necrosis is dependent upon the release of the proteases cathepsin G and elastase from the adherent neutrophils. lntercellular Adhesion Molecules In the late phase of hepatic IR injury, neutrophils first marginate to the sinusoidal wall and subsequently migrate across the endothelium into the space of Disse (Lichtman and Lemasters, 1999). If the endothelial cell damage is severe, neutrophils may have direct access to parenchymal cells without undergoing transmigration. However, when the endothelial cell barrier is intact, transmigration requires intercellular adhesion molecules. The activation and recruitment of neutrophils into the liver is the critical step in the pathogenesis of IR injury. lntercellular adhesion molecules are cell surface glycoproteins that are not only involved in cell-cell interactions, but also cell-matrix interactions as well. These molecules are crucial for leukocyte adhesion to the endothelium, transmigration, binding to target cells and cytotoxicity (Jaeschke et al.,1997). On the basis of structure and sequence homology, there are three main families of adhesion molecules that participate in inflammatory processes: integrins, the immunoglobulin gene superfamily, and selectins. The role of adhesion molecules has been most extensively studied in animal models of neutrophil-induced organ injury (Jaeschke et al., 1997). Investigations utilizing in vitro systems and intravital microscopy have identified several discrete steps involving the different adhesion molecules (Granger 1994; Kishimoto 1994). Since these events take place in the postcapillary venules, the 11 rapidly flowing neutrophils must be slowed down. This is accomplished by the expression of selectins. Following a rapid upregulation of P-selectin by mobilization of Weibel-Palade bodies in endothelial cells there is transcriptional activation of both P-selectin and E-selectin (McEver et al., 1995). The interaction between selectins expressed on endothelial cells with their ligands on neutrophils results in tethering and rolling of neutrophils along the vessel wall. During this hindered movement in the vicinity of an inflammatory focus, the neutrophils are exposed to a multitude of chemotactic factors, including cytokines, chemokines, complement and platelet activating factor. These inflammatory mediators induce upregulation of Mac-1 and increase the affinity of LFA-1 (CD11a/CD18) on neutrophils (Jaeschke and Smith, 1997). In addition, it has been shown that binding of neutrophil L-selectin to its ligand induced up-regulation of Mac-1 adhesion molecule expression on the surface of neutrophils (Crockett-Torabi et al., 1995). Further, cytokines can induce the expression of ICAM-1 on endothelial cells through transcription. As a result, these events lead to firm adhesion of neutrophils to the vessel wall mainly through interactions between [3; integrins and lCAM-1 (Granger and Kubes, 1994; Smith 1992). The next step is the migration of neutrophils through the endothelial cell barrier toward the inflammatory focus, which is facilitated by gradients of chemotactic factors. Transendothelial migration not only requires 82 integrin-lCAM-1 interaction, it also requires platelet-endothelial cell adhesion molecule-1 (PECAM-1), which is expressed on both neutrophils and endothelial cells (Wakelin et al., 1996). While 12 this mechanism applies to several organs, including heart, intestine, skeletal muscle and skin, there are a few significant differences in the liver. Regulation of Hepatic Adhesion Molecule Expression The integrin family of adhesion molecules consists of membrane glycoproteins that are made up of a8 heterodimers (Ruoslahti 1991). Two integrin receptor families that are important for inflammatory reactions define the very late antigens (VLA) (B1) and the leukocyte integrins ((32). The very late antigens and the leukocyte integrins are found on the surface of many cells including Kupffer cells, pit cells, monocytes, T lymphocytes and neutrophils (Bioulac—Sage et al., 1996). Quantitatively, the 82 integrin LFA-1(CD11a/CD18) is highly expressed on all leukocytes (Jaeschke et al., 1997). However, Mac-1 (CD11b/CD18) is present primarily on neutrophils, Kupffer cells, and monocytes. It is present to a lesser degree on pit cells and is not expressed on T cells (Jaeschke et al., 1997; Bioulac—Sage et al., 1996). Very late antigen-4 (VLA-4) (CD49d/C029), a B1 integrin, is expressed mainly on T cells, pit cells and monocytes, and less on neutrophils and Kupffer cells (Bioulac-Sage et al., 1996; Essani et al., 1997). The expression of integrins is regulated at multiple levels. First, synthesis of LFA—1 and Mac-1 takes place during hematopoietic maturation (Miller et al., 1986). Second, a further increase of cell surface receptors can occur during cell activation (Strassman et al., 1985). Finally, cell activation can lead to conformational changes in the adhesion molecule, resulting in exposure of ligand 13 binding sites. An increase in affinity after stimulation has been demonstrated for both LFA—1 (Keizer et al., 1988) and VLA-4 (Shimizu et al., 1990). The activation of integrins, particularly Mac-1, was observed in pathophysiological situations relevant to hepatic injury in vivo. Macrophage-1 antigen expression on circulating neutrophils was demonstrated in several models of inflammation including hepatic ischemia (Jaeschke et al., 1993), endotoxemia (Essani et al., 1995; Witthaut et al., 1994) and sepsis (Zhang et al., 1994). The inflammatory mediators that upregulate Mac-1 in these conditions include activated compliment factors, TNFa and platelet activating factor (PAF) (Essani et al., 1995; Witthaut et al., 1994; Zimmerman and McIntyre, 1988). The immunoglobulin gene superfamily includes ICAM-1, lCAM-2, lCAM-3, vascular endothelial adhesion molecule-1 (VCAM-1) and PECAM-1. In general, ICAM-2, lCAM-3 and PECAM-I are constitutively expressed on vascular lining cells, while ICAM-1 and VCAM-1 are both inducible (Jaeschke et al., 1997). In normal liver, there is low constitutive expression of lCAM-1 on the entire hepatic endothelium and Kupffer cells (Essani et al., 1995; Farhood et al., 1995; Jaeschke et al., 1996; Mochida et al., 1996; Ohira et al., 1995; Scoazec and Feldmann, 1994; Steinhoff and Brandt, 1996). However lCAM-1 is not detected on hepatocytes (Essani et al., 1995; Farhood et al., 1995; Ohlinger et al., 1993; Scoazec et al., 1994; Steinhoff and Brandt, 1996) or stellate cells (Hellerbrand et al., 1996). During inflammation, ICAM-1 is strongly upregulated on virtually all liver cells, including all endothelial cells, Kupffer cells, stellate cells and hepatocytes (Essani et al., 1995; Farhood et al., 1995; Jaeschke et al., 1996; 14 Mochida et al., 1996; Ohira et al., 1995; Scoazec et al., 1994; Steinhoff 1996; Hellerbrand 1996; Volpes 1990). Studies with isolated hepatocytes (Kvale and Brandzaeg, 1993; Mickelson et al., 1995; Morita et al., 1994) and endothelial cells (Ohira et al., 1994) have demonstrated that TNFa, IL-1 and interferon-y can induce ICAM-1 expression. There is low VCAM-1 expression on endothelial cells of arteries and veins, as well as on bile duct epithelial cells in control livers. But sinusoidal endothelial cells are very weakly stained (Essani et al., 1997). After administration of endotoxin, TNFa, or IL-1 in vivo, there is increased staining of VCAM-1 on all endothelial cells and Kupffer cells (Essani et al., 1997; Van Oosten et al., 1995). There has been no constitutive or inducible expression of VCAM-1 identified on hepatocytes (Essani et al., 1997; Van Oosten et al., 1995). Platelet-endothelial cell adhesion molecule-1, which is constitutively expressed on the endothelial cells of large vessels in both human and rodent livers, cannot be Induced by endotoxin or cytokines (Jaeschke 1997; Scoazec et al., 1991). It has been demonstrated to be important for leukocyte transendothelial migration in extrahepatic tissue (Jaeschke and Smith, 1997; Wakelin et al., 1996). The selectin family consists of three closely related cell surface molecules with differential expression by leukocytes (L-selectin, CD62L), platelets (P- selectin, CD62P), and vascular endothelium (E-selectin, CDS2E). Each of these molecules contains a calcium-dependent Iectin domain, an epidermal growth 15 factor-like domain and several complement binding repeats as the extracellular portion of the molecule (McEver et al., 1995; Tedder et al., 1995). L-selectin is constitutively expressed on neutrophils and is shed from the cell surface during the exposure of the neutrophil to chemotactic factors that upregulate Mac-1 (Kishimoto et al., 1989). P-selectin is stored in a-granules of resting platelets and Weibel-Palade bodies in endothelial cells (Hsu-Lin et al., 1984; McEver et al., 1989). Within minutes after activation by inflammatory and thrombogenic mediators, P-selectin is mobilized to the cell surface. These 1 mediators include histamine, complement, ROS, cytokines and thrombin (Tedder et al., 1995). Expression of P-selectin on the cell surface is short-lived. However, in vivo studies of P-selectin function suggest that it may also be important at later time points as a cytokine-induced adhesion molecule (Tedder et al., 1995). Studies have shown that P-selectin is not constitutively expressed in liver sinusoids and cannot be rapidly upregulated (Essani et al., 1998). The P- selectin gene can be activated in liver endothelial cells in response to endotoxin and cytokines (TNFa), but its expression is limited to the endothelium of large vessels and is not detected in sinusoids (Essani et al., 1995; Tedder et al., 1995). E-selectin is not found on normal human livers (Adams et al., 1994; Keelan et al., 1994; Redl et al., 1991). However, during septic shock, endotoxemia or administration of lL-1, there is expression of E-selectin on large-vessel endothelial cells and to a lesser degree on sinusoidal lining cells (Keelan et al., 1994; Redl et al., 1991). E-selectin protein production is also rapidly induced by TNFa, interferondy and substance P (Bevilacqua et al., 1987). 16 A common feature of the inducible adhesion molecules described (ICAM- 1, VCAM-1, P-selectin and E-selectin) is that each can be transcriptionally activated by TNFa and lL-1 (Essani et al., 1995, Essani et al., 1997, Essani et al., 1998). Both cytokines are strong inducers of the transcription factor NFKB and all inducible adhesion molecules have at least one NFKB binding site in their promoter region (Collin-et al., 1995). However, only ICAM-1 is upregulated in all cell types, with VCAM-1, P-selectin and E-selectin only induced in endothelial cells. This suggests that NFKB activation is necessary but not sufficient to enhance adhesion molecule expression (Jaeschke 1997). Thus a combination of several transcription factors may determine which adhesion molecule is upregulated in each liver cell type. Adhesion Molecules and Neutrophil-Induced Liver Damage Recent reports have demonstrated role for P-selectin in hepatic IR injury utilizing monoclonal antibody (mAb) blockade. Garcia-Criado et al. (1995) administered a mAb to P-selectin before and after reperfusion in a rat model of partial hepatic ischemia and showed reduced hepatic damage, decreased myeloperoxidase activity and improved survival. Another group of investigators that employed an anti-P-selectin mAb not only confirmed attenuated hepatocellular damage in a model of IR, but also found decreased neutrophil accumulation (Singh et al., 1998). In a study using soluble P-selectin glycoprotein ligand-1(PSGL-1), a ligand for P-selectin, given at the time of hepatic inflow occlusion in a warm ischemia model, there was reduced aspartate aminotransferase (AST), hepatic damage and neutrophil infiltration 17 (Dulkanchainun et al., 1998). Furthermore, there was improved survival when the soluble PSGL-1 ligand was administered before cold ischemia and at the time of reperfusion in a transplanted liver. One investigation, which utilized P-selectin deficient mice in a model of warm partial hepatic ischemia, demonstrated an important role for P-selectin in neutrophil and platelet sequestration (Yadav et al., 1999). Following 90 minutes of ischemia, there was a significant decrease in serum transaminase levels, hepatocellular necrosis, as well as neutrophil infiltration and adhesion (Yadav et al., 1999). In a model of total hepatic ischemia, there was improved survival in mice subjected to both 75 and 90 minutes of ischemia (Yadav et al., 1999). In addition, platelet sequestration, as determined by in situ immunohistochemical staining, was markedly reduced in the P-selectin deficient mice (Yadav et al., 1999) In other studies using P-selectin knockout mice in a model of partial hepatic ischemia, intravital microscopy was employed to investigate leukocyte recruitment in the postischemic hepatic vasculature. An increase in rolling leukocytes was observed in wild-type mice subjected to 20 minutes of ischemia and 5 hours of reperfusion (Zibari et al., 1998). This phenomenon was not observed in control mice, P-selectin deficient mice, or wild-type mice treated with a mAb to P-selectin (Zibari et al., 1998). Furthermore, when this same group looked at the number of rolling, saltating and adherent leukocytes in terminal hepatic venules after IR, there were significant increases in wild-type mice, 18 whereas in P-selectin deficient mice these responses were absent (Sawaya et al., 1999). Some studies have concluded that P-selectin, in contrast to many other organs, is not required for sinusoidal neutrophil sequestration, transendothelial migration, and subsequent neutrophil-induced liver parenchymal injury. Since there are no WeibeI-Palade bodies in sinusoidal endothelial cells of normal liver (Wisse 1972; Irving et al., 1984), P-selectin expression in the liver must be under transcriptional control (Essani et al., 1998). In a model of endotoxemia, P- selectin mRNA formation was demonstrated in Kupffer cells as well as endothelial cells (Essani et al., 1995). In a model of galactosamine and Salmonella abortus equi endotoxemia, immunohistochemical analysis indicated that P-selectin was expressed on endothelial cells of the periportal and central vein areas, with no P-selectin detected on sinusoidal endothelial cells (Essani et al., 1998). Furthermore, administration of an anti-P-selectin mAb neither reduced the number of accumulating neutrophils in the sinusoids, nor attenuated liver injury. I A study that focused on the role of L-selectin and lCAM-1 in hepatic IR injury, utilized mice that were deficient in L-selectin, ICAM-1, or both. A model of left lateral lobe IR injury was employed, with ischemia times of 30, 60 and 90 minutes, followed by reperfusion times of 15 minutes, 1 hour, 6 hours and 24 hours. In mice that were deficient in L-selectin alone, there was a wide range of protective effects on IR injury observed, including decreases in neutrophil infiltration, prevention of microcirculatory failure, lower transaminase levels 19 (Yadav et al., 1998). There was also improved overall survival in a model of total hepatic ischemia (Yadav et al., 1998). In mice deficient in lCAM-1, the protective effects were limited to a decrease in both neutrophil infiltration and transaminase levels only at 6 hours of reperfusion (Yadav et al., 1998). The L-selectin/lCAM-1 double mutant mice demonstrated a reduction in injury with a pattern that was very similar to the L-selectin deficient mice. However, unlike either the L-selectin or lCAM-1 deficient mice, there was a statistically significant reduction in transaminase levels after 24 hours of reperfusion, suggesting a possible additive effect (Yadav et al., 1998). Adhesion Molecule Deficient Mice in Models of IshemialReperfusion Recent developments in molecular genetic analyses of mice have made it possible to generate null mutations in any gene of interest (Hynes and Wagner, 1996). Much useful information has already been obtained from such “knockout” mice. Transgenic mice, to which genes have been added, have been available for longer and provide another avenue for manipulating the genome of mice. Moreover, it is now possible to generate subtle mutations and tissue-specific or regulatable expression or ablation of specific genes (Rossant and Naggy, 1995; Marth 1996). Mice can be interbred to combine mutations in multiple genes, providing animal models for multigenic defects. Even though the use of blocking agents is particularly applicable to designing strategies to prevent IR injury in a specific organ with potential clinical application, they provide only limited insight into the mechanism of injury. Moreover, they do not convincingly establish the involvement of a specific 20 adhesion molecule. Other ligands may interact with other adhesion molecules or receptors resulting in nonspecific stimulatory or inhibitory effects. In particular, for a substance such as soluble PSGL —1, various studies have shown concomitant blockage of L-selectin (Spertini et al., 1996; Tu et al., 1996), as well as E-selectin (Asa et al., 1995). One of primary advantages of knockout technology over immunological manipulation is the ability to specifically and completely ablate the gene product. This eliminates the potential inherent ability of antibodies to trigger secondary cellular responses or nonspecific immunological effects. Antibodies against adhesion molecules have been shown to mimic the effects of the natural ligands of the surface target molecule (Schachner 1993). For example, antibodies to neural cell adhesion molecule activate G-protein—coupled phospholipase C signaling pathways, increase cytosolic free calcium concentration, decrease intracellular pH, and activate protein kinases (Schuch et al., 1989). Antibodies to adhesion molecules have been shown to have multiple effects on signaling and genetic responses in cells (Schachner 1993). In cells treated with antibodies to B1 integrins, for example, there is increased tyrosine phosphorylation of a number of proteins (Kornberg et al., 1991). It is also possible for an antibody to induce a different set of signals and responses than the actual ligand. Furthermore, since antibodies vary in their specificity, blocking a particular epitope does not necessarily neutralize the functional activity of the protein to which the antibody is directed. In addition, the local availability of the blocking mAbs may be compromised, particularly in the presence of extensive occlusion 21 of the vasculature. Targeted gene inactivation technology thus affords the opportunity to study the role of a specific molecule in a pure system. Knockout technology is only in current routine use in mice. The mouse has a long history of use as a key biological tool for inflammation research. The advantages include its well-studied genetics, short reproductive cycle and well- characterized models. Disadvantages relate to the small size of mice for some sample preparation and variability in susceptibility to inflammatory stimuli due to genetic strain differences. Nonetheless, significant progress in the understanding of the mechanisms of inflammation and the immune response has been gained in the use of knockout mice. Neurologic lschemia/Reperfusion Injury There is accumulating evidence that cerebral ischemia elicits an inflammatory response that is augmented by reperfusion. Leukocyte infiltration has been well documented after cerebral IR and is known to mediate local tissue damage as well as alterations in microvascular perfusion (Frenette and Wagner, 1996; Garcia et al., 1994; Hallenbeck et al., 1985; Kochanek and Hallenbeck, 1992). In a murine model of reperfused stroke, it was demonstrated that neutrophil depletion before stroke reduced cerebral infarct volume by three-fold and improved functional outcome (Connolly et al., 1996). To investigate the role of P-selectin in stroke, Connolly, et al. utilized a murine model of focal cerebral IR via middle cerebral artery occlusion in homozygous P-selectin null mice and a strategy of administering a functionally blocking P-selectin antibody. Expression of P-selectin in the postischemic 22 cerebral cortex was localized to the ipsilateral cerebral microvascular endothelial cells by immunohistochemistry and by specific accumulation of radiolabeled anti- murine P-selectin lgG. Furthermore, neutrophil accumulation in the ischemic cerebral cortex of mice expressing the P-selectin gene was significantly greater than that of the P-selectin null mice. These mice also demonstrated smaller infarct volumes and improved overall survival compared to the wild-type mice (Connolly et al., 1997). In addition, functional mAb blockade of P-selectin in wild- type mice led to improved early reflow and stroke outcome compared with control mice. Cerebral infarction volumes were reduced even when the blocking antibody was administered after the occlusion of the middle cerebral artery (Connolly et al., 1997). Another investigation evaluated the role of P-selectin and E-selectin in a model of transient focal cerebral IR utilizing a double knockout mouse that was deficient in both selectins. After 3 hours of ischemia via MCAO and 21 hours of reperfusion, there was no difference in infarct volume (Soriano et al., 1999). Other studies have implicated an important role for lCAM-1 in neutrophil- mediated cerebral IR injury. lCAM-1 is upregulated on microvascular endothelium and glial cells during inflammatory processes in the brain (Fabry et al., 1992; Frohman et al., 1995). Specific blockade of lCAM-1 by a mAb has been shown to attenuate cerebral damage after transient focal ischemia (Zhang et al., 1994). Two studies utilizing an lCAM-1 deficient mouse in a model of transient focal cerebral IR both demonstrated a reduction in infarct volume compared to wild-type controls (Connolly et al., 1996; Soriano et al., 1996). In 23 the various models of cerebral IR injury employed in these investigations, there was a 3.7 to 7.8-fold reduction in infarct volume in the ICAM-1 null mice. In addition, lCAM-1 null mice demonstrated a 35% increase in survival and reduced neurologic deficit compared with wild-type controls (Connolly et al., 1996). An important role for ICAM-1 in the genesis of cerebral no-reflow was suggested by the finding that cerebral blood flow to the infarcted hemisphere was 3.1-fold greater in lCAM-1 null mice compared to wild-type controls (Connolly et al., 1996) The role of Mac-1 in neutrophil infiltration after transient focal IR has been investigated as well. In studies using a mAb to Mac-1 there was a reduced Infarct volume after IR (Chen et al., 1994; Springer 1995). However, administration of this antibody was not without complication, as a partial depletion of peripheral white blood cells was noted. Investigations that utilized a mAb to the CD18 subunit have yielded conflicting results. In a feline model, the antibody had no effect on infarct volume or cerebral blood flow (Takeshima et al., 1992). Yet in a primate model, there was improved microvascular patency after IR (Mori et al., 1992). In a study of cerebral using Mac-1 deficient mice, there was a significant decrease in infarction volume by 26%. Furthermore, there was a reduction in extravasated neutrophils in the infarcted brains of the mutant mice, though this finding was not statistically significant (Soriano et al., 1999). Myocardial lschemia/Reperfusion Injury Myocardial IR injury is considered a potent stimulus for tissue destruction and possible cardiac failure (Braunwald and Kloner, 1985; Entman et al., 1991). 24 A large extent of this injury is believed to result from intensified neutrophil- endothelial cell affinity via enhanced adhesion molecule expression subsequent to myocardial IR. Clinical investigations have provided some preliminary evidence that neutrophils and endothelial cell adhesion molecules are activated in the coronary circulation under conditions of unstable angina (Buerke et al., 1994; Lefer AM et al., 1994), coronary vasospasm (Eppihimer et al., 1996; Gill et al., 1996) and acute myocardial infarction (Lefer DJ et al., 1996) in humans. Studies have suggested a pivotal role for P-selectin in myocardial IR injury. It has been demonstrated that administration of monoclonal antibodies (Weyrich et al., 1993) or carbohydrate ligands (Lefer DJ et al., 1994) directed against P-selectin markedly attenuates myocardial reperfusion injury in animal models of myocardial IR. Palazzo et al. (1998) utilized P-selectin knockout mice to evaluate the role of P-selectin in myocardial IR injury. After 30 minutes of ischemia followed by 120 minutes of reperfusion, there was significant diminution of myocardial infarct size and neutrophil accumulation in the P-selectin deficient mouse heart compared to the wild-type mouse hearts. However, with prolonged coronary artery occlusion, (Le. 60 minutes) the infarct size was increased, but similar between P-selectin-deficient and wild-type mice. Yet there was no difference in PMN infiltration between 30 minutes and 60 minutes of myocardial ischemia. Therefore P-selectin deficiency may not confer cardioprotection during extended periods of coronary artery occlusion, which may be attributed to neutrophil-independent myocardial necrosis. 25 There Is a large amount of experimental evidence that suggests CD18 interactions with lCAM-1 play a significant role in neutrophil-mediated myocardial IR injury. The involvement of lCAM-1 in myocardial IR injury has been previously demonstrated (Kukielka et al., 1993; Youker et al., 1994). In addition, it has also been shown that neutrophil CD18 participates in the firm adhesion of activated neutrophils to cardiac myocytes through interaction its with lCAM-1, leading to myocyte injury and necrosis (Entman et al., 1992; Smith et al., 1989). Monoclonal antibodies against the neutrophil [lg-integrin complex CD11/CD18 have been demonstrated to have cardioprotective effects (Aral et al., 1996; Aversano et al., 1995; Lefer DJ et al., 1993; Ma et al., 1991; Simpson et al., 1988). Similarly, monoclonal antibodies against lCAM-1, the endothelial ligand for CD11/CD18, have been shown to decrease the injury produced by myocardial IR (Hartman et al., 1995; Lefer DJ et al., 1996; Ma et al., 1992; Yamazaki et al., 1993). However, other studies have reported that anti-CD18 monoclonal antibodies fail to reduce myocardial IR injury. Palazzo et al. (1998) examined the role of CD18 and lCAM-1 individually in a model of murine myocardial IR utilizing knockout mice. Mice were subjected to 30 minutes of myocardial ischemia and 120 minutes of reperfusion to determine the extent of myocardial cell necrosis and neutrophil infiltration. Myocardial infarction, as calculated in % of the area at risk, was 45.1 +/- 5.9 in wild-type mouse hearts. The extent of myocardial infarction was significantly attenuated in the CD-18-deficient mice (i.e. 19.3 +/- 5.1%), as well as in the lCAM-1-deficient mice (i.e. 17.9 +/- 3.2%). Similarly, neutrophil infiltration into 26 the IR myocardium was reduced by 54% in the CD18-deficient mice and by 32% in ICAM-1-deficient mice compared to wild-type hearts. Based on these data, there is strong evidence that both of these adhesion molecules play an important role in myocardial cell injury in IR. Lung lschemia/Reperfusion Injury Several studies have demonstrated P-selectin-mediated pulmonary PMN sequestration in models of remote tissue IlR injury, including intestine and skeletal muscle (Carden et al., 1993; Seekamp et al., 1994). However, one study investigated the role of P-selectin in pulmonary injury in models of hypothermic preservation with transplantation and direct ischemic insult. To determine whether P-selectin played a role in mediating early primary graft failure, left lungs harvested from male Lewis rats were preserved for 6 hours at 4°C and transplanted orthotopically into isogenic recipients. Recipients immunodepleted of neutrophils prior to transplantation demonstrated improved graft function and overall survival. Administration of a blocking P—selectin antibody 10 minutes prior to reperfusion diminished graft PMN infiltration and improved graft function as evidenced by reduced pulmonary vascular resistance and increased pulmonary arterial flow. Recipient survival was improved as well. To evaluate the role of P-selectin in normothermic pulmonary ischemia, mice were subjected to temporary left pulmonary artery ligation (Le. 30 or 60 minutes) and 30 minutes of reperfusion. After functional removal of the nonischemic right lung, P-selectin-deficient mice demonstrated reduced PMN infiltration, improved arterial oxygenation and improved survival compared to 27 controls. These studies indicate a central role of P-selectin in PMN recruitment and tissue injury in the setting of lung transplantation after hypothermic preservation and after normothermic ischemia. (Naka et al., 1997) Renal lschemia/Reperfusion Injury Studies in the rat utilizing mAbs have suggested a role for lCAM-1 in the pathogenesis of acute tubular necrosis. However, anti-lCAM-1 mAb may not block all lCAM-1-dependent interactions since lCAM-1 has multiple sites for integrin binding (Springer 1990). Furthermore, antibodies vary in their specificity and blocking a particular epitope does not necessarily neutralize the functional activity of the protein to which the antibody is directed. Since not all of the functions of ICAM—1 are known, it is not possible to determine in vivo or in vitro the extent to which the antibody is neutralizing. Kelly et al. (1996) studied the role of lCAM-1 in ischemic renal injury by determining the histological and functional consequences of bilateral IR injury in mice genetically deficient in lCAM-1 expression. Serum creatinine was significantly increased over baseline after bilateral renal ischemia and reperfusion at 24, 48 and 72 hours in control mice. lntercellular adhesion molecule-1-deficient mice were protected from renal IR injury. After 72 hours of reperfusion, all of the lCAM-1-deficent mice survived, whereas 50% of the control wild-type animals died. Histologic analysis of the kidneys of wild-type mice revealed significantly more tubular epithelial necrosis in the outer medulla than lCAM-1 deficient mice. Control wild-type mice had significantly more neutrophils in the outer medulla, as well as greater myeloperoxidase activity than lCAM-1- 28 deficient mice did. These data suggest an important role for ICAM-1 and leukocyte-endothelial adhesion in the pathophysiology of ischemic renal failure. P-selectinIICAM-1 Double Mutant Mice in Models of Inflammation Bullard et al first described P-selectin/ICAM-1 double mutant mice in 1995. P-selectin mutant mice were generated by gene targeting using a replacement vector designed to delete exons 3-5. Deletion of a 4.5-kb region containing these exons was confirmed by Southern blotting and reverse transcriptase polymerase chain reaction. Mutant animals demonstrated a loss of expression on both endothelium and platelets. P-selectin mutant mice were bred to mice containing a mutation in the lCAM-1 gene within exon 5 (Sligh et al., 1993) to generate double homozygotes. Examination of circulating leukocyte counts demonstrated a significant increase in the number of neutrophils in P-selectinllCAM-l double mutants when compared to mice of similar genetic background. The circulating lymphocyte and monocyte counts were increased as well. The first study with these mice investigated the roles of P-selectin and lCAM-1 in the acute inflammatory response induced by S. pneumoniae within the peritoneum and lung. In a model of S. pneumoniae peritonitis, there was a complete absence of neutrophil emigration in the peritoneum in the P- selectin/lCAM-1double mutant mice, compared to a 60-70% reduction with either mutation alone (Bullard et al., 1995). The data indicate that the roles of P- selectin and ICAM-1 are unlikely to be completely independent in this model. In contrast, the combined absence of these molecules had no effect on neutrophil 29 emigration into the parenchyma of the lung in response to the same stimulus. These data suggest that neutrophil emigration through the pulmonary microvasculature can occur through adhesion pathways that do not require P- selectin or lCAM-1. Another investigation examined the role of P-selectin and lCAM-1 in traumatic brain injury utilizing P-selectin/lCAM-1 double mutant mice. Experimental traumatic brain injury elicits an acute local inflammatory response, including up-regulation of adhesion molecules and accumulation of neutrophils, in injured brain (Carlos et al., 1997; Whalen et al., 1997). In a model of traumatic brain injury there was no difference in neutrophil accumulation between P- selectin/lCAM-1 double knockout mice and wild-type mice. However, brain edema was significantly decreased in the deficient mice. In comparing histopathology and tests of memory and motor function, no significant differences between the two groups were identified (Whalen et al., 2000). These data suggest a role for adhesion molecules in the pathogenesis of brain edema independent of leukocyte accumulation (Whalen et al., 2000). 30 MATERIALS AND METHODS Animals Gene-targeted mice deficient in P-selectin and lCAM-1 (P/l null), C57BL/6- lcam1WIBaySelpm‘Bay, were purchased from the Jackson Laboratory (Bar Harbor, ME). These mice demonstrate an increased number of neutrophils in the blood and a complete loss of neutrophil emigration into the peritoneum during Streptococcus pneumoniae-induced peritonitis (Bullard 1995). Homozygotes are viable and fertile with to obvious phenotypic abnormalities. Male double- knockout mice were either bred under the guidance of the University Laboratory Animal Resources, Michigan State University, or purchased directly from the Jackson Laboratory. Wild-type mice were C57BL/6 from Charles River Laboratories, Portage, MI. Mice were maintained on 12-hour light and 12-hour dark cycles. The animals had access to food and water ad libitum. All procedures were performed according to the Michigan State University All- University Committee on Animal Use and Care guidelines. Model of Partial Hepatic lschemia/Reperfusion Injury A murine model of lobar hepatic ischemia was utilized (Colletti et al., 1990; Kawano et al., 1989; Lichtman and Lemasters, 1999, Martinez-Mier et al., 2000). Mice were anesthetized with 1-2 ml of methoxyflurane (Schering-Plough, Union, NJ) applied to gauze in a face cone, which was fashioned from a 50-ml Corning polypropylene disposable centrifuge tube (Corning, NY). Once adequate anesthesia was obtained, as determined by a negative foot pinch reflex, an intraperitoneal injection (15 mg/kg) of pentobarbital sodium (50mg/ml, Abbott 31 Laboratories, North Chicago, IL) was administered. The face cone was applied as needed to maintain anesthesia. A midline incision was made from the xiphoid process to the pubis. The small and large bowel were eviscerated and packed in normal (0.9%) saline moistened gauze (2 in. x 2 in., The Kendall Company, Mansfield, MA). The liver was exposed completely. The ligamentous attachment of the left lateral lobe was carefully divided. The median and left lateral lobes were freed. The portal circulation to both of these lobes was carefully dissected. The portal vein and hepatic artery supplying the median and left lateral lobes was then interrupted with the application of an atraumatic vascular clamp (accurate Surgical and Scientific Instruments Corporation, Westbury, NY) to the vascular pedicle. The caudate and right lateral lobes, as well as the papillary and quadrate processes of the liver were perfused to prevent intestinal congestion. This results in the induction of ischemia to approximately 70% of the liver (Kawano et al., 1989; Lichtman and Lemasters, 1999). The small and large bowel were replaced into the abdominal cavity. One ml of sterile normal saline was dripped into the abdominal cavity to replace insensible fluid losses and prevent desiccation. During hepatic ischemia, the abdomen was covered with plastic wrap (S. C. Johnson & Son, Inc., Racine, WI) to prevent evaporation. After 90 minutes of partial hepatic ischemia, the clamp was removed and reperfusion was initiated. The midline Iaparotomy was closed in a single layer using 5-0 nylon on an FS-2 needle (Ethicon, lnc., Somerville, NJ). Sterile Lactated Ringers solution (Abbott Laboratories, North Chicago, IL), 0.8 ml, was administered subcutaneously to compensate for operative blood and fluid losses. 32 Sham mice underwent the same procedure but without vascular occlusion. Mice were sacrificed after 0, 1.5, 3 and 6 hours of reperfusion. The experimental procedure is summarized in Figure 1. 33 Median Lobe Left Lateral Lobe Papillary and Quadrate Processes Caudate and Right Lateral Lobes Figure 1. Model of Partial Hepatic lschemia The blood supply to the median and left lateral lobes Is interupted with an atraumatic microvascular clamp. The total ischemia time is 90 minutes, followed by various reperfusion times. Determination of Plasma Alanine Aminotransferase Levels Plasma levels of ALT were utilized as an established marker of hepatic I/R injury. Blood samples were obtained from the right ventricle via a left anterior thoracotomy at the time of sacrifice. The blood was collected in a sterile heparinized (50pl, 100 USP Units/ml, Abbott Laboratories, North Chicago, IL) 3 cc syringe (Becton Dickinson & 00., Franklin Lakes, NJ) with a 25G needle (Becton Dickinson & 00., Franklin Lakes, NJ). The blood was centrifuged (Eppendorf Model 5415C, Brinkmann Instruments, Inc., Westbury, NY) at 14,000 rpm in 18-20°C for 15 minutes. The plasma was then transferred to two sterile 1.5 ml eppendorf microcentrifuge tubes (Brinkmann Instruments, Inc., Westbury, NY) using a sterile transfer pipette (Sarstedt, Inc., Newton, NC). The plasma was then stored at —70°C until further use. Measurement of plasma ALT was performed using a diagnostic kit from Sigma Chemical Company (St. Louis, MO). This assay is based on colorimetric measurement of transaminase in plasma by formation of 2, 4- dinitrophenylhydrazones of the keto-acids produced by the enzymes (Reitman and Frankel, 1957). Briefly, 1.0 ml of Alanine-a-KG Substrate (Sigma Chemical Company, St. Louis, MO) was pipetted into disposable glass culture tubes (VWR Scientific, Chicago, IL). Plasma samples were diluted in 0.9% saline. Next 200 pl of each plasma sample was added to each tube and vortexed (Vortex Genie 2, Fisher Scientific, Pittsburgh, PA) for 10 seconds. The samples were then incubated in a 37°C water bath (Lauda, Model M20, VWR Scientific, Chicago, IL) for 30 minutes. Next, 1.0 ml of Sigma Color Reagent was added. The reaction 35 mixture was vortexed and incubated at room temperature for 20 minutes. The reaction was stopped with 10 ml of 0.40N sodium hydroxide solution (J. T. Baker Chemical Company, Phillipsburg, NJ). The tubes were then covered with Parafilm® “M”(American National Can, Menasha, WI) and the contents mixed using gentle inversion. After 5 minutes, the absorbance was measured at 500 nm using a Spectramax Plus Microplate Reader (Molecular Devices, Sunnyvale, CA). A standard curve was generated from a plot of the OD. values at 500 nm versus a known amount of ALT activity per ml (Sigma-Frankel Units/ml) (Sigma Chemical Company, St. Louis, MO). The plasma concentration of ALT activity In the experimental samples was then calculated from the standard curve and is presented as the international unit per liter of plasma (IU/L). Histological Assessment of Hepatocellular Damage Portions of the left lateral and caudate lobes were fixed in buffered 10% formalin and embedded in paraffin. Sections were cut to 5 microns, which underwent routine staining with hematoxylin and eosin. The tissues were examined by two including individuals, a board certified veterinary pathologist (J.H.), who were blinded from the experimental treatment of the individual mice. lmmunohistochemistry for ICAM-1 and Neutrophlls lnercellular adhesion molecule-1 expression was analyzed by immunohistochemistry using an Armenian hamster lgG anti-mouse primary mAb (3E2) specific for lCAM-1 (CD54) (01541 D, PharMingen, San Diego, CA). Biopsies of the ischemic median lobe were placed in intermediate Cryomolds (Miles lnc., Elkhart, IN) and embedded in optimal cutting temperature (OCT) 36 embedding medium (Sakura Finetek USA, Inc., Torrance, CA). The samples were then frozen by floating the specimen molds in 2-methylbutane (Fisher Scientific, Pittsburgh, PA) prechilled in liquid nitrogen and then stored at -70°C. Cryosections were cut at 5 microns, and placed on glass slides (VWR Scientific, Chicago, IL) and fixed in acetone. Slides were then placed in a humidified chamber. The tissue sections were rinsed with TBS (BioRad, Hercules, CA), pH 7.4, and incubated in Tween-buffered saline(TBS) for 5 minutes. This was repeated for a total of two washes. To prevent nonspecific binding, the tissue sections were incubated in goat serum (Vector Laboratories, Inc., Burlingame, CA)/hydrogen peroxide (Sigma Chemical Company, St. Louis, M0) for 10 minutes. The tissue sections were then rinsed twice with TBS for 5 minutes per rinse. Next the tissue sections were incubated with Avidin-D (Vector Laboratories, Inc., Burlingame, CA) and incubated for 15 minutes, followed by a wash with TBS. Biotin Blocking Reagent (Vector Laboratories, Inc., Burlingame, CA) was then applied to the tissue sections and incubated for 15 minutes. Tissue sections were again washed with TBS for 5 minutes and the excess was removed. Next 100pl of the anti-ICAM-1 antibody was applied to the tissue and incubated for 60 minutes. The tissue sections were then rinsed three times with TBS for a total of 10 minutes and the excess was wiped away. Next 100 pl of the secondary antibody, biotin-conjugated mouse anti-hamster lgG mAb (12102D, PharMingen, San Diego, CA) was applied to the tissue and incubated for 30 minutes. A 10-minute TBS rinse was again performed. Vectastain ABC Reagent (Vector Laboratories, Inc., Burlingame, CA) was applied to the tissue and 37 incubated for 30 minutes, followed by a 10—minute TBS rinse. The 3,3”— diaminobenzidine (DAB) solution (Vector Laboratories, Inc., Burlingame, CA) was then applied and incubated for 10 minutes. The tissue cultures were then rinsed with 0.05M sodium bicarbonate (J. T. Baker Chemical Company, Phillipsburg, NJ) and incubated for 10 minutes. Next, DAB Enhancing solution was applied for 5-10 seconds per slide and rinsed with dHZO. The tissues were then stained in Hematoxylin (Gill’s Formula, Vector Laboratories, I nc., Burlingame, CA) for 5 minutes. The tissue sections were then rinsed with dH20 until the effluent was clear. Next the tissue sections were dipped ten times in 2% acetic acid (EM Science, Gibbstown, NJ), followed by rinsing with dH20. The tissue sections were then placed in a bluing solution (1.5 ml NH4OH (EM Science, Gibbstown, NJ) and 98.5 ml 70% EtOH) for one minute. Tissue sections were then dipped ten times in dH20 and allowed to air dry. Finally a coverslip was applied with Dako Mounting Media (Dako Corporation, Carpinteria, CA) Immunohistochemical staining for neutrophils was performed on biopsies of the ischemic lobes embedded in OCT as previously described. The primary antibody was a rat Inga anti-mouse neutrophil mAb (7/4) (CL8993AP, Cedarlane, Accurate Chemical & Scientific Corporation, Westbury, NY); while the secondary antibody was the biotin-conjugated mouse anti-rat lgG mAb (121020, PharMingen, San Diego, CA). The procedure was otherwise as described above. 38 Statistical Analysis A two-way analysis of variance was utilized to test for differences in ALT between wild-type and P/l null mice. Analysis was performed using the Number Cruncher Statistical System (Number Cruncher Statistical Systems, Kaysville, UT). A p value less than 0.05 was considered statistically significant. 39 RESULTS Verification of lCAM-1 Deficiency in PII Null Mice The lCAM-1 expression was assessed in situ by specific immunohistochemical staining for lCAM-1 (0054) utilizing a specific mAb to mouse lCAM-1. lmmunostaining demonstrated that lCAM-1 was constitutively expressed in the wild-type control mice as indicated by intense brown staining along the endothelium of the central vein, sinusoids and portal vasculature (Figure 2A). In P/l null control mice, ICAM-1 deficiency was confirmed by the lack of staining along the endothelium (Figure ZB). After hepatic IR, lCAM-1 continued to be expressed as demonstrated by the continued intense staining of the endothelium (Figure 20). Further, there was no evidence of ICAM-1 expression in the PH null mice, even after 6 hours of reperfusion (Figure 20). 40 Figure 2A. Control, wild-type mouse (no ischemia and reperfusion). Figure 23. Control, P/I null mouse (no ischemia and reperfusion). Figure 2C. Wild-type mouse after 90 minutes of ischemia and 6 hours of reperfusion. Figure ZD. P/I null mouse after 90 minutes of ischemia and 6 hours of reperfusion. 41 42 Demonstration of Hepatocellular Injury by Determination of Alanine Aminotransferase Levels Hepatic IR caused significant hepatocellular damage in a time-dependent manner as demonstrated by plasma ALT levels. The plasma ALT levels of both wild-type and P/l null mice after 90 minutes of ischemia followed by 1.5, 3 and 6 hours of reperfusion were significantly elevated compared to the sham operated mice at the same time points. However, there was no statistically significant difference in ALT levels between the wild-type and PH null mice that underwent 90 minutes of ischemia and 0 hours of reperfusion, compared to the sham group at 0 hours for both types of mice (Table 1). In addition, there was no significant difference between wild-type and PH null sham operated mice at 0, 1.5, 3 and 6 hours of reperfusion (62 1 10 vs. 49 1 5, 105 1 17 vs. 83 1 13, 78 1 10 vs. 95 1 30, 68 1 15 vs. 56 1 22 IU/L, respectively). In comparing the wild-type and PH null mice after 1.5, 3 and 6 hour reperfusion, there was a trend toward decreased injury in PM null mice compared to the wild-types (1977 1 289 vs. 2090 1 472, 3877 1 807 vs. 4267 1 489, 4233 1 495 vs. 5613 1 576 lU/L, respectively) (Figure 3). However, this was not statistically significant. 43 Table 1. Summary of Plasma ALT Levels in Wild-Type and P/l Null Mice After Sham Operation and IR. Values are expressed as the mean 1 standard error of the mean (SEM) with n = 3 for sham groups and n z 5 for the IR groups. Reperfusion Time Treatment Wild-Type PII Null 0h Sham 62110 4915 IR 86 1 13 66 1 22 15h Sham 105117 83113 IR 2090 1 472 1977 1 290 3 h Sham 78 110 95 1 30 IR 4267 1 489 3877 1 807 6 h Sham 68 115 56 1 22 IR 5613 1 576 4233 1 495 44 7000 ‘ 6000 ‘ 5000 ‘ 4000 ‘ 3000 '1 ALT (lU/L) 2000 ‘ 1000 ‘ O- UWild-type I P/l Null 0 1.5 3 6 Reperfusion Time (hrs) Figure 3. Hepatocellular Injury After 90 Minutes of Hepatic lschemia With Various Reperfusion Times - Wild-Type vs. P/l Null Mice. Values are expressed as the mean 1 SEM with n _>_ 5 in each group. 45 Demonstration of Hepatocellular Injury by Histopathology — Hematoxylin and Eosin Staining The liver lobule consists of three main regions. The periportal area includes the portal triad, which is comprised of the portal vein (PV), hepatic artery and bile duct. The pericentral area surrounds the central vein (CV). The midzonal region is found between the periportal and pericentral areas. Histopathologic evidence of hepatic IR injury is classically includes sinusoidal congestion, cytoplasmic vacuolization, hepatocellular necrosis and neutrophil infiltration (Suzuki et al., 1994). Typically, after ischemia followed by a long period of reperfusion, there is relative sparing of the periportal region, and significant necrosis in the midzonal and pericentral regions. In this study, after 90 minutes of ischemia followed by 0 hours of reperfusion, there is minimal evidence of hepatocellular injury (Figure 4A). However, following 1.5 and 3 hours of reperfusion there was moderate hepatocellular necrosis in both the wild- type and P/I null mice, and marked hepatocellular necrosis after 6 hours of reperfusion (Figure 4B and 4C). As figure 4B shows, a greater number of neutrophils infiltrated into the midzonal region of ischemic liver after 6 hours of reperfusion in wild-type mice. There was no evidence of hepatocellular injury in the corresponding non-ischemic lobes of the livers of both animal groups that underwent 90 minutes of ischemia and 1.5, 3, and 6 hours of reperfusion. 46 Figure 4A. Control, wild-type mouse (no ischemia and reperfusion). Figure 4B. Wild-type mouse after 90 minutes of warm ischemia, followed by 6 hours of reperfusion. There is significant hepatocellular necrosis in the pericentral and midzonal regions, with relative sparing of the periportal areas. Note the presence of neutrophils in the midzonal region. Figure 4C. P/l null mouse after 90 minutes of warm ischemia followed by 6 hours of reperfusion. Again note the significant hepatocellular necrosis in the pericentral and midzonal regions, with relative sparing of the periportal areas. 47 48 Demonstration of Neutrophil Recruitment by Immunohistochemical Staining Immunohistochemical staining for neutrophils was utilized to assess neutrophil recruitment. In situ immunohistochemical staining for neutrophils was carried out using a specific mAb to mouse neutrophils. A scant number of neutrophils were identified in the livers of sham operated wild-type and P/l null mice at each time point (Figure 5A). However, after 90 minutes of ischemia and 6 hours of reperfusion a greater number of neutrophils infiltrated into the hepatic parenchyma of wild-type mice, with the majority of neutrophils concentrated in the pericentral and midzonal regions (Figure SB). Under a similar condition, P/l null mice exhibited less neutrophil infiltration, with the majority of neutrophils concentrated in the pericentral and periportal regions (Figure 5C). After 1.5 and 3 hours of reperfusion, there was minimal infiltration of neutrophils into the liver parenchyma of both groups. 49 Figure 5A. Wild-type, 6 hour sham (No ischemia and reperfusion). Figure 58. Wild-type mouse after 90 minutes of warm ischemia and 6 hours of reperfusion. Figure SC. P/l null mouse after 90 minutes of warm ischemia and 6 hours of reperfusion. 50 51 DISCUSSION The hypothesis of this study was P-selectin and lCAM-1 play an important role in neutrophil-mediated hepatic IR injury. To test the hypothesis, the roles of P-selectin and ICAM-1 in the pathogenesis of hepatic IR injury were investigated collectively using mice deficient in P-selectin and lCAM-1. Warm IR injury to the liver leads to an acute Inflammatory response that may cause significant cellular damage and organ dysfunction. It is an important clinical problem that has been demonstrated in a number of settings, including hepatic resection, liver transplantation and hemorrhagic shock with fluid resuscitation (Huguet et al., 1994; Liu et al., 1996; Lemasters and Thruman 1997; Vedder et al., 1989). Consequences of hepatic IR injury include liver failure and multiple organ dysfunction syndrome, both of which have significant rates of morbidity and mortality (Lentsch et al., 2000). By focussing on P-selectin and ICAM-1, each involved in the step-wise recruitment of neutrophils in hepatic injury, it was hoped that the mechanism of this phenomenon-could be further elucidated and provide a potential pathway to attenuate warm hepatic IR injury in a clinical arena. Adhesion of neutrophils involves a multi-step process mediated by a variety of cell surface molecules, with the selectins mediating the initial capture and rolling of leukocytes along the endothelium, while the immunoglobulin gene superfamily and integrins are involved in the subsequent firm adhesion and transmigration. The fact that each class of adhesion molecules consists of different members with similar functions raises the possibility of functional redundancy within the system. Thus, while the double knockout mice were 52 deficient in both P-selectin and ICAM-1, it is conceivable that neutrophil rolling and transmigration may have been facilitated through a different combination of adhesion molecules. Utilizing models of trauma and TNFor, Jung and Ley (1999) concluded that overlapping functions were present among the selectin family in mice lacking a combination of selectins and the elimination of E-, L- and P- selectin significantly impaired neutrophil recruitment. Furthermore, while it is known that the recruitment of neutrophils requires the interaction between the (32- integrins and lCAM-1, a recent study indicated that antibodies to VCAM-1 could also inhibit liver injury by preventing neutrophil extravasation (Essani et al., 1997). These results indicate that neutrophils may be able to utilize interactions between B1-integrin and VCAM-1, as well as between B2-integrin and ICAM-1 for transmigration. To further support the concept of overlapping functions between adhesion molecules, some investigators have challenged the paradigm of leukocyte recruitment as a cascade of events occurring in separable, sequential steps that are dependent upon different adhesion molecules. In TNFor-induced inflammation, leukocyte rolling velocities in CD18 deficient mice were significantly elevated, suggesting that BZ-integrins are involved in mediating leukocyte rolling (Jung et al., 1998). Utilizing CD18 and E-selectin double mutant mice in a model of TNFa-induced inflammation, Forlow et al (2000) demonstrated a significant increase in leukocyte rolling. These data indicate that the steps in leukocyte recruitment are not discrete, independent events, but rather the functions of 53 selectins and integrins overlap to mediate a crucial step in the leukocyte adhesion cascade. Several studies have shown that blocking P—selectin and lCAM-1 adhesion molecules with monoclonal antibodies or their ligands attenuates inflammatory tissue injury following IR (Garcia-Criado et al., 1995; Nakano et al., 1995; Marubayashi et al., 1997; Dulkanchainun et al., 1998; Singh et al., 1998; Yadav et al., 1998). Administration of an antibody to P-selectin before and after reperfusion, in models of partial or total hepatic ischemia has shown reduced hepatic damage, decreased neutrophil infiltration and improved survival (Garcia- Criado et al., 1995; Singh et al., 1998). Similarly, administration of soluble PSGL-1 at the time of hepatic inflow occlusion in a warm ischemia model in the rat has shown attenuated hepatic damage and neutrophil emigration (Dulkanchainun et al., 1998). In addition the use of soluble PSGL-1 in the University of Wisconsin cold preservation solution for organ transplantation, significantly increased the survival of rat liver isografts (Dulkanchainun et al., 1998). Further, blocking lCAM-1 receptors with monoclonal antibodies has resulted in hepatocellular protection with decreases in transaminase levels, neutrophil infiltration and oxygen radical formation (Nakano et al., 1995; Marubayashi et al., 1997). Bullard et al (1995), who first described P/l null double mutant mice, have shown that while mice with either a P-selectin or lCAM-1 mutation alone showed 60-70% reduction in acute neutrophil emigration into the peritoneum during 8. pneumoniae-induced peritonitis, double mutant mice showed a complete loss of 54 neutrophil emigration. In contrast, neutrophil migration into the alveolar spaces during 8. pneumoniae-induced acute pneumonia was normal in double mutant mice, indicating different pathways for neutrophil infiltration into various organs. Examination of circulating leukocyte counts demonstrated a significant increase in the number of neutrophils in the PH null double mutants when compared to wild-type mice of similar genetic background (Bullard et al., 1995). This could certainly be considered a confounding variable in this investigation. The exact mechanism for the increased circulating leukocyte counts in the Pll null mice compared to wild-type mice is not known (Bullard et al., 1995). A similar phenomenon has been observed in P-selectin deficient mice (Mayadas et al., 1993; Johnson et al., 1995). Mice deficient in lCAM-1 have an even higher elevation in circulating neutrophils (Sligh et al., 1993; Xu et al., 1994). In P- selectin knockout mice, the higher level of circulating neutrophils was attributable to a delay in neutrophil clearance, which suggests that P-selectin is important for normal turnover of neutrophils in the circulation (Johnson et al., 1995). To assess whether P-selectin deficient mice and wild-type mice had comparable marginated pools of neutrophils, Johnson et al. (1995) measured blood samples before and after injection of epinephrine. Their results revealed that while the ratios of response was higher in the wild-type mice relative to the P-selectin knockout mice, the absolute number of neutrophils released into the circulation was comparable. Therefore assuming a similar absolute neutrophil increase in response to stress occurs in the P/l null mice compared to wild-type mice, then the total number of circulating neutrophils available to respond to the hepatic HR 55 injury would be similar. Examination of neutrophil counts in P/l null mice compared to wild-type mice before and after stimulation with epinephrine, as well as with hepatic IR injury could provide some insight into the issue of neutrophilia. The double knockout of the P/l null mice was confirmed for both P-selectin and lCAM-1, using reverse-transcriptase polymerase chain reaction and immunohistochemical staining of the liver tissue, respectively on mice bred in our laboratory. P-selectin immunohistochemistry was not performed. The Jackson Laboratory confirmed the PH null double mutation on mice purchased directly from the company. In this investigation a model of lobar hepatic ischemia was utilized. lschemia was induced to the median and left lateral lobes, which results in damage to approximately 70-80% of the liver mass (Kawano et al., 1989; Martinez-Mier et al., 2000). Ninety minutes of partial hepatic ischemia followed by various times of reperfusion caused a significant hepatocellular injury in a time-dependent manner In both wild-type and P/I null mice. The severity of the hepatocellular damage between P/I null mice and wild-type mice was not statistically significant as judged by plasma ALT levels and liver histopathological analysis. These results suggest that the hepatocellular injury induced following reperfusion of the ischemic liver was independent of P-selectin and lCAM-1 molecules during the first six hours of post-ischemic reperfusion. Furthermore, IR injury sustained under these conditions may involve factors not directly related to neutrophil infiltration. 56 Several investigations have observed two phases of liver injury following hepatic IR (Colletti et al., 1990; Jaeschke et al., 1990). The first or earlier phase develops over the course of the first one to three hours of reperfusion. Increased levels of liver transaminases characterize this phase. It is mediated through an ROS mechanism and correlates with Kupffer cell activation. The injury that occurs during this phase is unrelated to intrahepatic neutrophils (Jaeschke et al., 1991; Jaeschke et al., 1992). The second or later phase develops after six to twenty-four hours of reperfusion. This is characterized by a further increase in hepatic transaminases. It is also associated with a massive emigration of neutrophils into the hepatic parenchyma (Jaeschke et al., 1993; Jaeschke et al., 1993; Langdale et al., 1993; Suzuki et al., 1993). These two phases are superimposed upon the damage that is initiated by the oxidative stresses that occurs during ischemia. A limitation of this project was the reperfusion time frame that was studied. Only the initial time point for neutrophil-mediated hepatic IR injury was measured, which may explain the lack of difference observed between the wild-type and P/l null mice. The project could be taken a step further by looking at longer reperfusion times. Several studies have used mice with double-gene targeted knockouts for adhesion molecules. Kamochi et al (1999), have studied lipopolysaccharide (LPS)—induced neutrophil recruitment and injury to the lung and liver. After intraperitoneal injection of LPS, overall mortality was not reduced. However, there was a significant delay in mortality in the P/l null mice compared with that in wild type mice (Kamochi et al., 1999). In addition, lung and liver edema was 57 significantly lower in LPS-treated P-selectln/lCAM-1 null mice compared with LPS-treated wild type mice. Similarly a study by Whalen et al., investigated the inflammatory response after traumatic brain injury in P-selectin/lCAM-1 null mice (Whalen et al., 2000). They observed no differences in brain neutrophil accumulation between the wild type and the null mice. However, brain edema was significantly decreased in the null mice. In our study, we did not examine liver edema formation in P/l null mice. This investigation was limited by its design in that only double mutant mice for P-selectin and lCAM-1 were studied. Applying the surgical model to mice that are deficient in either P—selectin or lCAM-1 may add significant information regarding a possible interaction between these two adhesion molecules. A study by Yadav et al (1998) investigated the role of L-selectin and lCAM-1 in hepatic I/R injury using the double mutant L-selectin/lCAM-1 null mice, as well as single mutants. They observed that in livers subjected to 90 minutes of ischemia followed by 1 hour of reperfusion, there was a significant decrease in infiltration of neutrophils between the null and the wild-type mice. However, after 6 hours of reperfusion, there was extensive infiltration of neutrophils in all areas of the hepatic tissue, and no marked differences were detected between the null and wild-type groups. Furthermore, mice subjected to 120 minutes of ischemia with 6 hours of reperfusion had serum transaminase levels that were paradoxically higher in the null mice compared with the wild-type. An investigation that utilized P-selectin deficient mice in a model of warm partial hepatic ischemia, demonstrated an important role for P-selectin in 58 neutrophil and platelet sequestration (Yadav et al., 1999). Following 90 minutes of ischemia, there was a significant decrease in serum transaminase levels, hepatocellular necrosis, as well as neutrophil infiltration and adhesion (Yadav et al., 1999). In a model of total hepatic ischemia, there was improved survival in mice subjected to both 75 and 90 minutes of ischemia (Yadav et al., 1999). In addition, platelet sequestration, as determined by in situ immunohistochemical staining, was markedly reduced in the P-selectin deficient mice (Yadav et al., 1999). In other studies using P-selectin knockout mice in a model of partial hepatic ischemia, intravital microscopy was employed to investigate leukocyte recruitment in the postischemic hepatic vasculature. An increase in rolling leukocytes was observed in wild-type mice subjected to 20 minutes of ischemia and 5 hours of reperfusion (Zibari et al., 1998). This phenomenon was not observed in control mice, P—selectin deficient mice, or wild-type mice treated with a mAb to P-selectin (Zibari et al., 1998). Furthermore, when this same group looked at the number of rolling, saltating and adherent leukocytes in terminal hepatic venules after IR, there were significant increases in wild-type mice, whereas in P-selectin deficient mice these responses were absent (Sawaya et al., 1999). Together, these investigations point toward a role for P-selectin in hepatic IR injury. Some studies have concluded that P-selectin, in contrast to many other non-hepatic tissues, is not required for sinusoidal neutrophil sequestration, transendothelial migration, and subsequent neutrophil-induced liver parenchymal injury. Since there are no Weibel-Palade bodies in sinusoidal endothelial cells of 59 normal liver (Wisse 1972; Irving et al., 1984), P-selectin expression in the liver must be under transcriptional control (Essani et al., 1998). In a model of endotoxemia, P-selectin mRNA formation was demonstrated in Kupffer cells as well as endothelial cells (Essani et al., 1995). In a model of galactosamine and Salmonella abortus equi endotoxemia, immunohistochemical analysis indicated that P-selectin was expressed on endothelial cells of the periportal and central vein areas. However P-selectin was not detected on sinusoidal endothelial cells (Essani et al., 1998). In addition, administration of an anti-P-selectin mAb neither reduced the number of accumulating neutrophils in the sinusoids, nor attenuated liver injury. This study suggests that the lack of P-selectin expression on sinusoidal cells and on hepatocytes makes a contribution of P-selectin to transmigration and adherence to parenchymal cells unlikely. The results of these investigations by Essani and Jaeschke lend support to a three-step mechanism of neutrophil-mediated parenchymal injury: 1) neutrophil sequestration in the hepatic vasculature, 2) transendothelial migration, and 3) adherence to hepatocytes which leads to the release of cytotoxic mediators (Jaeschke 1997). The first step is sequestration of activated neutrophils in the hepatic vasculature. Even though neutrophils can be found in sinusoids and marginated in post-sinusoidal venules, only cells in sinusoids appear to be relevant for injury (Chosay et al., 1997). The trapping hypothesis of neutrophil sequestration requires these cells to be mechanically trapped in the liver vasculature by swelling of the endothelial lining cells (McCuskey 1993), active sinusoidal constriction (Zhang et al., 1994), and decreased deformability of 60 neutrophil membranes (Worthen et al., 1989). The absence of P-selectin expression demonstrated by immunohistochemistry in a model of endotoxemia suggests that P-selectin is not involved in this initial inflammatory response. After the initial sequestration, neutrophils can transmigrate out of the vasculature into the liver parenchyma, adhere to the hepatocytes, and induce adherence- dependent cytotoxicity (Jaeshke 1997). A potential shortcoming of this surgical model is the prolonged duration of ischemia. While another study has utilized this model successfully, identification of a critical ischemia time that results in the obstruction of sinusoids by blood elements has not been identified. The contribution of adhesion molecules in mediating microcirculatory disturbances due to Ieukostasis has remained controversial. While some authors have proposed that the development of microcirculatory occlusion is solely dependent upon factors such as endothelial swelling, protrusion of blebs, and hemoconcentration (Menger et al., 1993), others have suggested that neutrophil plugging is an important event in the occlusion of microvasculature after reperfusion (Ferguson et al., 1993; Koo et al., 1992). This phenomenon of no-reflow has been described to occur as early as after 30 minutes of hepatic ischemia (Koo et al., 1992). Intravital microscopy could be used to determine the ischemia time point at which microvascular failure occurs in this model. Yadav et al (1998) utilized this technique to establish a critical ischemia time in a model of murine partial hepatic ischemia where 39% of the liver mass was affected. After 60 minutes of ischemia, microcirculatory failure was manifested as petechial bleeding. In livers subjected to 90 and 120 61 minutes of ischemia, there was extensive clot formation and vascular plugging (Yadav et al., 1998). It is possible that the lack of significant difference in ALT levels observed between the wild-type and P/l null mice may be secondary to poor perfusion of liver tissue with the inability to completely release enzymes in the circulation. In situations where there is marked damage to sinusoidal endothelial cells, transmigration may not be a significant factor in the pathophysiology of neutrophil-induced hepatic parenchymal injury. This may occur during severe IR injury (McKeown et al., 1988; Caldwell-Kenkel et al., 1989; Fisher et al., 1997). Under these conditions, neutrophils may have direct access to parenchymal cells without undergoing transmigration. This may explain the neutrophil infiltration seen in both wild-type and P/l null mice after 90 minutes of ischemia and 6 hours of reperfusion. Furthermore, this may also explain the differential effect of adhesion molecules in various models of inflammation. As an example, an lCAM-1 antibody was found to be highly effective in attenuating liver injury due to the prevention of neutrophil transmigration during endotoxemia (Essani et al., 1995). Yet, a similar antibody was significantly less effective in preventing liver injury during hepatic IR (Farhood et al., 1995). lschemic injury of the liver has been generally considered to result in necrosis. However, it has been recently recognized that mediators of apoptosis are activated during IR injury in both sinusoidal endothelial cells and hepatocytes. In a study of normothermic IR injury, sinusoidal endothelial cells showed evidence of apoptosis earlier than hepatocytes (Kohli et al., 2000). With 62 a longer duration of ischemia, a greater number of sinusoidal endothelial cells and hepatocytes undenNent apoptosis. This was also observed with increasing duration of reperfusion. These results suggest that apoptosis of endothelial cells followed by hepatocytes is an important mechanism of cell death after IR injury in the liver. In support of this suggestion a study Cursio et al (1999), has shown that inhibition of liver apoptosis by applying a capsase inhibitor fully protected rats from death induced by normothermic hepatic IR. In addition, a capsase inhibitor reduced sinusoidal endothelial cell apoptosis and improved survival in a model of orthotopic liver transplantation (Natori et al., 1999). An investigation that utilized an in vivo adenovirus-mediated gene transfer of the antiapoptotic gene Bel-2, demonstrated a significant decrease in hepatic IR injury based on transaminases, necrosis and apoptosis, and permanent survival (Bilbao et al., 1999). Collectively these studies further support the hypothesis that severe IR injury could damage the sinusoidal endothelial cells to such an extent that adhesion-dependent transendothelial migration may not be a crucial step in neutrophil-induced hepatic parenchymal injury. The study presented here demonstrated that there was significant neutrophil infiltration after 6 hours of reperfusion in both wild-type and P/l null mice seen on histopathology and immunohistochemistry. Qualitatively, there was slightly less hepatic neutrophil infiltration in P/l null mice compared to wild- type mice. This result could be further quantified either by formal neutrophil counts from the fixed slides or by measuring myeloperoxidase activity as described by Mullane et al (Mullane et al., 1985; Bradley et al., 1982). 63 The presented study and the published studies collectively suggest that murine regulation of adhesion pathways differs during lifelong deficiency of P- selectin and lCAM-1 compared with inhibition of the adhesion molecules by antibodies. In addition, the studies indicate that adhesion-dependent and -independent activation of neutrophil emigration exists and can be differentially regulated by the targeted tissues. In conclusion, the presented study has shown hepatocellular injury following IR in double P-selectin/ICAM-1 deficient mice. The study suggests that P-selectin and lCAM-1 adhesion molecules may not be critical factors in hepatocellular injury induced by IR during the first six hours of post-ischemic reperfusion. 64 REFERENCES 65 REFERENCES . Adams DH, Burra P, Hubscher SG, et al. Endothelial activation and circulating vascular adhesion molecules in alcoholic liver disease. Hepatology 1994; 19: 588-594. . Ames A, Wright R, Kowada M, et al. The no-reflow phenomenon. Am J Pathol 1968; 52:437-453. . Aral M, Lefer DJ, So T, et al. An anti-CD18 antibody limits infarct size and preserves left ventricular function in dogs with ischemia and 48-hour reperfusion. JAm Coll Cardiol 1996; 27: 1278-1285. . Arora AS, de Groen P, Emori Y, et al. A cascade of degradive hydrolase activity contributes to hepatocyte necrosis during anoxia. Am J Physiol 1996; 270: G238-G245. . Asa D, Raycroft L, Ma L, et al. The P-selectin glycoproteins ligand functions as a common human leukocyte ligand for P- and E-selectins. J Biol Chem 1995; 270: 11662-11670. . Atalla SL, Toledo-Pereyra LH, MacKenzie GH, et al. Influence of oxygen- derived-free radical scavengers on ischemic livers. Transplantation 1985; 40: 584-590. . Aversano T, Zhou W, Nedelman M, et al. A chimeric lgG4 monoclonal antibody directed against CD18 reduces infarct size in a primate model of myocardial ischemia and reperfusion. J Am Coll Cardiol 1995; 25: 781-788. . Batelli MG. Enzymatic conversion of rat liver xanthine oxidase from dehydrogenase (D form) to oxidase (0 form). FEBS Left 1980; 113: 47-51. . Bevilacqua MP, Pober JS, Mendrick DL, et al. Identification of an inducible endothelial-leukocyte adhesion molecule. Proc Natl Acad Sci USA 1987; 84:9238-9243. 10. Bilbao G, Contreras JL, Eckhoff DE, et al. Reduction of ischemia-reperfusion injury of the liver by an in vivo adenovirus-mediated gene transfer of the antiapoptotic Bel-2 gene. Ann Surg 1999; 230: 185-193. 11.Bilzer M and Lauterburg BH. Oxidant stress and potentiation of ischemia/reperfusion injury to the perfused rat liver by human polymorphonuclear leukocytes. J Hepatol 1994; 20: 473-477. 66 12. Bioulac-Sage P, Kuiper J, Van Berkel TJC, et al. Lymphocyte and macrophage populations in the liver. Hepatogastroenterology1996; 43: 4-14. 13. Bond JM, Herman N and Lemasters JJ. Protection by acidotic pH against anoxia/reoxygenation injury to rat neonatal cardiac myocytes. Biochem Biophys Res Commun 1991; 719: 798-803. 14. Bond JM, Chacon E, Herman B, et al. Intercelluar pH and calcium hemostasis during the pH paradox of reperfusion injury to cultured neonatal rat cardiac myocytes. Am J Physiol 1993; 263: C129-C137. 15. Bonventure JV and Cheung JC. Effects of metabolic acidosis on viability of cells exposed to anoxia. Am J Physiol 1985: 249: 0149-0159. 16. Bradley PP, Preibat DA, Christensen RD, et al. Measurement of cutaneous inflammation: estimation of neutrophil content with an enzyme marker. J Invest Dennatol 1982; 78: 206-209. 17. Braunwald E and Kloner RA. Myocardial reperfusion: a double-edged sword. J Clin Invest 1985; 76: 1713-1719. 18. Bremer C, Bradford BU, Hunt kJ, et al. Role of Kupffer cells in the pathogenesis of hepatic reperfusion injury. Am J Physiol 1994; 267: G630- 6636. 19. Bronk SF and Gores GJ. pH-dependent nonlysosomal proteolysis contributes to lethal anoxic injury of rat hepatocytes. Am J Physiol 1993; 264: G774-G751. 20. Buerke M, Weyrich AS, Zheng Z, et al. Sialyl Lewisx-containing oligosaccharide attenuates myocardial reperfusion injury in cats. J Clin Invest 1994; 93: 1140-1148. 21. Bullard DC, Kunkel EJ, Kubo H, et al. Infectious susceptibility and severe deficiency of leukocyte rolling and recruitment in E-selectin and P-selectin double-mutant mice. J Exp Med 1996; 183: 2329-2336. 22. Bullard DC, Qin L, Lorenzo I, et al. P-selectin/lCAM-1 double mutant mice: acute emigration of neutrophils into the peritoneum is completely absent but is normal into pulmonary alveoli. J Clinlnvest1995; 95: 1782-1788. 23. Caldwell-Kenkel JC, Currin RT, Tanaka Y, et al. Reperfusion injuryto endothelial cells following cold ischemia storage of rat liver. Hepatology 1989; 10: 292-299. 67 24. Caldwell-Kenkel JC, Currin RT, Tanaka Y, et al. Kupffer cell activation and endothelial cell damage after storage of rat livers. Effects of reperfusion. Hepatology 1991; 13: 83-95. 25. Camargo CA, Madden JF, Gao W, et al. Interleukin-6 protects liver against warm ischemia/reperfusion injury and promotes hepatocyte proliferation in the rodent. Hepatology 1997; 26:1513-1520. 26. Camargo CA, Madden JF, Selvan RS, et al. lL-6 protects liver against warm ischemia/reperfusion injury and promotes hepatocyte proliferation in the rodent. Hepatology 1997; 26: 1513-1520. 27. Carden DL and Granger DN. Pathophysiology of ischaemia-reperfusion injury. J Pathol 2000; 190: 255-266. 28. Carden DL, Young JA and Granger DN. Pulmonary microvascular injury after intestinal ischemia-reperfusion: role of P-selectin. J Appl Physiol 1993; 75: 2529-2534. 29. Carlos TM, Clark RS, Franicola-Higgins D, et al. Expression of endothelail adhesion molecules and recruitment of neutrophils after traumatic brain injury in rats. J Leukoc Biol 1997; 61: 279-285. 30. Chazouilleres O, Guechot J, Balladur P, et al. TNFa in liver transplantation" and resection: No evidence for a key role in ischemia-reperfusion injury. J Hepatol 1992; 16: 376-379. 31. Chen H, Chopp M, Zhang RL, et al. Anti-CD11b monoclonal antibody reduces ischemic cell damage after transient focal cerebral ischemia. Ann Neurol 1994; 35: 458-463. 32. Chien KR, Abrams J, Pfau RG, et al. Prevention by chlorpromazine of ischemic liver cell death. Am J Pathol 1977; 88: 539-553. 33. Chopp M, Zhang RL, Chen H, et al. Postischemic administration of an anti- Mac-1 antibody reduces ischemic cell damage after transient middle cerebral artery occlusion in rats. Stroke 1994; 26: 869-876. 34. Chosay JG, Essani NA, Dunn CJ, et al. Neutrophil margination and extravasation in sinusoids and venules of the liver during endotoxin-induced injury. Am J Physiol 1997; 272: G1195-G2000. 35. Clavien PA. lL-6, a key cytokine in liver regeneration. Hepatology1997; 25:1294-1296. 68 36. Clavien PA. Sinusoidal endothelial cell injury during hepatic preservation and reperfusion. Hepatology1998; 28: 281-285. 37. Clavien PA, Camargo CA, Gorczynski R, et al. Acute reactant cytokines and neutrophil adhesion after warm ischemia in cirrhotic and noncirrhotic human livers. Hepatology 1996; 23: 1456-1463. 38. Collins T, Read MA, Neish AS, et al. Transcriptional regulation of endothelial cell adhesion molecules: NF-KB and cytokine-inducible enhancers. FASEB J 1995; 9: 899-909. 39. Colletti LM, Burtch GD, Remick DG, et al. The production of tumor necrosis factor alpha and the development of pulmonary capillary injury following hepatic ischemia/reperfusion. Transplantation 1990; 49: 268-272. 40. Colletti LM, Remick DG, Burtch GD, et al. 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. 41. Connolly ES, Winfree CJ, Prestigiacomo CJ, et al. Exacerbation of cerebral injury in mice that express the P-selectin gene. Circ Res 1997; 81: 304-310. 42. Connolly ES, Winfree CJ, Springer TA, et al. Cerebral protection in homozygous null ICAM-1 mice after middle cerebral artery occlusion: role of neutrophil adhesion in the pathogenesis of stroke. J Clin Invest 1996; 97: 209-216. 43. Crockett-Torabi E, Sulenbarger B, Smith CW, et al. Activation of human neutrophils through L-selectin and Mac-1 molecules. J Immunol 1995; 154: 2291-2302. 44. Currin RT, Gores GJ, Thurman RG, et al. Protection by acidotic pH against anoxic cell killing in perfused rat liver: Evidence for a “pH paradox.” FASEB J 1991; 5: 205-210. 45. Currin RT, Caldwell-Kenkel JC, Lichtman SN, et al. Protection by Carolina rinse solution, acidotic pH and glycine against lethal reperfusion injury to sinusoidal endothelial cells of rat livers stored for tranplantation. Transplantation 1996; 62: 1549-1558. 46. Cursio R, Gugenheim J, Ricci JE, et al. A capsase inhibitor fully protects against lethal normothermic liver ischemia by inhibition of liver apoptosis. FASEB J 1999; 13: 253-261. 47. Cywes R, Packham MA, Tietze L, et al. Role of platelets in allograft preservation injury in the rat. Hepatology1993; 18: 635-647. 69 48. Dahm LJ, Scultze AE and Roth RA. Activated neutrophils injure the isolated, perfused at liver by an oxygen radical-dependent mechanism. Am J Pathol1991; 139: 1009-1020. 49. Del Zopppo GJ, Schmid-Schonbein GW, Mori E, et al. Polymorphonuclear leukocytes occlude capillaries following cerebral artery occlusion and reperfusion. Stroke 1991 , 22: 1 276-1 283. 50. Della Corte E and Stirpe F. The regulation of rat liver xanthine oxidase. Involvement of thiol groups in the conversion of the enzyme activity from the dehydrogenase (type D) into oxidase (type 0) and purification of the enzyme. Biochem J 1972; 126: 739-745. 51. Drath DB and Karnovsky ML. Superoxide production by phagocytic leukocytes. J Exp Med 1975; 141: 257-262. 52. Dulkanchainun TS, Goss JA, lmagawa DK, et al. Reduction of hepatic ischemia/reperfusion injury by a soluble P-selectin glycoproteins ligand-1. Ann Surg 1998; 227: 832-840. 53. Entman ML, Michael LH, Rossen RD, et al. Inflammation in the course of early myocardial ischemia. FASEB J 1991; 5: 2529-2537. 54. Entman ML, Youker K, Shoji T, et al. Neutrophil induced oxidative injury of cardiac myocytes: a compartmented system requiring CD11b/CD18-lCAM-1 adherence. J Clin Invest 1992; 90: 1335-1345. 55. Eppihimer MJ, Wolitzky B, Anderson DC, et al. Heterogeneity of expression of E- and P-selectins in vivo. Circ Res 1996; 79:560-569. 56. Essani NA, Bajt ML, Vonderfecht SL, et al. Transcriptional activation of vascular cell adhesion molecule-1 (VCAM-1) gene in vivo and its role in the pathophysiology of neutrophil-induced liver injury in murine endotoxin shock. Immunolgy 1997; 158: 5941 -5948. 57. Essani, NA, Fisher MA, Farhood A, et al. Cytokine-induced hepatic intercellular adhesion molecule-1 (lCAM-1) mRNA expression and its role in the pathophysiology of murine endotoxin shock and acute liver failure. Hepatology 1995; 21 : 1632-1639. 58. Essani, NA, Fisher MA and Jaeschke H. Inhibition of NF-xB activation by dimethyl sulfoxide by correlates with suppression of TNF-a formation, reduced lCAM-1 gene transcription and protection against endotoxin-induced liver injury. Shock 1997; 7: 90-96. 70 59. Essani NA, Fisher MA, Manning AM, et al. Differential induction of mRNA for lCAM-1 and selctins in hepatocytes, Kupffer cells and endothelial cells during endotoxemia. Biochem Biophys Res Commun 1995; 211: 74-82. 60. Essani NA, Fisher MA, Manning AM, et al. Endotoxin-induced activation of nuclear transcription factor KB in hepatocytes, Kupffer cells and endothelial cells in vivo. J Immunol 1996; 156: 2956-2963. 61. Essani NA, Fisher MA, Simmons CA, et al. Increased P-selectin gene expression in the liver vasculature and its role in the pathophysiology of neutrophil-induced liver injury in murine endotoxin shock. J Leukocyte Biol 1998; 63: 288-296. 62. Fabry Z, Waldschmidt MM, Hendrick D, et al. Adhesion molecules on murine brain microvascular endothelial cells: expression and regulation of lCAM-1 and Lpg 55. J Neuroimmunol 1992; 36: 1-11. 63. Faist E, Baue AE, Dittmer H. et al. Multiple organ failure in polytrauma patients. J Trauma 1983; 23: 775-787. 64. Farhood A, McGuire GM, Manning AM, et al. lntercellular adhesion molecule-1 (lCAM-1) expression and its role in neutrophil-induced ischemia- reperfusion injury in rat liver. J Leukoc Biol 1995; 57: 368-374. 65. Feliciano DV, Mattox KL, Jordan GL, et al. Management of 1000 consecutive cases of hepatic trauma. Ann Surg 1986; 204: 438-445. 66. Ferguson D, McDonagh P, Biewer J, et al. Spatial relationship between leukocyte accumulation and microvascular injury during reperfusion following hepatic ischemia. Int J Micorcirc Clin Exp 1993; 12: 45-60. 67. Fisher MA, Eversole RR, Beuving LJ, et al. Sinusoidal endothelial cell and parenchymal injury during endotoxemia and hepatic ischemia-reperfusion: Protection by the 21-aminosteroid tirilazad mesylate. Int Hepatol Commun 1997; 6: 121-129. 68. Forlow SB, White EJ, Barlow SC, et al. Severe inflammatory defect and reduced viability in CD18 and E-selectin double mutant mice. J Clin Invest 2000; 106: 1457-1466. 69. Frenette PS, Mayadas TN, Rayburn H, et al. Susceptibility to infection and altered hematopoiesis in mice deficient in both P- and E-selectins. Cell 1996; 84: 563-574. 70. Frenette PS and Wagner DD. Adhesion molecules 1. N Engl J Med 1996; 334: 1526-1529. 71 71. Frenette PS and Wagner DD. Adhesion molecules 2: blood vessels and blood cells. N Engl J Med 1996; 335: 43-45. 72. Frohman EM, Frohman TC, Dustin ML, et al. The induction of intercellular adhesion molecule-1 (lCAM-1) expression on fetal astrocytes by interferon, tumor necrosis factor a, lymphotoxin and interleukin-1: relevance to intracerebral antigen presentation. J Neuroimmunol 1995; 23: 117-124. 73. Gao W, Bentley RC, Madden JF, et al. Apoptosis of sinusoidal endothelial cells is a critical mechanism of preservation injury in rat liver transplantation. Hepatology 1998; 27: 1652-1660. 74. Garcia JH, Liu KF, Yoshida Y, et al. Influx of leukocytes and platelets in an evolving brain infarct (Wishtar rat). Am J Pathol 1994; 144: 188-199. 75. Garcia-Criado, FJ, Toledo-Pereyra LH, Lopez-Neblina F, et al. Roll of P- selectin in total hepatic ischemia and reperfusion. J Am Col Surg 1995; 181: 327-334. 76. Gill EA, Kong Y, and Honlvitz LD. An oligosaccharide sialyl-Lewis(x) analogue does not reduce myocardial infarct size after ischemia and reperfusion in dogs. Circulation 1996; 94: 542-546. 77. Gores GJ, Fleishman KE, Dawson TE, et al. Extracellular acidosis delays onset of cell death in ATP depleted hepatocytes. Am J Physiol 1988; 255: C315-C322. 78. Gores GJ, Nieminen A-L, Wray BE, et al. lntercellular pH during chemical hypoxia in cultured hepatocytes. J Clin Invest 1989; 255: 386-396. 79. Granger DN and Kubes P. The microcirculation and inflammation: modulation of leukocyte-endothelial cell adhesion. J Leukoc Biol 1994; 55: 662-675. 80. Hallenbeck JM, Dutka AJ, Tanishima T, et al. Polymorphonuclear leukocyte accumulation in brain regions with low blood flow during the early postischemic period. Stroke 1985; 17: 246-253. 81. Harper IS, Bond JM, Chacon E, et al. Inhibition of Na‘IH" exchange preserves viability, restore mechanical function, and prevents the pH paradox in reperfusion injury to rat neonatal myocytes. Basic Res Cardiol1993; 88: 430-442. 72 82. Harrison DC, Lemasters JJ and Herman B. A pH-dependent phospholipase A2 contributes to loss of plasma membrane integrity during chemical hypoxia in rat hepatocytes. Biochem Biophys Res Commun 1991; 174: 654-659. 83. Harrison-Schostak DC, Lemasters JJ, Edgell CJ, et al. Role of ICE-like proteases in endothelial cell hypoxic and reperfusion injury. Biochem Biophys Res Commun 1997; 231: 844-847. 84. Hartman JC, Anderson DC, Wiltse AL, et al. Protection of ischemic/reperfused canine myocardium by CL18/6, a monoclonal antibody to adhesion molecule ICAM-1. Cardiovasc Res 1995; 30: 47-54. 85. Hellerbrand c, Wang SC, Tsukamoto H, et al. Expression of intercellular adhesion molecule-1 by activated hepatic stellate cells. Hepatology1996; 24: 670-676. 86. Holloway CM, Harvey PR, Strasberg SM, et al. Viability of sinusoidal lining cells in cold-preserved liver allografts. Transplantation 1990; 49: 225-229. 87. Horie Y, Wolf R, Russell J, et al. Roll of Kupffer cells in gut ischemia/reperfusion-induced hepatic microvascular dysfunction in mice. Hepatology 1997; 26: 1499-1505. 88. Hsu-Lin SC, Berrnan CL, Furie BC, et al. A platelet membrane protein expressed during platelet activation and sevretion. Studies using a monoclonal antibody specific for thrombin-activated platelets. J Biol Chem 1984: 258: 9121-9126. 89. Huguet C, Gavelli A, and Bone S. Hepatic resection with ischemia of the liver exceeding one hour. J Am Coll Surg 1994; 178: 454-458. 90. Hynes R0 and Wagner DD. Genetic manipulation of vascular adhesion molecules in mice. J Clin lnvest1996; 98: 2193-2195. 91. Irving MG, Roll FJ, Huang S, et al. Characterization and culture of sinusoidal endothelium from normal rat liver: Lipoprotein uptake and collagen phenotype. Gastroenterol1984; 87: 1233-1247. 92. Jaeschke H. Cellular adhesion molecules: regulation and functional significance in the pathogenesis of liver diseases. Am J Physiol 1997; 273: G602-GG11. 93. Jaeschke H, Bautista AP, Spolarics 2, et al. Superoxide generation by Kupffer cells and priming of neutrophils during reperfusion after hepatic ischemia. Free Radic Res Commun 1991; 15:277-285. 73 94. Jaeschke H, Bautista AP, Spolarics 2, et al. Superoxide generation by neutrophils and Kupffer cells during in vivo reperfusion after hepatic ischemia in rats. J Leukoc Biol 1992; 52: 37-382. 95. Jaeschke H, Essani NA, Fisher MA, et al. Release of soluble intercellular adhesion molecule-1 (leAM-1) into bile and blood in murine endotoxin shock. Hepatology1996; 23: 530-536. 96. Jaeschke H and 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. 97. Jaeschke H, Farhood A, Bautistia AP, et al. Functional inactivation of neutrophils with Mac-1 (CD11b/CD18) monoclonal antibody protects against ischemia-reperfusion injury in rat liver. Hepatology1993; 17: 915-923. 98. Jaeschke H, Farhood A, Smith CW. Cell adhesion and migration lll. Leukocyte adhesion and transmigration in the liver vasculature. Am J Physiol 1997; 273: G1169-G1173. 99. Jaeschke H, Farhood A, Smith CW. Neutrophils contribute to ischemia/reperfusion injury in rat liver in vivo. FASEB 1990; 423355-3359. 100. Jaeschke H and Smith CW. Mechanisms of neutrophil-induced parenchymal cell injury. J Leukoc Biol 1997; 61: 647-653. 101. Johnson RC, Mayadas TN, Frenette PS, et al. Blood cell dynamics in P- selectin-deficient mice. Blood 1995; 86: 1106-1114. 102. Jung U and Ley K. Mice lacking two or all three selectins demonstrate overlapping and distinct functions for each selectin. J Immunol 1999; 162: 6755-6762. 103. Jung U, Norman KE, Scharfetter-Kochanek K, et al. Transit time of leukocytes rolling through venules controls cytokine-induced inflammatory cell recruitment in vivo. J Clin Invest 1998; 102: 1526-1533. 104. Kaplan SH, Yang H, Gilliam DE, et al. Hypercapnic acidosis and dimethyl amiloride reduce perfusion induced cell death in ischemic rabbit ventricular myocardium. Cardiovasc Res 1995; 29: 231-238. 105. Kawano K, Kim Yl, Kaketani K, et al. The beneficial effect of cyclosporine on liver ischemia in rats. Transplantation 1989; 48: 759-764. 74 106. Keelan ETM, License ST, Peters AM, et al. Characterization of E- selectin expression in vivo with use of radiolabeled monoclonal antibody. Am J Physiol 1994; 266: H279-H290. 107. Keizer GD, Visser W, Vliem M, et al. A monoclonal antibody (NK1-L16) directed against a unique epitope on the a-chain of human lymphocyte- associated antigen-1 induces homotypic cell-cell interactions. J Immunol 1988; 140: 1393-1400. 108. Keller GA, West ME, Cerra FB, etal. Macrophage-mediated modulation of hepatic function in multiple-system failure. J Surg Res 1985; 39: 555-563. 109. Kelly KJ, Williams WW, Colvin RB, et al. lntercellular adhesion molecule-1 deficient mice are protected against ischemic renal injury. J Clin Invest 1996; 97: 1056-1063. 110. Kishimoto TK and Anderson DC. The role of integrins in inflammation. In: Inflammation. Basic Principles and Clinical Correlates, edited by Jl Gallin, IM Goldstein and R Snyderman. New York: Raven, 1992, p. 353-406. 111. Kishimoto TK, Jutila MA, Berg EL, et al. Neutrophil Mac-1and MEL-14 adhesion proteins inversely regulated by chemotactic factors. Science 1989; 245: 1238-1241. 112. Kishimoto TK and Rothlein R. lntegrins, ICAMs and selectins: role and regulation of adhesion molecules in neutrophil recruitment to inflammatory sites. Adv Phannacol 1994; 25: 117-169. 113. Kobayashi H, Nonami T, Kurokawa T, et al. Role of endogenous nitric oxide in ischemia-reperfusion in rat liver. J Surg Res 1995; 27: 163-169. 114. Kochanek PM and Hallenbeck JM. Polymorphonuclear leukocytes and monocytes/macrophages in the pathogenesis of cerebral ischemia and stroke. Stroke 1992; 23: 1367-1379. 115. Kohli V, Selzner M, Madden JF et al. Endothelial cell and hepatocyte deaths occur by apoptosis after ischemia-reperfusion injury in the rat liver. Transplantation 1999; 67: 1099-1105. 116. Koo A, Komatsu H, Tao G, et al. Contribution of no-reflow phenomenon to hepatic injury after ischemia-reperfusion: Evidence for a role for superoxide anion. Hepatology1992; 15:507-514. 117. Kornberg LJ, Earp HS, Turner CE, et al. Signal transduction of integrins: increased tyrosine phosphoryation caused by clustering of [31 integrins. Proc Natl Acad Sci USA 1991; 88: 8392-8396. 75 118. Kuchan MJ, Jo H and Frangos JA. Role of G proteins in shear stress- mediated nitric-oxide production by endothelial cells. Am J Physiol 1994; 267: C753-C758. 119. Kukielka GL, Hawkins HK, Michael L, et al. Regulation of intercellular adhesion molecule-1 (lCAM-1) in ischemic and reperfused canine myocardium. J Clin Invest 1993; 92: 1504-1516. 120. Kvale D and Brandzaeg P. Immune modulation of adhesion molecules lCAM-1 (CD54) and LFA-3 (CD58) in human hepatic cell lines. J Hepatol 1993; 17: 347-352. 121. Langdale LA, Flaherty LC, Liggit HC, et al. Neutrophils contribute to hepatic ischemia-reperfusion injury by a CD18-independent mechanism. J Leukoc Biol 1993; 53: 511-517. 122. Lefer AM, Weyrich AS and Buerke M. Role of selectins, a new family of adhesion molecules, in ischemia-reperfusion injury. Cardiovasc Res 1994; 28: 289-294. 123. Lefer DJ, Flynn DM, Anderson DC, et al. Combined inhibition of P- selectin and lCAM-1 reduces myocardial injury following ischemia and reperfusion. Am J Physiol 1996; 271: H2421-H2429. 124. Lefer DJ, Flynn DM, and Buda AJ. Effects of a monoclonal antibody directed against P-selectin after myocardial ischemia and reperfusion. Am J Physiol 1996; 270: H88-H98. 125. Lefer DJ, Flynn DM, Phillips ML, et al. A novel sialyl Lewis" analog attenuates neutrophil accumulation and myocardial necrosis after ischemia and reperfusion. Circulation 1994; 90: 2390-2401. 126. Lefer DJ, Shandelya SM, Serrano, Jr CV, et al. Cardioprotective actions of a monoclonal antibody against CD-18 in myocardial ischemia-reperfusion injury. Circulation 1993; 88: 1779-1787. 127. Lemasters JJ and Thurman RG. Reperfusion injury after liver preservation for transplantation. Annu Rev Phannacol Toxicol 1997; 37: 327- 338. 128. Lentsch AB, Kato A, Yoshidome H, et al. Inflammatory mechanisms and therapeutic strategies for warm hepatic ischemia/reperfusion injury. Hepatology 2000; 32: 169-173. 76 129. Lentsch AB, Yoshidome H, Cheadle WG, et al. Chemokine involvement in hepatic ischemia/reperfusion injury in mice: Roles for macrophage inflammatory protein-2 and KC. Hepatology1998; 27: 1172-1177. 130. Lichtman SN and Lemasters JJ. Role of cytokines and cytokine- producing cells in reperfusion injury to the liver. Sem Liv Dis 1999; 19: 171- 187. 131. Lichtman SC, Wang J, Schwab JH, et al. Comparison of peptidoglycan- polysaccharide and LPS stimulation of Kupffer cells to produce TNFa and IL- 1. Hepatology1994; 19: 1013-1022. 132. Lindert KA, Caldwell-Jenkel JC, Nukina S, et al. Activation of Kupffer cells on reperfusion following hypoxia: Particle phagocytosis in a low-flow, reflow model. Am J Physiol 1992; 262: G345-G350. 133. Liu DL, Jeppsson B, Hakansson CH, et al. Multiple-system organ failure damage resulting from prolonged hepatic inflow interruption. Arch Surg 1996; 131: 442-447. 134. Liu P, Yin K, Nagle R, et al. Inhibition of nitric oxide synthase attenuates peroxynitrite generation, but augments neutrophil accumulation in hepatic ischemia-reperfusion in rats. J Phannacol Exp Ther1998; 284: 1139-1146. 135. McCord JM. Oxygen-derived free radical in postischemic tissue injury. New Engl J Med 1985; 312: 159-163. 136. McCuskey RS. Hepatic microvasculature responses to endotoxemia and sepsis. ProgAppIMicrocirc1993; 19: 76-84. 137. McEver RP, Beckstead JH, Moore KL, et al. GMP-140, a platelet alpha granule membrane protein, is also synthesized by vascular endothelial cells and is localized in Weibel-Palade bodies. J Clin Invest 1989; 84: 92-99. 138. McEver RP, Moore KL and Cummings RD. Leukocyte trafficking mediated by selectin-carbohydrate interactions. J Biol Chem 1995; 270: 11025-11028. 139. McKeown CMB, Edwards V, Phillips MJ, et al. Sinusoidal lining cell damage: the critical injury in cold preservation of allografts in the rat. Transplantation 1988; 46: 178-191. 140. Ma X-L, Lefer DJ, Lefer AM, et al. Coronary endothelial and cardiac protective effects of a monoclonal antibody to intercellular adhesion molecule- 1 in myocardial ischemia and reperfusion. Circulation 1992; 86: 937-946. 77 141. Ma X-L, Tsao PS, and Lefer AM. Antibody to 00-18 exerts endothelial and cardioprotective effects in myocardial ischemia and reperfusion. J Clin Invest 1991 ; 88: 1237-1243. 142. Marabayashi S, Dohi K, Ochi K, et al. Role of free-radicals in ischemic rat liver cell injury: Prevention of damage by alpha-tocopherol administration. Surgery 1986; 99: 184-192. 143. Marubayshi S, Oshiro Y, Maeda T, et al. Protective effects of monoclonal antibodies to adhesion molecules on rat liver ischemia-reperfusion injury. Surgery 1997; 122: 45-52. 144. Marth JD. Recent advances in genome mutagenesis by site-directed recombination. J Clin Invest 1996; 97: 1999-2002. 145. Martinez-Mier G, Toledo-Pereyra LH, McDuffie JE, et al. P-selectin and chemokine response after liver ischemia and reperfusion. J Am Coll Surg 2000; 191: 395-402. 146. Mayadas TN, Johnson RC, Rayburn H, et al. Leukocyte rolling and extravasation are severely compromised in P-selectin-deficient mice. Cell 1993; 74: 541-554. 147. Menger MD, Vollmar B, Glasz J, et al. Microcirculatory manifestations of hepatic ischemia/reperfusion injury. In: Liver Microcirculation and Hepatobiliary Function, edited by K Messmer and MD Menger. Basel: Karger, 1993, p 106-124. 148. Metzger J, Dore SP and Lauterberg BH. Oxidant stress during reperfusion of the ischemic liver: no evidence for a role of xanthine oxidase. Hepatology1988; 8: 580-584. 149. Mickelson JK, Kukielka G, Bravenec JS, et al. Differential expression and release of CD54 induced by cytokines. Hepatology1995; 22: 866-875. 150. Miller LJ, Scharting R and Springer TA. Regulated expression of the Mac- 1, LFA-1, p150.95 glycoprotein family during leukocyte differentiation. J Immunol 1986; 137: 2891 -2900. 151. Mochida S, Ohno A, Arai M, et al. Role of adhesion molecules in the development of massive hepatic necrosis in rats. Hepatology1996; 23: 320- 328. 152. Mori E, del Zoppo GJ, Chambers JD, et al. Inhibition of polymorphonuclear leukocyte adherence suppresses no—reflow after focal cerebral ischemia in baboons. Stroke 1992; 23: 712-718. 78 153. Morita M, Watanabe Y and Akaike T. Inflammatory cytokines upregulate intercellular adhesion molecule-1 expression on primary cultured mouse hepatocytes and T-lymphocyte adhesion. Hepatology1994; 19: 426-431. 154. Mullane KM, Kraemer R and Smith B. Myeloperoxidase activity as a quantitative assessment of neutrophil infiltration into ischemic myocardium. J Pharrnacol Methods 1985; 14: 157-167. 155. Muraoka K-I, Shimizu K, Sun X, et al. Hypoxia, but not reoxygenation induces IL-6 gene expression through NFKB activation. Transplantation 1997; 64: 466-470. 156. Muller MJ, Vollmar B, Friedl HP, et al. Xanthine oxidase and superoxide radicals in portal triad crossclamping-induced microvascular reperfusion injury of the liver. Free Radic Biol Med 1996; 21: 189-197. 157. Naka Y, Toda K, Kayano K, et al. Failure to express the P-selectin gene or P-selectin blockade confers early pulmonary protection after lung ischemia or transplantation. Proc Natl Acad Sci USA 1997; 94: 757-61. 158. Natori S, Selzner M, Valentino KL, et al. Apoptosis of sinusoidal endothelial cells occurs during liver preservation injury by a capsase- dependent mechanism. Transplantation 1999; 68: 89-96. 159. Nishimura Y, Romer LH and Lemasters JJ. Mitochondrial dysfunction and cytoskeletal dysruption during chemical hypoxia to cultured hepatic sinusoidal endothelial cells: The pH paradox and cytoprotection by glucose, acidotic pH and glycine. Hepatology1998; 27: 1039-1049. 160. Nishimura Y, Takei Y, Kawano S, et al. The F(ab')2 fragment of anti- lCAM-1 monoclonal antibody attenuates liver injury after orthotopic liver transplantation. Transplantation 1996; 61 : 99-104. 161. Ohira H, Ueno T, Shakdo S, et al. Cultured rat heaptic sinusoidal endothelial cells express intercellUlar adhesion molecule-1 (lCAM-1) by tumor necrosis factor-a or interleukin-1a stimulation. J Hepatol 1994; 20: 729-734. 162. Ohira H, Ueno T, Torimura T, et al. Leukocyte adhesion molecules in the liver and plasma cytokine levels in endotoxin-induced rat liver injury. Scand J Gastroenterol 1995; 30: 1027-1035. 163. Ohlinger W, Dinges hp, Zatloukal K, etal. Immunohistochemical detection of tumor necrosis factor-a, other cytokines and adhesion molecules in human livers with alcoholic hepatitis. Virchows Arch 1993; 423: 169-176. 79 164. Pachter HL and Spencer FC. Recent concepts in the management of hepatic trauma. Ann Surg 1979; 190: 423-429. 165. Palazzo AJ, Jones SP, Anderson DC, et al. Coronary endothelial P- selectin in pathogenesis of myocardial ischemia-reperfusion injury. Am J Physiol 1998; 275: H1865—H1872. 166. Palazzo AJ, Jones SP, Girod WG, et al. Myocardial ischemia-reperfusion injury in CD18- and lCAM-1-deficient mice. Am J Physiol 1998; 275: H2300- H2307. 167. Pannen BH, Al-Adili F, Bauer M, et al. Role of endothelins and nitric oxide in hepatic reperfusion injury in the rat. Hepatology1998; 27: 755-764. 168. Qian T, Nienimen A-L, Herman B, et al. The role of pH, Na+ and the MPT in reperfusion injury to rat heaptocytes: Protection by cyclosporin A and glycine. Am J Physiol 1997; 273: C1783-C1792. 169. Redl H, Dinges HP, Burrman WA, et al. Expression of endothelial leukocyte adhesion molecule-1 in septic but not traumatic/hypovolemic shock in the baboon. Am J Pathol 1991; 139: 461-466. 170. Reitman S and Frankel S. A colorimetric method for the determination of serum glutamic oxalacetic and glutamic pyruvic transaminases. Am J Clin Pathol1957; 28: 56-63. 171. Rodriguez AA, LaMorte WW, Hanrahan LM, et al. Liver viability after ischemia-reperfusion. Arch Surg 1991; 126: 767-772. 172. Rossant J and Naggy A. Genome engineering: the new mouse genetics. Nat Med 1995; 1: 592-594. 173. Ruoslahti E. lntegrins. J Clin Invest1991; 87: 1-5. 174. Sawaya, Jr DE, Zibari GB, Minardi A, et al. P-selectin contributes to the initial recruitment of rolling and adherent leukocytes in hepatic venules after ischemia/reperfusion. Shock 1999; 12: 227-232. 175. Schachner M. The analysis of neural recognition molecules: benefits and vicissitudes of functional knock-outs using antibodies and gene ablation. Curr Opin Cell Biol 1993; 5: 786-790. 176. Scharffetter-Kochanek K, Lu H, Norman K, et al. Spontaneous skin ulceration and defective T cell function in CD18 null mice. J Exp Med 1998; 188: 119-131. 80 177. Schuch U, Lohse MJ and Schachner M. Neural adhesion molecules influence second messenger systems. Neuron 1989; 3: 13-20. 178. Scoazec JY, Durand F, Degott C, et al. Expression of cytokine-dependent adhesion molecules in postperfusion biopsy specimens of liver allograft. Gastroenterology 1994: 107: 1094-1 102. 179. Scoazec JY and Feldmann G. In situ immunophenotyping study of endothelial cells of the human hepatic sinusoid: results and functional implications. Hepatology 1991; 14: 789-797. 180. Seekamp A, Till GO, Mulligan MS, et al. Role of selectins in local and remote tissue injury following ischemia and reperfusion. Am J Pathol 1994; 144: 592-598. 181. Shanley TP, Schmal H, Warner RL, etal. Requirement of C-X-C chemokines (MlP-2 and CINC) in lgG immune complex-induced lung injury. J Immunol 1997; 158: 3439-3448. 182. Shiatori Y, Kiriyama H, Fukishi Y, et al. Modulation of ischemia- reperfusion-induced hepatic injury by Kupffer cells. Dig Dis Sci 1994; 39: 1265-1272. 183. Shimizu Y, Van Seventer GA, Horgan KJ, et al. Regulated expression and binding of three VLA (B1) integrin receptors on T cells. Nature 1990; 345: 250-253. 184. Shirasugi N, Wakabayashi G, Shimizu M, et al. Upregulation of oxygen- derived free radicals by lL-1 in hepatic ischemia/reperfusion injury. Transplantation 1 997; 64: 1398-1403. 185. Shito M, Wakabayashi G,Ueda M, et al. lL-1 receptor blockade reduces TNFa production, tissue injury and mortality after hepatic ischemia- reperfusion injury in the rat. Transplantation 1997; 63: 143-148. 186. Simpson PJ, Todd RFI, Fantone JC, et al. Reduction of experimental canine myocardial reperfusion injury by a monoclonal antibody (anti-M01, anti-CD11b) that inhibits leukocyte adhesion. J Clin Invest 1988; 81: 624- 629. 187. Sindram D, Porte RJ, Hoffman MR, et al. Platelets induce sinusoidal endothelial cell apoptosis upon reperfusion of the cold ischemic rat liver. Gastroenterology 2000; 118: 183-191. 188. Singh l,Zibari GB, Brown MF, et al. Hepatic ischemia-reperfusion injury causes E-selectin upregulation. Transplant Proc 1998; 30: 2321-2323. 81 189. Sligh JE, Ballantyne CM, Rich SS, et al. InflammatoryI and immune responses are impaired in mice deficienrt in intercellular adhesion molecule 1. Proc Natl Acad Sci USA 1993; 90: 8529-8533. 190. Smith CW. Transendothelial migration. In: Adhesion. Its Role in Inflammatory Disease, edited by JM Harlan and DY Liu. New York: Freeman, 1992, p. 85-115. 191. Smith CW, Marlin SD, Rothlein R, et al. Cooperative interactions of LFA-1 and MAC-1 with intercellular adhesion molecule-1 in facilitating adherence and transendothelial migration of human neutrophils in vitro. J Clin Invest 1989; 83: 2008-2017. 192. Soriano SG, Coxon A, Wang YF, et al. Mice deficient in Mac-1 (CD11b/CD18) are less susceptible to cerebral ischemia/reperfusion injury. Stroke 1999; 130: 134-139. 193. Soriano SG, Wang YF, Wagner DD, et al. P- and E-selectin-deficient mice are susceptible to cerebral ischemia-reperfusion injury. Brain Res 1999: 835: 360-364. 194. Spertini O, Cordey AS, Monai N, et al. P-selectin glycoprotein ligand-1 is a ligand for L-selectin on neutrophils, monocytes, and CD34+ hematopoietic progenitor cells. J Cell Biol 1996; 135: 523-531. 195. Springer TA. Adhesion receptors of the immune system. Nature 1990; 346: 425-434. 196. Springer TA. Traffic signals on endothelium for lymphocyte recirculation and leukocyte emigration. Annu Rev Physiol 1995; 57: 827-872. 197. Steinhoff G and Brandt M. Adhesion molecules in liver transplantation. Hepatogastroenterology 1996; 43: 11 17-1 123. 198. Strassman G, Springer TA and Adams DO. Studies on antigens associated with the activation of murine mononuclear phagocytes: kinetics of and requirements for induction of lymphocyet-associated antigen (LFA-1) in vitro. J Immunol 1985; 135: 147-151. 199. Suzuki S, Toledo-Pereyra, Rodriguez FJ, etal. Neutrophil infiltration as an important factor in liver ischemia and reperfusion injury. Modulating effects of FK506 and cyclosporine. Transplantation 1993; 55:1265—1272. 82 200. Suzuki S, Toledo-Pereyra LH and Rodgriguez FJ. Role of neutrophils during the first 24 hours after liver ischemia and reperfusion injury. Transplant Proc 1994; 26: 3695-3700. 201. Takeshima R, Kirsch JR, Koehler RC, et al. Monclonal leukocyte antibody does not decrease the injury of transient focal cerebral ischemia in cats. Stroke 1992; 23: 247-252. 202. Tedder TF, Steeber DA, Chen A, et al. The selectins: vascular adhesion molecules. FASEB J 1995; 9: 866-873. 203. Toledo-Pereyra LH. Liver transplantation reperfusion injury. Factors in its development and avenues for treatment. Klin Wochenschr1991; 69: 1099- 1104. 204. Tu L, Chen A, Delahunty MD, et al. L-selectin binds to P-selectin glycoprotein ligand-1 on leukocytes: interactions between the lectin, epidermal growth factor, and consensus repeat domains of the selectins determine ligand binding specificity. J Immunol 1996; 157: 3995-4004. 205. Van Oosten M, Van DeBilt E, De Vries HE, et al. Vascular adhesion molecule-1 and intercellular adhesion miolecule-1 expression on rat liver cells after lipopolysaccharide administration in vivo. Hepatology1995; 22: 1538- 1546. 206. Vedder NB, Fouty BW, Winn RK, et al. Role of neutrophils in generalized reperfusion injury associated with resuscitation from shock. Surgery 1989; 106: 509-516. 207. Vollmar B, Glasz J, Menger MD, et al. Leukocytes contribute to hepatic ischemia-reperfusion injury via intercellular adhesion molecule-1-mediated venular adherence. Surgery 1995; 117: 195-200. 208. Volpes R, Van den Oord JJ and Desmet VJ. Immunohistochemical study of adhesion molecules in liver inflammation. Hepatology1990; 12: 59-65. 209. Wagner GA, Ertel W, Muller P, et al. Liver ischemia and reperufion induces a systemic inflammatory response through Kupffer cell activation. Shock 1996; 5: 34-40. 210. Wakelin MW, Sanz MJ, Albelda SM, et al. An anti-platelet endothelial cell adhesion molecule-1 antibody inhibits leukocyte extravasation from mesenteric microvessels in vivo by blocking the passage through the basement membrane. J Exp Med 1996; 184:229-239. 83 211. Wang Y, Matthews WR, Guido DM, et al. Inhibition of nitric oxide synthesis aggravates reperfusion injury after hepatic ischemia and endotoxemia. Shock1995; 4: 282-288. 212. Wanner GA, Ertel W, Muller P, et al. Liver ischemia and reperfusion induces a systemic inflammatory response through Kupffer cell activation. Shock 1996; 6: 34-40. 213. Weyrich AS, Ma XY, Lefer DJ, et al. In vivo neutralization of P-selectin protects feline heart and endothelium in myocardial ischemia and reperfusion injury. J Clin lnvest1993; 91: 2620-2629. 214. Whalen MJ, Carlos TM, Clark RS, et al. The effect of brain temperature on acute inflammation after traumatic brain injury in rats. J Neurotrauma 1997; 14: 561-572. 215. Whalen MJ, Carlos, TM, Dixon CE, et al. Reduced brain edema after traumatic brain injury in mice deficient in P-selectin and intercellular adhesion molecule-1. J Leukoc Biol 2000; 67: 160-168. 216. Wisse E. An ultrastructural characterization of the endothelial cell in the rat liver sinusoid under normal and various experimental conditions, as a contribution to the distinction between endothelial cells and Kupffer cells. J Ultrastruct Res 1972; 38: 528-562. 217. Witthaut RA, Farhood C, Smith CW, et al. Complement and tumor necrosis factor-a contribute to Mac-1 (CD11b/CD18) upregulation and systemic neutrophil activation during endotoxemia in vivo. J Leukoc Biol 1994; 55: 105-111. 218. Worthem GS, Schwab B, Elson E, et al. Mechanics of stimulated neutrophils: cell stiffening induces retention in capillaries. Science 1989; 245: 183-186. 219. Xu H, Gonzalo JA, S. Pierre Y, et al. Leukocytosis and resistance to septic shock in intercellular adhesion molecule 1-deficient mice. J Exp Med 1994; 180: 95-109. 220. Yadav SS, Howell DN, Geo W, et al. L-selectin and lCAM-1 mediates reperfusion injury and neutrophil adhesion in the warm ischemic mouse liver. Am J Physiol 1998; 275: G1341-G1352. 221. Yadav SS, Howell DN, Steeber DA, et al. P-selectin mediates reperfusion injury through neutrophil and platelet sequestration in the warm ischemic mouse liver. Hepatology1999; 29: 1494-1502. 84 222. Yamazaki T, Seko Y, Tamatani T, et al. Expression of intercellular adhesion molecule-1 in rat heart with ischemia/reperfusion and limitation of infarct size by treatment with antibodies against cell adhesion molecules. Am J Pathol 1993; 143: 410-418. 223. Youker KA, Hawkins HK, Kukielka GL, et al. Molecular evidence for induction of intercellular adhesion molecule-1 in the viable border zone associated with ischemia-reperfusion injury of the dog heart. Circulation 1994; 89: 2736-2746. 224. Zager RA, Schimpf BA and Gmur DJ. Physiological pH effects on post hypoxic proximal tubular injury. Circ Res 1993; 72: 837-846. 225. Zhang JX, Pegoli W and Clemens MG. Endothelin-1 induces direct constriction of hepatic sinusoids. Am J Physiol 1994; 266: G624-G632. 226. Zhang P, Xie M, Zagorski J, et al. Hepatic neutrophil sequestration in early sepsis: enhanced expression of adhesion molecules and phagocytic activity. Shock 1994; 2: 133-140. 227. Zhang RL, Chopp M, Jiang N, et al. Anti-lCAM-1 antibody reduces ischemic cell damage of after transient middle cerebral artery occlusion in the rat. Neurology 1994; 44: 1747-1751. 228. Zibari GB, Brown MF, Burney DL, et al. Role of P-selectin in the recruitment of leukocytes in mouse liver exposed to ischemia and reperfusion. Transplant Proc 1998; 30: 2327-2330. 229. Zimmerman GA and McIntyre TM. Neutrophil adherence to human endothelium in vitro occurs by CD18 glycoprotein-dependent and -independent mechanisms. J Clin Invest 1988; 81: 531-537. 85 IIIIIIIIIIIIIIIIIIIIII