t . It? fihuuUH f , nqluhuuyaauh qgvi‘hhu 4‘1 . > V .‘ .. 3-. It .9 n - ol n~|lvvn‘n ‘AtltW‘gcu ‘ . j x .no .. .1 I l HDEADV l O u g “INC! huge? Jita‘ie f) O 0 v . k x \ UniverSlty __i This is to certify that the dissertation entitled MECHANISMS OF SULINDAC/LPS-INDUCED LIVER INJURY IN RATS: AN ANIMAL MODEL OF DRUG-INDUCED IDIOSYNCRATIC HEPATOTOXICITY presented by Wei Zou has been accepted towards fulfillment of the requirements for the ph_ [)_ degree in Microbiology and Molecular Genetics Emu-,4. 7170 Major Professor’s Sighature I-ZE-LEHQ Date MSU is an Affirmative Action/Equal Opportunity Employer 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 5/08 K IPrq/Achres/CIRC/Daleoue indd MECHANISMS OF SULINDAC/LPS-INDUCED LIVER INJURY IN RATS: AN ANIMAL MODEL OF DRUG-INDUCED IDIOSYNCRATIC HEPATOTOXICITY By Wei Zou A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Microbiology and Molecular Genetics 2010 ma:- lc‘ :s :ch :r A"! A O 1 . .~ .3 U *3 Ilpn‘ 379W 3-. , 3 war ea": 333.21; 3'2: Of 1 ABSTRACT SULINDAC/LPS-INDUCED LIVER INJURY IN RATS: AN ANIMAL MODEL OF IDIOSYNCRATIC DRUG-INDUCED LIVER INJURY By Wei Zou Idiosyncratic adverse drug response is a type of adverse reaction that occurs in a minority of patients during drug therapy. Liver is one of the major organ targets. All of the nonsteroidal anti-inflammatory drugs (NSAIDs) have been associated with hepatic idiosyncratic adverse drug reactions (IADRs) in patients, and the risk from sulindac (SLD) is reported to be 5-10 fold greater than for NSAle as a class. However, the mechanism of SLD-induoed hepatotoxicity has not been clarified because of the lack of experimental animal models. Previous studies suggest that inflammatory stress is a susceptibility factor for lADRs. The work in this dissertation supports this hypothesis. Cotreatment of rats with lipopolysaccharide (LPS), which induces modest inflammation, and SLD resulted in liver necrosis, whereas neither LPS nor SLD was hepatotoxic alone. After we developed a SLD- inflammation interaction model of idiosyncratic liver injury by treating rats with SLD and LPS, the mechanisms of SLD/LPS- induced liver injury were investigated. SLD/LPS cotreatrnent causes an increase in the production of tumor necrosis factor-a (T NF), activation of the hemostatic system and of neutrophils (PMNs) as well as oxidative stress in the liver. Neutralization of TNF, anticoagulant administration, PMN depletion or antioxidant treatment attenuated liver injury in this model. Results of neutralization or inhibition studies A0 ' Ii. in vivo and in vitro suggest roles for TNF, the hemostatic system, PMNs and oxidative stress in the pathogenesis of liver injury-induced by SLD/LPS. Moreover, these mediators are not independent players. They contribute to liver injury by interacting with each other and with the SL0 toxic metabolite, SLD sulfide. The studies in this dissertation provide an understanding of mechanisms of liver injury resulting from SLD- inflammation interaction. 727‘? 31 :A‘n q N «van; I" . U. W“ ‘l fl ACKNOWLEDGMENTS In almost five years at Michigan State University pursuing my Ph.D. degree, a lot of people have supported and assisted me along the way. Without these kindnesses, my thesis project might not have been done now, and this dissertation would not come out. There are no words sufficient to express my gratitude to them First of all, I would like to thank my mentor, Dr. Robert Roth. At the first month arriving MSU as well as United State, I attended a seminar in which I was attracted by the brilliant hypothesis that Bob proposed. I talked to Bob right after his presentation and expressed my will to join the lab. The more I worked with Bob, the better I knew it is a decision I would never regret. Bob has given me tremendous training and help. He leads me to think independently and helps me without reservation whenever there is an overwhelming obstacle for me in the project. I am also influenced by his passion and perseverance in scientific research, which sets a role model for me. Dr. Patricia Ganey is also my mentor in the lab and deserves my gratitude. Along with Bob, Patti plays a significant role in training me throughout my graduate life. She not only gives me ideas and suggestions in the project, but also helps me polish my scientific English in my manuscripts, which I will benefit from in my whole career. It is my fortune to work with so many talented and warm-hearted people in our lab. I would like to thank Shawn Deng. He is the person I closely worked with during the rotation in the lab. He was always ready to answer all kinds of iv . r llazscx i. Q. ‘ c5 OI Q“I‘Am Hwy. ‘5 8 Ag I R A N A" 31“....2a questions I came up with and never weary of showing me lab techniques. Erica Sparkenbaugh also deserves my gratitude. She helped me a lot with the in vitro experiments, including hepatocyte isolation. I also thank Kevin Beggs who worked side by side with me for months on this project. In addition to them, I would like to thank all the people in the lab who I worked with. They are Dr. Jane Maddox, Dr. Francis Tukov, Dr. Sachin Devi, Dr. Jesus Olivero—Verbel, Dr. Rohit Sinhal, Pat Shaw, Christine Dugan, Jingtao Lu, Aaron Fullerton, Kyle Poulsen, Nicole Crisp, Allen MacDonald, Theresa Eagle and Sandy Newport. Beside my colleagues in the lab, Dr. Daniel Jones in the Department of Biochemistry and Molecular Biology also contributed to this project. He spent a lot of time with me to figure out the protocol for LC/MS/MS. I would also like to thank Kazuhisa Miyakawa and Mike Scott from the Department of Pathobiology and Diagnostic Investigation, who helped me with immunohistochemistry. I also own a thank to Dr. Husam Younis from Pfizer Global Research and Development, Drug Safety R&D. It was a great experience for me to collaborate with people from industry. The last but not the least, I would like to say thanks to my parents. Although my parents are thousands of miles away from me, I am always their priority. They never stop caring about me and encouraging me whenever I am down and discouraged. Without them, I would not be a graduate student and go this far. TABLE OF CONTENTS LIST OF TABLES .................................................................................. x LIST OF FIGURES ................................................................................ xi LIST OF ABBREVIATIONS .................................................................... xvi CHAPTER 1 GENERAL INTRODUCTION ................................................................................. 1 1.1 Idiosyncratic adverse drug reactions (IADRs) .............................................. 2 1.1.1 Overview of drug-induced idiosyncratic hepatotoxicity ......................... 2 1.1.2 Conventional explanations for IADRs ................................................... 4 1.1.3 Inflammatory stress hypothesis as potential eXplanation for IADR ....... 9 1.2 Idiosyncratic adverse drug reactions (IADRs) ............................................ 14 1.2.1 Overview of the inflammatory response ............................................ 14 1.2.2 Tumor necrosis factor-a (T NF) ............................................................ 18 1.2.3 The hemostatic system and hypoxia .................................................. 22 1.2.4 Neutrophils (PMNs) ............................................................................ 23 1.2.5 Reactive oxygen species .................................................................... 25 1.3 Models of drug-inflammation interaction ................................................... 28 1.3.1 Ranitidine/LPS-induced hepatotoxicity in rats ..................................... 28 1.3.2 Diclofenac/LPS-induoed hepatotoxicity in rats .................................... 30 1.3.3 Trovafloxacin-inflammation interaction model of idiosyncratic liver injury ..................................................................................................................... 32 1.4 Sulindac-induced idiosyncratic liver injury ................................................. 35 1.5 Hypothesis and specific aims .................................................................... 39 CHAPTER 2 HEPATOTOXIC INTERACTION OF SULINDAC WITH LIPOPOLYSACCHARIDE: ROLE OF THE HEMOSTATIC SYSTEM ................ 41 2.1 Abstract ..................................................................................................... 42 2.2 Introduction ............................................................................................... 43 2.3 Materials and methods .............................................................................. 44 2.3.1 Materials ............................................................................................. 44 2.3.2 Animals ............................................................................................... 44 2.3.3 Experimental protocol ......................................................................... 44 2.3.4 Evaluation of liver injury ...................................................................... 45 2.3.5 Evaluation of serum TNFo concentrations .......................................... 47 2.3.6 Evaluation of hemostasis and fibrin deposition ................................... 47 2.3.7 Evaluation of liver hypoxia .................................................................. 48 2.3.8 Hepatocyte (HPC) isolation and hepatocytotoxicity assessment in vitro ............................................................................................................. 48 2.3.9 Statistical analysis .............................................................................. 49 2.4 Results ...................................................................................................... 50 2.4.1 Dose-response and timecourse of liver injury ..................................... 50 vi SE' 244A» 2458 rt ENSAC 32 intro: 24.2 H 24 CnAFTE" NtiIJI.“ 3;”. H CL EC CL nb m CL my Cc 9A rCrCndUEnivS u? «I. «J 4 Ev Cu 7- 03 Cu ”Aw «I «I. 1v 4 a... Ru .3. 1e 10 «J flxv nrv atv arv 63 R 4 4 4 4 4 AH. firU 1U AI‘U “TV 4. 3 1U "11‘ ”AU 3 “(V 5 Al‘V "(TV A!" 1U 1U 1U 1U 2.4.2 Histopathological findings ................................................................... 50 2.4.3 Effect of SLD/LPS cotreatrnent on serum TNF concentration ............ 57 2.4.4 Activation of the hemostatic system ................................................... 57 2.4.5 Effect of heparin on liver injury induced by SLD/LPS cotreatrnent ...... 62 2.4.6 Effect of low oxygen on hepatocytotoxicity induced by SLD sulfide in vitro ..................................................................................................... 69 2.5 Discussion ................................................................................................. 74 CHAPTER 3 SULINDAC METABOLISM AND SYNERGY WITH TNF IN A DRUG- INFLAMMATION INTERACTION MODEL OF IDIOSYNCRATIC LIVER INJURY ............................................................................................................................ 81 3.1 Abstract ..................................................................................................... 82 3.2 Introduction ............................................................................................... 83 3.3 Materials and methods .............................................................................. 84 3.3.1 Materials ............................................................................................. 84 3.3.2 Animals ............................................................................................... 84 3.3.3 Experimental protocol ......................................................................... 84 3.3.4 Evaluation of liver injury ...................................................................... 85 3.3.5 Determination of TNF concentration in serum .................................... 86 3.3.6 LC/MS/MS analysis ............................................................................ 86 3.3.7 Evaluation of cytotoxicity of SLD and its metabolites in vitro .............. 87 3.3.8 Cytotoxicity from TNF and SLD metabolites ....................................... 88 3.3.9 Statistical analysis .............................................................................. 89 3.4 Results ...................................................................................................... 90 3.4.1 Timecourse of TNF concentration in plasma ...................................... 90 3.4.2 Effect of TNF inhibition on liver injury ................................................. 90 3.4.3 Effect of LPS on SLD metabolism in rat ............................................. 94 3.4.4 Effect of etaneroept on SLD metabolism in rats ................................ 101 3.4.5 Cytotoxicity of SLD and its metabolites in HepG2 cells and rat primary hepatocytes ...................................................................................... 101 3.4.6 Effect of TNF on cytotoxicity of SLD and its metabolites in HepGZ and rat primary hepatocytes .................................................................... 101 3.5 Discussion ............................................................................................... 109 CHAPTER 4 THE CRITICAL ROLE OF TNF AND PAl-1 MEDIATED NEUTROPHIL ACTIVATION IN A SULINDAC/ LIPOPOLYSACCHARIDE- MODEL OF IDIOSYNCRATIC LIVER INJURY IN RATS .................................................... 115 4.1 Abstract ................................................................................................... 116 4.2 Introduction ............................................................................................. 1 17 4.3 Materials and methods ............................................................................ 118 4.3.1 Materials ........................................................................................... 1 18 4.3.2 Animals ............................................................................................. 118 4.3.3 Animal model and sample collection ................................................ 118 vii I.” A Q 4 E. u n» u 0.9 Add 4. 4 a..- any new 90 Ate Aid 4 4 4 “Res 441 A 44 L Ah» q I an Pt 4 4 ”U v. I. 4 4 :u Mum 4. IHL A v nyna’u VIUUH i I EN FR In. win“ we. .... m: 4 z. :. 7. uh. -nr NW 3 19 «(v 5 EU 4 . n/s .Iv .Kv rhv Exv {v pHv :v phv phv «I. 1; In 3 :v Add .40 10. any ad. NH 4 i :.U 5 4 PRU. 4.3.4 Anti-PMN serum, eglin C, PAl039 and etaneroept treatment protocol ........................................................................................................... 1 19 4.3.5 Evaluation of hepatotoxicity .............................................................. 119 4.3.6 Determination of ClNC-1, MIP-1q and PAI-1 concentrations in plasma ........................................................................................................... 120 4.3.7 Evaluation of liver PMN accumulation and activation ....................... 120 4.3.8 Assessment of fibrin deposition in liver ............................................. 121 4.3.9 Statistical analysis ............................................................................ 121 4.4 Results .................................................................................................... 123 4.4.1 Evaluation of PMN accumulation and activation in livers .................. 123 4.4.2 Time course of ClNC-1 and MIP-1q concentrations in plasma ......... 123 4.4.3 Effect of PMN depletion and PMN protease inhibition on SLD/LPS- induced liver injury ............................................................................ 126 4.4.4 Effect of TNF on PMN accumulation and activation ......................... 131 4.4.5 Role of PAH in liver injury and accumulation and activation of PMNs ........................................................................................................... 131 4.4.6 Effect of TNF on plasma PAl-1 concentration .................................. 131 4.4.7 Effect of TNF and PAl-1 on fibrin deposition ..................................... 136 4.5 Discussion ............................................................................................... 140 CHAPTER 5 OXIDATIVE STRESS IS A POTENTIAL PLAYER IN THE PATHOGENESIS OF LIVER INJURY INDUCED BY SULINDAC AND LIPOPOLYSACCHARIDE ....‘I47 5.1 Abstract ................................................................................................... 148 5.2 Introduction ............................................................................................. 149 5.3 Materials and methods ............................................................................ 150 5.3.1 Materials ........................................................................................... 150 5.3.2 Animals ............................................................................................. 150 5.3.3 Design of experiments in vivo ........................................................... 150 5.3.4 Gene expression analysis ................................................................ 151 5.3.5 Evaluation of liver injury and protein carbonyls in mitochondria ....... 152 5.3.6 Evaluation of mitochondrial membrane potential .............................. 153 5.3.7 Evaluation of reactive oxygen species in HepG2 cells ..................... 153 5.3.8 Evaluation of cytotoxicity and capase 3 activity ................................ 154 5.3.9 Statistical analyses ........................................................................... 154 5.4 Results .................................................................................................... 155 5.4.1 Gene expression changes regulated by treatment with SLD, LPS or SLD/LPS ........................................................................................... 155 5.4.2 Gene expression changes specifically regulated by SLD/LPS point to oxidative stress ................................................................................. 155 5.4.3 Oxidative stress in SLD/LPS-cotreated rats ..................................... 158 5.4.4 Effect of SLD sulfide on mitochondrial membrane potential ............. 158 5.4.5 Effect of SLD sulfide on the production of reactive oxygen species in HepG2 cells ...................................................................................... 164 5.4.6 Cytotoxicity of hydrogen peroxide and TNF to HepG2 cells ............. 164 viii r: PHIU I. Pay A ., w Rb 5.4.7 Effect of antioxidant treatment and caspase inhibition on cytotoxicity ........................................................................................................... 164 5.4.8 Effect of antioxidant treatment on caspase 3/7 activity induced by SLD sulfide and TNF cotreatrnent ............................................................. 169 5.5 Discussion ............................................................................................... 172 CHAPTER 6 SUMMARY AND CONCLUSIONS ................................................................... 177 6.1 Summary and conclusions ...................................................................... 178 6.2 Commonality and differences among liver injury models of drug- inflammation interaction .......................................................................... 183 6.3 Potential future studies ........................................................................... 187 APPENDICES ................................................................................................... 189 BIBLIOGRAPHY .............................................................................................. 201 ix LIST OF TABLES TABLE 1.1 LIST OF IDIOSYNCRATIC DRUGS TO CAUSE LIVER INJURY IN DRUG/LPS MODELS IN RODENTS .................................................................. 13 TABLE 2.1 MIDZONAL HEPATIC NECROSIS IN LIVERS OF RATS TREATED WITH SLD/LPS .................................................................................................. 58 TABLE 2.2 SERUM TNFA CONCENTRATION IN SLD/LPS-TREATED RATS .59 TABLE 3.1 EFFECT OF ETANERCEPT ON SLD METABOLISM ................... 102 TABLE 5.1 PATHWAYS ASSOCIATED WITH GENES SPECIFICALLY CHANGED BY SLD/LPS COTREATMENT ..................................................... 159 TABLE 6.1 CHARACTERISTICS AND UNDERLYING MECHANISMS OF IDIOSYNCRATIC LIVER INJURY MODELS OF DRUG-INFLAMMATION INTERACTION ................................................................................................. 184 LIST OF FIGURES IMAGES IN THIS DISSERTATION ARE PRESENTED IN COLOR FIGURE 1.1 INFLAMMATION MIGHT PRECIPITATE DRUG— INDUCED IADRS ............................................................................................................................ 11 FIGURE 1.2 SUMMARY OF THE INFLAMMATORY RESPONSE .................... 15 FIGURE 1.3 TNF SIGNAL TRANSDUCTION PATHWAY ................................. 20 FIGURE 1.4 METABOLISM OF SLD ................................................................. 36 FIGURE 2.1 EXPERIMENTAL PROTOCOL FOR ANIMAL TREATMENT ........ 46 FIGURE 2.2 SULINDAC DOSE-RESPONSE IN THE ABSENCE AND PRESENCE OF LPS .......................................................................................... 51 FIGURE 2.3 DEVELOPMENT OF HEPATOCELLULAR INJURY INDUCED BY SLD/LPS COTREATMENT ................................................................................ 52 FIGURE 2.4 MARKERS OF HEPATIC CHOLESTASIS INDUCED BY SLD/LPS COTREATMENT ................................................................................................ 54 FIGURE 2.5 LIVER HISTOPATHOLOGY IN SLD/LPS-COTREATED RATS ....56 FIGURE 2.6 ACTIVATION OF THE HEMOSTATIC SYSTEM ........................... 60 FIGURE 2.7 FIBRIN DEPOSITION IN LIVER .................................................... 63 FIGURE 2.8 HYPOXIA STAINING IN LIVER ..................................................... 64 FIGURE 2.9 EVALUATION OF FIBRIN DEPOSITION AND HYPOXIA IN LIVEF;5 FIGURE 2.10 EFFECT OF HEPARIN ON LIVER FIBRIN AND HYPOXIA ........ 67 FIGURE 2.11 EFFECT OF HEPARIN ON SLD/LPS-INDUCED LIVER INJURY 70 FIGURE 2.12 EFFECT OF HEPARIN ON HEPATIC LESIONS INDUCED BY SLD/LPS ............................................................................................................ 72 FIGURE 2.13 EFFECT OF HYPOXIA ON SLD SULFIDE- INDUCED CYTOTOXICITY ................................................................................................. 73 xi FIGURE 3.1 TIMECOURSE OF TNF CONCENTRATION IN RAT SERUM ...... 91 FIGURE 3.2 EFFECT OF TNF INHIBITION ON LIVER INJURY INDUCED BY SLD/LPS ............................................................................................................ 92 FIGURE 3.3 EFFECT OF LPS ON PLASMA CONCENTRATIONS OF SLD, SLD SULFONE AND SLD SULFIDE .......................................................................... 95 FIGURE 3.4 EFFECT OF LPS ON LIVER CONCENTRATIONS OF SLD, SLD SULFONE AND SLD SULFIDE .......................................................................... 97 FIGURE 3.5 CONCENTRATIONS OF SLD, SLD SULFONE AND SLD SULFIDE IN GI TRACT AND FECES ................................................................................ 99 FIGURE 3.6 EVALUATION OF CYTOTOXICITY INDUCED BY SLD, SLD SULFONE OR SLD SULFIDE .......................................................................... 103 FIGURE 3.7 CYTOTOXICITY INDUCED BY TNF AND SLD OR ITS METABOLITES ................................................................................................ 1 05 FIGURE 3.8 EFFECT OF TNF ON SLD SULFIDE-INDUCED INJURY TO RAT PRIMARY HEPATOCYTES ............................................................................. 108 FIGURE 4.1 EVALUATON OF PMN ACCUMULATION AND ACTIVATION IN RAT LIVERS .................................................................................................... 124 FIGURE 4.2 CONCENTRATIONS OF PMN CHEMOKINES IN RAT PLASMA .......................................................................................................................... 127 FIGURE 4.3 EFFECT OF PMN DEPLETION ON SLD/LPS-INDUCED LIVER INJURY ............................................................................................................ 129 FIGURE 4.4 EFFECT OF PMN PROTEASE INHIBITION ON SLD/LPS- INDUCED LIVER INJURY ............................................................................... 130 FIGURE 4.5 EFFECT OF TNF INHIBITION ON PMN ACCUMULATION AND ACTIVATION .................................................................................................... 1 32 FIGURE 4.6 EFFECT OF PAI-1 INHIBITION ON LIVER INJURY AND PMN ..134 FIGURE 4.7 EFFECT OF TNF INHIBITION ON PLASMA PAl-1 CONCENTRATION .......................................................................................... 137 FIGURE 4.8 EFFECT OF TNF AND PAI-1 ON LIVER FIBRIN DEPOSITION .138 FIGURE 4.9 MECHANISMS OF SLD/LPS-INDUCED LIVER INJURY ............ 146 xii name 5 rtlftPS C A F‘P P Fig.3.: a T l w'"| j. ' :KI'.I | 5r. . 9"- SC IIJVr\_ tot. .. 5 IIEIJERRN FIGURE 5.1 VENN DIAGRAM OF PROBE SETS REGULATED BY SLDNEH, VEH/LPS OR SLD/LPS .................................................................................... 156 FIGURE 5.2 EVALUATION OF PROTEIN CARBONYL CONCENTRATION IN LIVER MITOCHONDRIA ................................................................................... 161 FIGURE 5.3 EFFECT OF ANTIOXIDANTS ON LIVER INJURY ...................... 162 FIGURE 5.4 EFFECT OF SLD AND ITS METABOLITES ON MITOCHONDRIAL MEMBRANE POTENTIAL ............................................................................... 165 FIGURE 5.5 EFFECT OF SLD SULFIDE AND TNF ON PRODUCTION OF REACTIVE OXYGEN SPECIES IN HEPG2 CELLS ........................................ 166 FIGURE 5.6 CYTOTOXICITY INDUCED BY HYDROGEN PEROXIDE AND TNF .......................................................................................................................... 167 FIGURE 5.7 EFFECT OF ANTIOXIDANT ON CYTOTOXICITY INDUCED BY SLD SULFIDE AND TNF ................................................................................. 168 FIGURE 5.8 EFFECT OF ANTIOXIDANT ON CASPASE 3/7 ACTIVITY ........ 170 FIGURE 5.9 EFFECT OF PAN-CASPASE INHIBITOR ON CYTOTOXICITY INDUCED BY SLD SULFIDE AND TNF .......................................................... 171 FIGURE 6.1 PROPOSED PATHWAY IN THE PATHOGENESIS OF SLD/LPS- INDUCED LIVER INJURY ............................................................................... 182 xiii 1"“ VI \. LIST OF ABBREVIATIONS ALP - alkaline phosphatase ALT- alanine aminotransferase ANOVA— analysis of variance AST— aspartate aminotransferase CINC-1- cytokine-induced neutrophil chemoattractant-1 DMEM- Dulbecco's modified Eagle’s medium DMSO- dimethyl sulfoxide FBS- fetal bovine serum FMO- flavin containing monooxygenase GGT— y-glutamyltransferase H&E- hematoxylin and eosin HOCI- hypochlorous acid lADRs- idiosyncratic adverse drug reactions lDlLl- idiosyncratic drug-induced liver injury LDH- lactate dehydrogenase LPS- lipopolysaccharide MPO- myeloperoxidase MRM- multiple reaction monitoring MSR- methionine sulfoxide reductase NSAID- nonsteroidal anti-inflammatory drug PAI-1- plasminogen activator inhibitor-1 PMN- polymorphonuclear neutrophil xiv I j I Hi 5V- L ROS- reactive oxygen species SLD— sulindac TAT — thrombin-antithrombin TNF- tumor necrosis factor-a UPLC- ultra performance liquid chromatography XV CHAPTER 1 General introduction L ‘Cv53‘ H a “'t ‘ U 1.1 Idiosyncratic adverse drug reactions (IADRs) 1.1.1 Overview of drug-induced idiosyncratic hepatotoxicity Adverse drug reactions (ADRs) remain a major issue for affected patients as well as a huge challenge to health providers. The safety of new compounds are sometimes not well understood until a drug has been on the market for many years. As a result, serious ADRs commonly emerge after approval of a drug by the Food and Drug Administration (FDA). More than 10% of newly approved drugs from 1975 to 2000 in the United States either had to be withdrawn from the market or received a warning due to adverse reactions (Lasser et al., 2002). The liver, which plays an important role in the metabolism of drugs, is a frequent target of IADRs. There are two types of adverse drug reactions: dose-related reactions (Type A reactions) and idiosyncratic reactions (Type B reactions). Type A reactions occur during drug therapy and they are dose-dependent, and most likely occur in overdosed individuals. A typical example is acetaminophen-induced adverse reactions that are due to acetaminophen overdose (Amar and Schiff, 2007;Larson et al., 2005). lADRs differ from type A adverse drug reactions in that they are unpredictable and not apparently dose-dependent. Typically, lADRs occur only in a minority of patients who are treated with a specific drug. lADRs do not relate to the known pharmacologic effects of the drug (Kaplowitz, 2005; Uetrecht, 2006; Uetrecht, 2007). Drug-induced idiosyncratic hepatotoxicity, which might result in permanent disability or death, has great importance to human health. In addition, these hall! 5 lug“ a h ‘IIA. U€."v'\- fiflflfl ‘UUU I "AI!“A h J“ .3. ' “new. ' A LVVUI 39:" s‘. that ti.“ 37.: I. '15.. rug} reactions are a major issue for the pharmaceutical industry, because they lead to a large number of withdrawals and restrictions to the use of efficient drugs on the markets. A typical example of a drug which induced idiosyncratic hepatotoxicity is troglitazone, which contributed 10% of overall idiosyncratic drug reactions between 1998 and 2001 and was withdrawn from the market by the US. FDA in 2000 (Ostapowicz et al., 2002). The main reason for the withdrawal was that troglitazone was associated with the development of acute liver failure (Chojkier, 2005). Troglitazone is a peroxisomal proliferator-activated receptor (PPAR)-y agonist that is used to treat type 2 diabetes. Although troglitazone was beneficial at improving insulin resistance, among 1.92 million patients who took troglitazone, ninety-four cases of liver failure were reported (89 acute, 5 chronic) (Graham et al., 2003). The risk of idiosyncratic adverse drug reactions is difficult to predict, and most idiosyncratic reactions are not discovered until a drug is on the market. That is because a clear mechanistic understanding of IADRs is still absent, and lADRs are generally not reproducible in traditional animal models. For example, oral administration of troglitazone to monkeys at large doses (60- to 120—fold larger than the therapeutic dose) for 52 weeks did not increase serum liver enzymes and had little gastrointestinal, hematologic or hepatic effects (Rothwell et al., 2002). The lack of effective preclinical animal models causes mechanisms of IADRs to be poorly understood, so that appropriate action cannot be applied to prevent or treat lADRs. 1.1.2 Cc AK". $29.5 I 565787. 1.1.2 Conventional explanations for IADRs Although the mechanisms are still not fully understood, extensive studies on IADRs have been performed. Several hypotheses have been raised to explain mechanisms of IADRs, including the metabolic polymorphism hypothesis, the hapten hypothesis, the danger hypothesis, the mitochondrial abnormality hypothesis, the failure to adapt hypothesis and the multiple determinant hypothesis. The detailed hypotheses, their supporting evidence and limitations are described below. Metabolic polymorphism hypothesis The metabolic polymorphism hypothesis suggests that drug metabolites are responsible for the toxicity of IADRs. Drugs are metabolized into electrophiles or free radicals, which can covalently bind to proteins and/or unsaturated fatty acids, or induce lipid peroxidation (Kaplowitz et al., 1986). As a result, cell functions can be impaired, and cytotoxicity can be causediby inducing a cell death signaling pathway, causing impaired calcium homeostasis or decreased energy generation. Cytochrome P450 is a superfamily of enzymes that transform drugs into their reactive metabolites. Overexpression of P450 in a fraction of patients can lead to excessive reactive metabolite formation and accumulation in the liver that can result in hepatotoxicity. Therefore, polymorphism of P450 that is responsible for drug-induced hepatotoxicity is present in a specific fraction of patients. O F ”-3.3. .|" a; 2: :c :e' R .‘l-‘I' I4» iii-3'53 66-30 T Take troglitazone (TGZ) as an example. TGZ is metabolized into reactive metabolites by CYP3A4, a member of the cytochrome P450 superfamily (He at al., 2004). CYP3A4 mediates oxidative cleavage of the thiazolidinedione ring, generating a highly electrophilic metabolite, TGZ quinine, which covalently binds to cellular macromolecules (Kassahun et al., 2001). This is supported by the evidence that administrating TGZ to HepGZ cells transfected with CYP3A4 led to increased cytotoxicity (Vignati et al., 2005). However, another study suggests that TGZ quinone is less toxic to hepatocytes and HepG2 cells than TGZ itself (Tettey et al., 2001). Moreover, the role of CYP3A4 in the TGZ-induced toxicity has not been tested in vivo. Although many epidemiological studies have been performed attempting to link susceptibility to drug-induced toxicity with genes involved in drug metabolism (Kumashiro et al., 2003; Daly et al., 2007), there is no direct evidence proving that genetic/metabolic polymorphism contributes to idioscyncratic drug toxicity in vivo. Hapten hypothesis The hapten hypothesis is another prevalent theory of IADRs. It suggests that prodrugs or more likely their reactive metabolites form drug-protein adducts through covalent binding (Macher and Chase, 1969; Uetrecht, 2006). The drug- protein adducts are recognized as non-self antigens by the immune system and are taken up by antigen presenting cells. The processed adduct peptides are presented to helper T cells. Thus, an active immune response can be elicited rot-CIT l ”3"." a , yvu Tr A't‘taz tic-3“” vhv M2” '5 (I) (h {“H' I " ' ..wu“ 5r — w-nl c 5“" 3”“); MW! " 5‘ ‘IV'I. ESQ Thfi';f’. ‘A “Var-1' IVER S‘s-r93." which causes the production of specific antibodies to adducts. The production of antibodies may target self proteins and lead to the destruction of host tissues. This theory is supported by case reports in patients with liver injury. Antibodies have been detected after exposure to drugs causing idiosyncratic reactions, including halothane, diclofenac and trogalitazone (Sallie et al., 1991; Maniratanachote et al., 2005; Nguyen et al., 2008). However, evidence against this theory is also accumulating. Autoantibodies are present in only a portion of patients having drug-induced idiosyncratic hepatotoxicity. Moreover, IADRs are induced in some patients after the first exposure to drug (Clay et al., 2006), whereas a second exposure is required in this theory of adaptive immune response. Several animal models of drug-induced autoimmunity have been developed, including penicillamine-induced autoimmune syndrome and nevirapine—induced skin rash in rats (T ournade et al., 1990; Shenton et al., 2003). Although antibodies against penicillin-modified proteins are present in rats treated with penicillin, liver injury is not induced in the animal model (Shenton et al., 2004). In the nevirapine—induced skin rash model, liver injury is not induced either. Therefore, the results in animal models and human patients suggest that the autoantibodies are not necessarily pathogenic, and additional evidence supporting this theory is needed. Danger hypothesis F‘ q ‘ 3‘35 c I““-A‘~‘ “it V v i3"ci 0. ‘y‘ u The immune response induced by a foreign antigen is weak in the absence of adjuvant, which is a possible explanation of why the immune response induced by a drug itself is insufficient to cause liver injury (Uetrecht, 2006). The danger hypothesis was proposed as an alternative (Matzinger, 1994; Uetrecht, 1999). It suggests that necrosis or cell stress imposed by reactive drug metabolites provides a “danger signal” that activates macrophages, antigen presenting cells or other cells. The danger signals from stressed cells lead to upregulation of costimulatory molecules or production of cytokines which cause an enhanced antibody or T-cell-mediated specific immune response. However, it is not clear what these danger signals are. Activation of the innate immune system, eg. inflammation, was proposed to be a potential danger signal (Kaplowitz, 2005). Mitochondrial abnormality hypothesis This theory proposes that the mitochondria are targeted by idiosyncratic drugs, and mitochondrial abnormality is the underlying mechanism of IADRs. Mitochondria are a critical player in mediating cell death and also a common target of xenobiotics (Wallace and Starkov, 2000). Numerous drugs associated with IADRs, such as troglitazone, tolcapone, nimesulide and valproic acid are reported to cause mitochondrial dysfunction in vitro (Bjorge and Baillie, 1991; Mingatto et al., 2000; Bedoucha et al., 2001; Haasio et al., 2002). There is also clinical evidence associating IADRs with mitochondrial dysfunction. In one case .M , 5' OCXUUEU An I m. “I" H“ , ,,,, rafzr: f5 ra'spc-n For mart} Oar-BIA IT. .vnv- Iv! Hzfi fihfi advu VCR v . Ila-I ‘IW H‘finah,‘ ' - -. «9'91 IV~ M‘Aahfi. \U'vr U JMRM‘- gluav't P R ‘A.A. “3.3.3.... “rezsrw ‘VLVI report, mitochondrial swelling, loss of cristae and reduced matrix density was observed in hepatocytes of a patient with tolcapone-induced liver injury. An inherited mitochondrial dysfunction or mutation in mitochondrial DNA might make people more susceptible to drug-induced toxicity. In one case of valproate-induced liver injury, an inherited dysfunction of mitochondrial electron transport chain complexes was observed in the patient (Krahenbuhl et al., 2000). For many unrelated drugs, age is one of the risk factors for IADRs. Interestingly, mitochondrial DNA mutations also increase with age, which could explain why aged people are more sensitive to IADRs (Kujoth et al., 2005). Models have been developed using an animal model of silent genetic mitochondrial abnormality. Superoxide dismutase (SOD) eliminates superoxide in mitochondria, and heterozygous SOD2 mice have a decreased ability to manage oxidative stress in the liver. Either nimesulide or troglitazone induced hepatotoxicity in heterozygous SOD2 mice, whereas these drugs had no hepatotoxic effect on normal mice (Ong et al., 2006; Ong et al., 2007). Failure to adapt hypothesis It has been observed that the majority of patients with ALT elevations due to drugs associated with idiosyncratic liver injury will eventually recover from liver injury despite continued exposure to the drug (Watkins, 2005). Therefore, another hypothesis has been raised that a small fraction of patients fail to adapt to the initial injury, which then leads to the progression of severe liver injury. TI‘ .5 myrreql xcunenl ea:ll 9“ hypothes- fi.’"~fl A P'VL': ‘51 lie-”‘7'? 1.1.3 lnl'li In at ppm-n y wu'vse: el al. 2’ ii Mans. exp-Esme L333 cfys. tr“; ~ awa- (Hewett - C ”2A. Uflb‘rs‘“.- ., k: Few animal models have been developed to support this hypothesis, and the mechanisms underlying injury are not known. One possibility is that a single inherited defect in adaptation precipitates drug-induced liver injury. Multiple determinant hypothesis This theory proposed that idiosyncratic drug toxicity is a result of “the occurrence of multiple critical and discrete events, with the probability for the occurrence of idiosyncratic drug toxicity being a product of the probabilities of each event” (Li, 2002). This theory is not exclusive with some of the other hypotheses already discussed. The critical events could include chemical properties of the drug, exposure, environmental factors and genetic factors. Inflammation could be a critical event to take into account. 1.1.3 Inflammatory stress hypothesis as potential explanation for IADRs In addition to the hypotheses mentioned above, a hypothesis has been proposed that inflammation may render an individual susceptible to IADRs (Roth et al., 2003; Ganey et al., 2004). Inflammatory episodes are commonplace in humans. Inflammation can be induced by infections, inflammatory diseases or exposure to endotoxin, which is a potent inducer of inflammation. Lipopolysaccharide (LPS), a cell wall component of gram-negative bacteria, is an inflammagen that can cause damage to several organs, including the liver (Hewett and Roth, 1993). Various conditions, including alcohol consumption, gastrointestinal distress, changes in diet, antibiotic treatment and surgery can IDC'EHSE 03“.".8.’ mpsenr ma. tx*~e V. .VL. increase LPS concentrations in human plasma (Roth et al., 1997). These conditions are not usually severe enough to cause overt illness, but they may potentiate the toxicity of xenobiotics through activating inflammatory cells and cytokines. Previous studies proved that an inflammatory stress precipitates hepatotoxicity of numerous xenobiotics, e.g. allyl alcohol and aflatoxin (Sneed et al., 1997; Yee et al., 2000; Barton et al., 2001). The characteristics of inflammation described above led to the inflammatory stress hypothesis for IADRs. As illustrated in Fig. 1.1, the decrease in threshold for toxicity results from the inflammatory episodes that occur in the lifetime of normal humans. When an individual is undergoing drug therapy, the drug concentration reaches its therapeutic concentration without causing toxicity under normal conditions. However, if an inflammatory stress decreases the threshold of toxicity below the drug concentration, an IADR may occur. This hypothesis can provide a plausible explanation for the characteristics of IADRs. The occurrence and magnitude of a modest inflammatory episode can be unnoticeable in humans. Therefore, IADRs due to the interaction of an inflammatory episode with idiosyncratic drugs would be expected to have an inconsistent temporal relationship to exposure and not appear to be dose dependent. Numerous drug-induced idiosyncratic liver injury models have been developed in rodents that support the inflammatory stress hypothesis. Specifically, drugs including diclofenac (DCLF), sulindac (SLD), halothane (HAL), chlorpromazine (CPZ), trovafloxacin (TVX) and ranitidine (RAN), when administered at nonhepatotoxic doses, induced significant liver injury in rodents 10 Fig. 1.1 I 2003). Th stats : treshcld ll?" Tina: x'certt'a A ”and Drug Conc. In Plasma Fig. 1.1 Inflammation might precipitate drug- induced IADRs (Roth et al., 2003). The subcurve indicates the drug concentration in plasma increases and remains at a therapeutic concentration during drug therapy. However, the threshold for drug toxicity (upper dotted line) might be decreased as a result of inflammatory stress. IADRs occur if the threshold for toxicity drops below drug concentration. y / Threshold for Toxicity LII-Io. .Il-lll- ..II-u-II-n. . e 4‘ a . ‘ E E ‘ f ‘- : ' E 5 O i : 1 .' : :. E E.‘ i i 1" a a. ...l.|"... .E .5 ‘ 6 S U \ 3 IIIIII I IIIIIIIIIIILK I ldlosyncratlc D Threshold for Toxic Res onse Phannacologic w Time on Maintenance Drug Therapy_ 11 pretreated with LPS (Table 1.1). Levofloxacin and famotidine are in the same pharmacological class with trovafloxacin and ranitidine, respectively, but they have far lower tendency to cause IADRs in humans and also failed to induce liver injury with LPS in rodents. Inflammatory stress induced by the viral RNA mimetic, poly (I:C) also precipitated hepatotoxicity caused by halothane in mice (Cheng et al., 2009). Therefore, these results suggest that inflammation may precipitate the toxicity of drugs that cause IADRs. Mechanisms underlying drug/LPS interaction models of idiosyncratic liver injury have been investigated. In the following sections of this chapter, the innate immune response and mechanisms of several drug/LPS interaction models are introduced. 12 Tame ‘ modem t L. A04 r u-V—‘V‘: s «a v -u [I . in“ A..- r: -,_ ch ..n “an ' 9' law“. LEI, R .3. :. Fa"? Table 1.1 List of Idiosyncratic drugs that cause liver injury in drug/LPS models in rodents (Deng et al., 2009). LPS + Drug Drug Human IADRs? Hepatotoxicity in Rodents? Diclofenac Yes Yes Sulindac Yes Yes Halothane Yes Yes Chlorpromazine Yes Yes Trovafloxacin Yes Yes Levofloxacin No No Ranitidine Yes Yes Famotidine No No 13 1.2 1.2.1 2. ...\.. nu )- I it" We a 1.2 Inflammation 1 .2.1 Overview of the inflammatory response People are exposed to infectious microorganisms in everyday life. Inflammation is a first-line defense mechanism by the innate immune system against infections. Not only infections, but also tissue trauma and non-infectious disease can contribute to inflammation (Trunkey, 1988). Tissue injury leads to activation of the innate immune response after trauma, which presents as a systemic inflammatory response syndrome (Lenz et al., 2007). Inflammation is also associated with anemia (Buck et al., 2009), Parkinson’s disease (Barcia et al., 2003), diabetes (Granic et al., 2009), obesity (Elmarakby and Imig, 2009; Olefsky, 2009), heart disease and other vasculopathies (Linde et al., 2006) and cancers (Sevinir et al., 2003; De Marzo et al., 2007). Inflammation is a complex response coordinated by various inflammatory cells (Fig. 1.2). Neutrophils, macrophages, endothelial cells, epithelial cells and platelets are activated by inflammatory stimuli (Ganey et al., 2004). Transcriptional activation occurs, and several genes are upregulated in inflammatory cells. Proteases and reactive oxygen species are released by neutrophils and macrophages and can kill parenchymal cells directly. Proinflammatory cytokines, such as tumor necrosis factor-a (T NF) are also released by macrophages. These cytokines can either activate cell death signaling pathway or lead to other inflammatory events. The coagulation system as well as the complement system can also be activated as a result of inflammation. 14 Fig. 1.2 Summary of the inflammatory response (Ganey et al., 2004). Inflammatory cells are activated by stimulus through receptor binding e.g. TLRs. As a result, numerous mediators of inflammation are released and cause homeostatic imbalance in the target tissue. These mediators might lead to tissue injury in aggravated conditions or be inconsequential, beneficial or increase tissue sensitivity when the response is modest. Inflammatory Stimulus (eg, LPS Ree. l v n: W Macrophages ’ ‘I Neutrophils Endothelial Cells Epithelial Cells Platelets Transcription factor activatio Genelprotein expression Cytoldnee Activated coagulation system Arachidonlc Acid Metabolites Activated complement Platelet Activating Factor Reactive Oxygen Species Proteases Nitric Oxide, Etc., Etc. M: Microbe destruction W: Tissue lnconsequential, + Hemostasis, Edema _) Injury & Beneficial, or Altered perfusion Dysfunction Increased Sensitivity Host cell injury, Etc. 15 I." .A" 1% :1. an.— IN! .3... 47.. I.\~ gnu |I *1 B. a h ad. .3 Hi» Severe inflammation can lead to dramatic changes in physiology, such as redness, swelling, pain and heat at the site of inflammation. Moreover, severe tissue damage or organ failure can result from a marked inflammatory response. More commonly, modest inflammatory episodes occur in humans, which may be unnoticeable. Although modest inflammation resolves and may have no detrimental effect alone, it might potentiate the toxicity of xenobiotics (Sneed et al., 1997; Yee et al., 2000; Barton et al., 2001). The Toll-like receptor (TLR) signaling pathway is a well studied pathway involved in the initiation of the inflammatory response (O'Neill, 2008). TLRs are expressed by inflammatory cells and recognize specific structures conserved among microorganisms (Takeda et al., 2003). LPS binds to TLR4, which results in signal transduction leading to a cascade of inflammatory events (Linde et al., 2006). Macrophages are the primary immune cells responding to LPS. TLR4 is also expressed on the membranes of hepatocytes, endothelium and mast cells (Migita et al., 2004). TLR4 signals through two intracellular Toll/lL-1 receptor (T IR) domain-containing adaptors, myeloid differentiation factor 88 (MyD88) and TlR-containing adapter molecule (T RIF). In the MyD88 dependent pathway, interleukin-1 receptor-associated kinase (IRAK) 1 and IRAK 4 are recruited to MyD88. Once phosphorylated, IRAK dissociates from MyD88 and activates TNF receptor associated Factor (I' RAF) 6. TRAF6 binds to the complex of TGF-B— activated kinase 1 (TAK1) and TAK-1 binding protein (TAB). In turn, TAK1 is phosphorylated and leads to the activation of MAPK pathway or NFkB-regulated genes. In the TRIF dependent pathway, IKB kinase is activated and leads to the 16 I ‘ Mar HIV. ‘ activation of NFKB. TRIF also leads to the activation of interferon regulatory factor 3 (IRF3) and the expression of interferon-inducible genes. 17 1.2.1 TI TNF raC'Opl: lGO'dcn res dent '5TNF 3335a; TNF P'n ‘ DAV. DOC“:: 1.2.1 Tumor necrosis factor-a (TNF) TNF is a proinflammatory cytokine that is produced by many cell types, eg. macrophages, mast cells, endothelial cells and stellate cells in response to LPS (Gordon and Galli, 1990; Thirunavukkarasu et al., 2006). Kupffer cells are resident macrophages attached to the outer surface of endothelial cells in liver sinusoids (Hewett and Roth, 1993), and they are the major macrophage population exposed to inflammagens in blood. After LPS binding to TLR4, pro- TNF is produced by Kupffer cells as a result of the activation of NFKB signaling pathway. Pro-TNF is a 26 kDa membrane-bound precursor form which is not biologically active (Solomon et al., 1997). TNF converting enzyme (l' ACE), a membrane bound metalloprotease, recognizes a cleavable signal sequence on pro—TNF and leads to shedding of the active form of TNF from the cell membrane (lshisaka et al., 1999; Wullaert et al., 2007). TNF exerts its biological effects by binding two plasma membrane receptors, TNF-receptor 1 (TNF-R1) and TNF-R2. In a few cell types, e.g., T cells, the binding of TNF to TNF-R2 leads to proliferation, NFkB activation and cytokine production (Rothe et al., 1994; Vandenabeele et al., 1994). However, in most cells, TNF-R2 has no direct effect on signal transduction and plays an indirect role in TNF-R1 responses by delivering TNF to the low affinity TNF-R1. The ligand passing activity of TNF-R2 allows fine-tuning of TNF-R1 mediated signal transduction. TNF plays an important role in regulating liver homeostasis (Wullaert et al., 2007). TNF-R1 activation leads to either hepatocyte proliferation via the 18 and Fas la the f: 01 p33; activation of NFkB, the initiation of MAPK cascades or cell death (Fig. 1.3). Activated TNF-R1 recruits adapter proteins, TNF-R-associated death domain (TRADD) protein, TNF-R associated factor (TRAF) 2, and receptor interacting protein (RIP) to the cytoplasmic part of TNF-R1. These proteins form complex I. TRAF2 possesses ubiquitin ligase activity, which leads to the ubiquitination of RIP and itself. This serves as an important signal for the binding of TAK1-TAB2- TAB3 complex. TAK1 leads to the activation of the MAPK pathway as well as lKK complex, which leads to the expression of NFkB-inducible genes. On the other hand, activated TNF-R1 can recruit adapter proteins TRADD and Fas—associated death domain (FADD) as well as pro-caspase 8, which leads to the formation of the death-inducing signaling complex (DISC). Autoactivation of procaspase 8 results in the generation of active caspase 8, which directly cleaves procaspase 3 into caspase 3 and leads to apoptosis (Hehlgans and Pfeffer, 2005). Caspase 8 converts Bid into truncated Bid (tBid). tBid contributes to mitochondrial dysfunction via the formation of permeability transition pores and Bak/Bax pores on the outer mitochondrial membrane, which leads to the release of cytochrome c and reactive oxygen species to cytosol. Cytochrome c activates caspase 9 and 3, which lead to apoptosis through activating caspase activated DNase (CAD). Thereby, cell death is induced as a result of TNF-R1 activation. Whether TNF leads to cell survival or death is determined by NFkB (Wullaert et al., 2007). NFkB plays a protective role against cell death by inducing the expression of several antiapoptotic genes, which include caspase-8 inhibitor c- 19 Fig. 1.3 TI InCJw GEEj Fig. 1.3 TNF signal transduction pathway. TNF can either activate NFkB or induce cell death. MAPK ® Caspase 3 Caspase 9 Apoptosis NFkB I mitochondria‘ 0° survival gene 0 expression cytochrome c 20 FLU”L.1 a; 3:135 Ilse-3w flan .fi‘ :31 . -, JNK 3: 51.3873): AAA AJHVI FLIPL, the Bcl-2 family members Bcl—xL and A1/Bfl-1, and X-linked inhibitor of apoptosis (XIAP) (Zong et al., 1999; Chen et al., 2000; Micheau et al., 2001). Moreover, prolonged activation of JNK resulting from ROS accumulation or TNF signaling, causes cell death (Kamata et al., 2005). NFkB can rapidly terminate JNK activation by upregulating the expression of antioxidant manganese superoxide dismutase (MnSOD) and ferritin heavy chain (FHC) (Sakon et al., 2003). 21 1.2.3 1 Va: 1.2.3 The hemostatic system and hypoxia Vascular hemostasis is regulated by coagulation and fibrinolysis. The coagulation system is activated in many inflammatory diseases (Vrij et al., 2003). It can be activated through the extrinsic pathway or the intrinsic pathway. In the extrinsic pathway, tissue factor is exposed to blood and forms a complex with factor VII, which activates factor X through the activation cascade. Factor X cleaves prothrombin into thrombin, which converts fibrinogen into fibrin monomers by cleaving fibrinopeptides. Fibrin can cross-link to form fibrin clots in blood vessels (Mosesson et al., 2001). Fibrin clots are controlled and dissolved by the fibrinolytic system, in which plasmin plays an important role by degrading fibrin clots into D-dimers (Fay et al., 2007). Plasminogen activators (PAs) cleave plasminogen to active plasmin. PAs can be inhibited by active plasminogen activator inhibitor (PAl-1), which is synthesized by hepatocytes, endothelial cells and platelets in response to TNF or LPS (Levi et al., 2003; Westrick and Eitzman, 2007). Therefore, an increase in active PAl-1 can dampen the fibrinolytic system and enhance fibrin deposition. Thrombin and PAl-1 are two critical factors in regulating the hemostatic system, but they also play a role in activating inflammatory cells. Thrombin cleaves and activates G protein-coupled receptors, including protease-activated receptor-1 (PAR-1), which can significantly increase production of pro- inflammatory cytokines (T NF, lL-1 and lL-6) from target cells (Fan et al., 2005). PAI-1 can potentiate LPS-induced neutrophil activation through a JNK-mediated pathway in vitro and enhance nuclear translocation of NFkB, which increases 22 p'oduc 2336:. cl TNF F it $51.". (It detrime :rrpagrir effects t'ated 17253.55 Vittoria :"cg . 3696515 A . LIV-3'3"- ‘v RP., A a P IS'V \. I: “he a ,. ,e"' 3"" he \s IV‘ ' Lr: iL'u' 1.2.4 I IF. ate i SC’iCN, v3“: production of the proinfiammatory cytokines lL-1, TNF and MlP-2 (Kwak et al., 2006). Previous studies in vivo also suggest that PAl-1 contributes to production of TNF, IL-10, KC and MCP-1 (Shaw et al., 2009c). Fibrin deposition controls the magnitude and area of infection, through which fibrin deposition plays a protective role against inflammagen exposure. However, detrimental effects can also be induced by fibrin deposition in liver sinusoids by impairing blood flow and thereby causing tissue hypoxia. Hypoxia exerts various effects on hepatocytes. It activates numerous intracellular signaling pathways related to transcription of hypoxia-responsive genes, mostly mediated by hypoxia inducible factor-1a. Reactive oxygen species that accumulate after exposure to hypoxia lead to the MPT (Qu et al., 2001; Schild and Reiser, 2005). Energy deficit (loss of ATP) and inhibition of aerobic metabolism are also caused by the decrease of available oxygen. Hepatic metabolism and function are highly oxygen-dependent. As a consequence of decreased cellular ATP level, physiological function is impaired and liver damage can occur (Semenza, 2004). Furthermore, hypoxia enhances LPS-induced liver damage (Shibayama, 1987) and potentiates the toxic effects of PMN-derived proteases on hepatocytes in vitro (Luyendyk et al., 2005). 1.2.4 Neutrophils (PMNs) PMNs are abundant blood leukocytes and play an important role in the innate immune response. PMNs are a contributor to tissue injury induced by ischemia-perfusion and alcohol. (Jaeschke et al., 1990; Hewett et al., 1992; 23 a fifths-IO Paul ' A? ‘ fl .. U‘ ‘ 'rj s (I) ‘ Jaeschke, 2002). PMNs are involved in several models of drug-induced liver injury (e.g., acetaminophen) and in drug-LPS interaction models of idiosyncratic liver injury (Deng et al., 2006; Deng et al., 2007b; Jaeschke and Liu, 2007; Ramaiah and Jaeschke, 2007; Shaw et al., 2009d). PMNs are primed and activated by systemic or local exposure to proinflammatory mediators. TNF, lL-1 or CXC chemokines (ClNC-l, MIP-2) may increase the expression of integrin on the surface of PMNs as well as lCAM-1 and VCAM-1 expression on the surface of endothelial cells (Jaeschke and Smith, 1997; Jaeschke et al., 1998). The binding of an adhesion molecule (lCAM-1 or VCAM-1) to its counter-receptor (integrin) results in PMNs adhering tightly to endothelial cells. However, in the RAN/LPS model, neutralization of integrin did not reduce PMN numbers in the liver (Deng et al., 2007b). Thus, accumulation of PMNs in vasculature is not necessarily dependent on the binding of adhesion molecules. PMN migration from the vasculature and infiltration into the parenchyma is a prerequisite for neutrophil cytotoxicity (Jaeschke and Smith, 1997). During this process, a signal from parenchymal cells is required. A chemotactic gradient of CXC chemokines in liver can lead to PMN infiltration, which contributes to liver injury (Okaya and Lentsch, 2003). Necrotic cells can release a mediator called high mobility group box 1 (HMGB1) which can signal to PMNs and cause their migration (T sung et al., 2005). Anti-HMGB1 antibody significantly reduced the production of proinflammatory cytokines and PMN infiltration in an ischemia- reperfusion model of liver injury (T sung et al., 2005). Interestingly, apoptotic cells might also trigger PMN migration (Jaeschke, 2006). It was observed that 24 T) "A A :a 3.. ’ In. we. A. «Jrf' N engulfment of apoptotic bodies by Kupffer cells promotes cytokine expression and PMN activation (Canbay et al., 2003). Alternatively, it has been proposed that gaps in the sinusoidal endothelial cells may facilitate the direct contact of PMNs with altered membranes of apoptotic cells (Jaeschke, 2006). After PMNs cross the endothelial cell barrier, they localize close to hepatocytes and degranulate, leading to the release of proteases and reactive oxygen species (ROS) including hydrogen peroxide and hypochlorous acid (Dahlgren and Karlsson, 1999). Both proteases and ROS contribute to liver injury in animal models (Jaeschke, 2006). Cathepsin G and elastase are two important proteases which are released by activated PMNs. They are important mediators of hepatic parenchymal cell killing (Ho et al., 1996). Inhibition of cathepsin G and elastase protected against liver injury in a drug-inflammation interaction model (Luyendyk et al., 2005). Besides direct effect on hepatocytes, proteases released by PMNs can also contribute to fibrin deposition by activating plasminogen activator inhibitor-1 (PAl-1) (Deng et al., 2007a). The role of ROS in cell death is discussed in the following section. 1.2.5 Reactive oxygen species Reactive oxygen species (ROS) are oxygen free radical and nonradical (but reactive) oxygen species. They include hydrogen peroxide, superoxide, hydroxyl radicals, etc. ROS are produced by several sources within the cell. Mitochondria, in which the electron transport chain transfers electrons to oxygen, are a major source of ROS. During normal cellular respiration, about 2% of electrons escape 25 irt'eas m-dase pfasma gewefa' cage" ..m. ,u l-‘IUI I f??? FA " «I fit: r~~ w VUU'I and lead to production of superoxide anion (Boveris and Cadenas, 1975). As described in previous sections, TNF and hypoxia have been shown to cause increased ROS generation in a mitochondria-dependent manner. Another major source of ROS is nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex. The NADPH oxidase complex assembles on phagosomal, plasma and granule membranes of activated PMNs and macrophages and generates ROS (Dahlgren and Karlsson, 1999). NADPH oxidase reduces oxygen to superoxide at the same time it oxidizes NADPH. Cytochrome b from NADPH is responsible for the transfer of electrons to oxygen present in intracellular compartments (Babior, 1999). In addition, many other enzymes, including xanthine oxidase, cyclooxygenases, lipoxygenases, myeloperoxidases, hemeoxygenase, monoamine oxidases, aldehyde oxidase, and cytochrome P450, can cause ROS accumulation. However, the capacity of these enzymes to generate ROS is less robust than the mitochondrial electron chain complex or NADPH oxidase (Morgan et al., 2008). Excessive ROS may tilt the prooxidant-antioxidant balance, causing oxidative stress in cells. ROS can directly oxidize proteins and lipids as well as nucleic acids, leading to cellular damage and dysfunction (Morgan et al., 2008). ROS may induce cell death through a JNK-dependent pathway (Schwabe and Brenner, 2006). ROS directly inactivate JNK phosphatase, which leads to prolonged activation of JNK (Kamata et al., 2005). Activated JNK phosphorylates antiapoptotic Bcl-2 family proteins (Bcl-2, BcI-XL) and inactivates them (Yamamoto et al., 1999a; Fan et al., 2000). 26 a'ésa' 0'55!“ 3U. . il'sE'S l ctalier 1r] FA: U Oxidative stress plays a role in several models of liver injury. It occurs and contributes to ischemia/reperfusion injury in mice (Serteser et al., 2002). Involvement of oxygen free radicals and consequently of oxidative stress has also been proven in alcoholic liver disease (Koch et al., 2004). R08 are involved in the pathogenesis of inflammatory liver diseases (Jaeschke, 2000). In an animal model of LPS-induced organ injury, a significant decrease in reduced glutathione and an increase in lipid peroxidation were observed in the lungs and livers of rats (Suntres and Shek, 1996). The administration of antioxidants after challenge with LPS resulted in a significant alleviation of both lung and liver injuries. Mice deficient in antioxidant enzyme showed greater susceptibility to PMN-mediated liver injury (Jaeschke et al., 1999). An NADPH oxidase inhibitor protected against endotoxin-induced PMN-mediated liver injury, suggesting a critical role for ROS (Gujral et al., 2004). Oxidative stress has also been proposed as a potential mechanism of NSAlD-induced hepatotoxicity (Boelsterli, 2002). Although the role of ROS in SLD-induced liver injury has not been determined in vivo, SLD and its toxic metabolite were reported to increase ROS production in several cell lines (Galati et al., 2002; Adachi et al., 2007). 27 1.3.1 F Re *‘ROM ”6 rl “ V a . 7 ha» 0.- I :vv \~ 1.3 Models of drug-inflammation interaction There are few animal models to study the mechanisms of drug-induced idiosyncratic hepatotoxicity. The main reason for this is that idiosyncratic hepatotoxicity-induced by drugs may only occur in a small fraction of animals, as it often does in humans. However, based on the hypothesis that inflammation precipitates drug toxicity, several idiosyncratic liver injury models based on drug- inflammation interaction have been developed. 1.3.1 RanitidineILPS- induced hepatotoxicity in rats Ranitidine (RAN) is a histamine-2 (H2)-receptor antagonist used for the treatment of duodenal ulcers, gastric hypersecretory diseases and gastroesophageal reflux disease. RAN is associated with idiosyncratic hepatotoxicity with an incidence of less than 1 in 1000 patients taking the drug (Vial et al., 1991). Famotidine (FAM), although in the same pharmacological class with RAN, has a decreased propensity to cause idiosyncratic reactions. An animal model of RAN-induced idiosyncratic liver injury was developed by pretreating rats with LPS (Luyendyk et al., 2003). In this model, LPS (44.4X106 EU/kg) or its saline vehicle was administered to rats via a tail vein. Two hours later, RAN (30 mg/kg), FAM (6 mg/kg), or their vehicle (sterile phosphate— buffered saline) was administered i.v. at a rate of approximately 0.15 mllmin. Neither RAN nor LPS given alone had a significant hepatotoxic effect as measured by ALT activity compared to control animals. FAM was also not hepatotoxic to rats in the presence or absence of LPS. In contrast, cotreatrnent of 28 rats wi ALT. l quififl' .LVLV'UI aorta} R; “C rats with RAN/LPS led to a significant increase in markers of liver injury (e.g., ALT, AST and GGT) in serum at 6, 12 and 24 hr after RAN treatment. Histopathology demonstrated the presence of midzonal hepatic necrosis in animals receiving RAN/LPS cotreatrnent. RAN but not FAM enhanced the LPS-induced TNF increase before the onset of hepatocellular injury (Tukov et al., 2007b). It was also observed that a large concentration of RAN enhanced LPS-induced TNF release in a Kupffer cell- hepatocyte coculture system. RAN enhanced the activation of p38 induced by LPS, which led to increased TACE activation (Deng et al., 2008). TACE cleaved pro-TNF into TNF and led to an increase in TNF concentration in the plasma. TNF plays a critical role in the pathogenesis of liver injury, which is supported by the evidence that TNF neutralization protected against liver injury induced by RAN/LPS. RAN also enhanced the increase in serum interleukin (lL)-1beta, lL-6 and lL-10 induced by LPS. In plasma of rats treated with RAN/LPS, a decrease in fibrinogen and increases in thrombin-antithrombin (TAT) dimers and PAl-1 occurred before the onset of liver injury, suggesting that the hemostatic system was activated by the cotreatrnent (Luyendyk et al., 2004). Hepatic fibrin deposition was observed in livers of rats cotreated with RAN/LPS at 3 after RAN (ie, before the onset of liver injury). The anticoagulant heparin or the fibrinolytic agent streptokinase significantly reduced liver injury induced by RAN/LPS. Hypoxia probably resulting from sinusoidal fibrin deposition was observed in livers of RAN/LPS-treated rats at 3 hr, and this was significantly attenuated by heparin. 29 I ccfiflr (gag. nct a.“ tasma PMNs are also critical to RAN/LPS-induced liver injury (Luyendyk et al., 2006). PMN accumulation occurred in livers of rats treated with LPS/RAN. Depletion of PMNs using anti-PMN serum protected against liver injury, suggesting that PMNs are involved in the pathogenesis. TNF, the hemostatic system and PMNs do not act independently of each other. TNF contributes to RAN/LPS-induced liver injury by enhancing production of inflammatory cytokines, chemokines (MlP-2) and hemostatic factors including TAT and PAl-1 (T ukov et al., 2007b). However, hepatic PMN accumulation was not affected by TNF. Heparin had little effect on liver PMN accumulation or plasma chemokine concentration, indicating that PMN accumulation is not affected by fibrin deposition in the liver (Luyendyk et al., 2006). However, both TNF and PAl-1 contribute to PMN activation (Deng et al., 2007b; Deng et al., 2008). PMN depletion reduced the plasma concentration of active PAl-1 and fibrin deposition in livers of rats treated with RAN/LPS, which suggests that PMNs promote fibrin deposition by increasing PAl-1concentration (Deng et al., 2007b). As a result, PMNs also promote hypoxia in the liver. These studies suggest that mediators involved in the pathogenesis of liver injury induced by RAN/LPS are not isolated, but interact with each other. 1.3.2 Diclofenac/LPS-induced hepatotoxicity in rats Diclofenac (DCLF) is a nonsteroidal anti-inflammatory drug (NSAID) associated with serious idiosyncratic hepatotoxicity in humans (Aithal, 2004). The incidence of DCLF-induced liver injury is approximately one to five cases per 30 IIVer min DCLF. I. 01' 15 58 “A ‘1‘. 93h. No Was E 100,000 persons exposed (Garcia Rodriguez et al., 1994). Liver failure has been recorded after the administration of DCLF (Greaves et al., 2001). Several hypotheses of DCLF-induced liver injury have been proposed, including metabolic polymorphism and allergic hypersensitivity (Daly et al., 2007). However, in vivo evidence that supports these hypotheses is still lacking. As in the RAN/LPS-induced liver injury model, a model of DCLF-induced liver injury in rats was developed by treating rats with a nonhepatotoxic dose of DCLF, LPS or their vehicles (Deng et al., 2006). Generally, LPS (29 X106 EU/kg) or its saline vehicle was administered to rats via a tail vein. Two hours later, rats were given DCLF (20 mg/kg, i.p.) or sterile saline. Neither LPS nor DCLF alone had an effect on ALT activity. However, cotreatment with LPS and DCLF caused a significant increase in serum ALT activity. Hepatocellular apoptosis, parenchymal edema, and hemorrhage induced by LPS were also significantly increased by DCLF cotreatrnent. A gene array study was performed to compare the gene expression patterns among LPS, DCLF and cotreatrnent groups. Genes encoding the neutrophil chemokines, such as MlP-2 and MlP-1, and the adhesion molecule lCAM-l, were greatly increased by DCLF/LPS cotreatrnent compared to LPS or DCLF alone. Both LPS alone and DCLF/LPS treatment led to hepatic PMN accumulation. DCLF did not enhance the effect of LPS on PMN accumulation, although the increase in MlP-2 concentration in serum of rats treated with LPS was enhanced by DCLF. Anti-PMN serum reduced PMN accumulation in liver 31 rats, lnAEarfi I V ‘ l“ I ; ”Na and attenuated liver injury induced by DCLF/LPS. This result suggests that PMNs play a critical role in the pathogenesis. Interestingly, a larger dose of DCLF (100 mglkg, i.p.) caused liver injury in rats, which was attenuated by treatment with nonabsorbable antibiotics (polymyxin B and neomycin) for 4 days before DCLF administration. This suggests that bacterial translocation from intestine to liver plays a critical role in DCLF-induced hepatotoxicity though interacting with DCLF. 1.3.3 Trovafloxacin-inflammation interaction model of idiosyncratic liver injury Trovafloxacin (TVX) is a broad spectrum antibiotic which functions through inhibiting bacterial topoisomerase IV. TVX was approved for marketing in 1997. Two years later, its use was severely limited due to the risk of hepatotoxicity. TVX is associated with idiosyncratic hepatotoxicity with an incidence of 1 in 18,000 prescriptions (Stahlmann, 2002). Another fluoroquinolone antibiotic, levofloxacin (LVX), has not been associated with idiosyncratic liver injury and was used in model development as a control (Shaw et al., 2007). Both a rat and mouse model of liver injury induced by TVX/LPS cotreatrnent was developed (Shaw et al., 2009d). In the mouse model (Shaw et al., 2007), TVX (150 mglkg), LVX (375 mg/kg) or their vehicle (saline) was administered to mice by oral gavage. LPS or its vehicle was administered intraperitoneally to mice 3 hr after the drug. TVX, LVX, LPS alone or LVX/LPS cotreatment did not increase plasma ALT activity, whereas TVX/LPS cotreatrnent significantly 32 increased after LPS treated W‘ .09chth fl et al. 23 liver inju' .pporfec ”7.qu in “53}. l ii”... 50’ increased this biomarker of hepatocellular injury at 9 hr and peaked at 15-21 hr after LPS administration. Hepatocellular necrosis was observed in livers of mice treated with TVX/LPS, but not in those treated with TVX, LVX or LPS alone. The necrotic foci observed in the TVX/LPS-treated group were found in midzonal and centrilobular regions. TVX prolonged the appearance of TNF induced by LPS in the plasma (Shaw et al., 2007). TNF neutralization using etanercept attenuated TVX/LPS-induced liver injury, suggesting that TNF is an important mediator. This was further supported by evidence that TVX and TNF cotreatrnent caused significant liver injury in mice, whereas neither TVX nor TNF was hepatotoxic (Shaw et al., 2009a). Interestingly, TVX prolonged the appearance of TNF in the plasma of mice cotreated with TVX/TNF compared to mice given only TNF. This prolonged appearance of TNF was caused by both enhanced production and decreased clearance of this cytokine. Comparison of hepatic gene expression profiles from mice treated with TVX/LPS to those treated with LPS or TVX alone suggested that the interferon v (lFNv) signaling pathway was selectively activated by TVX/LPS (Shaw et al., 2009b). TVX enhanced the appearance of interleukin (lL)-18 that contributes to the production of lFNy. In turn, IFNy can feedback to increase lL-18. TVX/LPS-induced liver injury was attenuated in either lFNy -/- or lL-18 -/- mice, which indicate both lFNy and lL-18 are important mediators in the pathogenesis of TVX/LPS-induced liver injury. Compared to LPS, TVX, LVX alone or LVX/LPS cotreatrnent, TVX/LPS caused a significant increase in plasma concentration of TAT dimers and PAl-1, 33 WTlCl'l w Ezner F WXLPE tSnW PM TNF is l Za'lclK which was accompanied by fibrin deposition in the liver (Shaw et al., 2009c). Either PAl-1 knockout or heparin treatment reduced liver injury caused by TVX/LPS, indicating that PAH and fibrin deposition contributed to liver injury in this model. PMNs also contribute to TVX/LPS-induced liver injury (Shaw et al., 2009d). TNF is responsible for the production of PMN chemokines including MlP-1, MIP- 2 and KC in this model (Shaw et al., 2009e). In addition to the TVX/LPS interaction model of idiosyncratic liver injury, inflammation induced by a Gram-positive stimulus, a peptidoglycan-lipoteichoic acid (PGN-LTA) mixture isolated from Staphylococcus aureus, also precipitates TVX-induced liver injury in mice (Shaw et al., 2009d). PGN and LTA activate TLR2 to induce inflammation, which indicates that liver injury induced by drug- inflammation interaction is not necessarily dependent on TLR4 pathway. Besides the three animal models introduced here, there are other models of IADRs, in which liver injury is induced by the cotreatrnent of chlorpromazine/LPS, halothane/LPS and halothane/poly l:C. However, the mechanisms underlying these models are not yet fully understood. 34 1.4 Sulindac-induced idiosyncratic liver injury Sulindac (SLD) is a prodrug in the therapeutic class of NSAIDs. SLD was introduced into the market in 1978 by Merck under the brand name Clinoril to relieve pain, tenderness, swelling, and stiffness caused by osteoarthritis, rheumatoid arthritis and ankylosing spondylitis. Typically, the dose of SLD for human patients is 150-200 mg, twice per day as a result of its 8 hr half life. The bioavailability of SLD is more than 90%. SLD is absorbed rapidly upon oral administration, and reaches a peak in human plasma in 2-4 hr (Davies and Watson, 1997). SLD and its metabolites are secreted into bile and undergo enterohepatic circulation (Bolder et al., 1999). SLD and its metabolites are excreted in urine and feces. Unlike prodrugs that are irreversibly bioactivated to active metabolites, SLD can be reversibly converted to the active metabolite, SLD sulfide, and irreversibly converted to SLD sulfone (Fig. 1.4). According to previous studies, two enzymes are responsible for SLD metabolism: methionine sulfoxide reductase (MSR) in both liver and gut flora reduces SLD to SLD sulfide, and a flavin-containing monooxygenase (FMO) converts SLD to SLD sulfone and also catalyzes the conversion of SLD sulfide to SLD (Etienne et al., 2003b). The SLD active metabolite, SLD sulfide, performs its pharmacological function by inhibiting cyclooxygenases (COX) -1 and -2 (Lin et al., 1985). Both COX-1 and COX-2 are responsible for the synthesis of prostaglandins. COX-1 is constitutively expressed in normal cells and plays a beneficial role, e.g.,maintain the normal function of GI tract, renal tract, platelet function. COX-2 is inducible 35 Fig. 1.4 Metabolism of SLD (Duggan et al., 1980). SLD can be reversibly converted to the active metabolite, SLD sulfide by methionine sulfoxide reductase (MSR), and irreversibly converted to SLD sulfone by flavin-containing monooxygenase (FMO). F 0|chsz F enzcozn F CHM We"; 32+ cu; fl‘°_. cu; ca 73;— CN 0 cu c333/E:][’ IE::]’ ca§§’E::l' 3‘ 5° 0 0 SULFIDE SULINDAC SULFONE (SULFOXIDE) 36 enzyme expressed by macrophages and contributes to inflammation. Therefore, the inhibition of COX—2 is effective to resolve inflammation. SLD has been used in the United State for over a decade, during which it has been associated with increased risk of heart attack, ulcer, stroke and liver injury (Tarazi et al., 1993). The Food and Drug Administration (FDA) Arthritis Advisory Committee wrote that “the potential for producing liver injury is a class characteristic of NSAIDs” (Paulus, 1982; Tarazi et al., 1993). SLD was associated with a 5—10 fold higher incidence of hepatic injury than other NSAIDs, which induced liver injury at an incidence of about 1 in 100,000 (Walker, 1997). According to the analysis of cases reported to FDA, SLD-induced liver injury often occurred within 8 weeks of taking the drug, whereas about 20% of the reported reactions occurred after 8 weeks of treatment (T arazi et al., 1993). Females are more susceptible to toxicity of SLD than males, and two thirds of the patients were over 50 years of age. Histopathology showed that the pattern of SLD-associated liver injury can be cholestatic, hepatocellular or mixed. In the cases of hepatocellular injury, the lesions were spotty and panacinar in most cases (8 out of 9). Portal inflammation and eosinophil infiltration was observed in a portion of patients. Mechanisms of SLD-induced idiosyncratic liver injury are not well understood. Because clinical characteristics consistent with hypersensitivity were observed in some patients, it was proposed that hypersensitivity accounts for a significant proportion of SLD-induced liver injury. However, no direct evidence for this has been found, and the mechanisms of pathogenesis still require further 37 investigation. Another hypothesis for liver injury induced by NSAIDs including SLD is through mitochondrial injury. A variety of NSAIDs or their metabolites (e.g., nimesulide, DCLF and SLD) have a toxic effect to the mitochondria of hepatocytes in vitro. There is little evidence in vivo supporting this hypothesis, and an idiosyncratic liver injury model for SLD has not been developed on the basis of this hypothesis. 38 1.5 Hypothesis and specific aims The overall hypothesis of this dissertation is that an inflammatory episode induced by LPS precipitates SLD-induced liver injury in rats. According to previous studies on the mechanism of LPS- or drug/LPS- induced liver injury, we hypothesize that TNF, the hemostatic system, PMNs and ROS are elevated by SLD/LPS cotreatrnent and play a critical role in the pathogenesis of liver injury. The SLD toxic metabolite, SLD sulfide also contributes to liver injury by synergistically interacting with those inflammatory mediators. Aim 1 Hypothesis Cotreatment with nonhepatotoxic doses of SLD and LPS causes idiosyncrasy- like Iiver injury in rats. (Chapter 2) Aim 2 Hypothesis The hemostatic system is activated by SLD/LPS cotreatrnent in rats and contributes to liver injury by causing hypoxia. (Chapter 2) Aim 3 Hypothesis TNF, which is increased by SLD/LPS cotreatrnent, plays an important role in liver injury by interacting with SLD sulfide. (Chapter 3) Aim 4 Hypothesis 39 PMNs are activated in livers of rats treated with SLD/LPS and contribute to liver injury by releasing toxic proteases. (Chapter 4) Aim 5 Hypothesis ROS production is increased in livers of rats treated with SLD/LPS. ROS are involved in the pathogenesis of liver injury by enhancing the cytotoxicity of TNF. (Chapter 5) 40 CHAPTER 2 Zou W, Devi SS, Sparkenbaugh E, Younis HS, Roth RA and Ganey PE (2009) Hepatotoxic interaction of sulindac with lipopolysaccharide: role of the hemostatic system. Toxicol Sci 108:184-193. 41 2.1 Abstract Sulindac (SLD) is a nonsteroidal anti-inflammatory drug (NSAID) that has been associated with a greater incidence of idiosyncratic hepatotoxicity in human patients than other NSAIDs. One hypothesis regarding idiosyncratic adverse drug reaction (IADRs) is that interaction of a drug with a modest inflammatory episode precipitates liver injury. In this study, we tested the hypothesis that lipopolysaccharide (LPS) interacts with SLD to cause liver injury in rats. SLD (50 mglkg) or its vehicle was administered to rats by gavage 15.5 hr before LPS (8.3X105 EU/kg) or its saline vehicle (i.v.). Thirty min after LPS treatment, SLD or vehicle administration was repeated. Rats were killed at various times after treatment, and serum, plasma and liver samples were taken. Neither SLD nor LPS alone caused liver injury. Cotreatment with SLD/LPS led to increases in serum biomarkers of both hepatocellular injury and cholestasis. Histological evidence of liver damage was found only after SLD/LPS cotreatrnent. As a result of activation of hemostasis induced by SLD/LPS cotreatment, fibrin and hypoxia were present in liver tissue before the onset of the hepatotoxicity. Heparin treatment reduced hepatic fibrin deposition and hypoxia and protected against liver injury induced by SLD/LPS cotreatrnent. These results indicate that cotreatrnent with nontoxic doses of LPS and SLD causes liver injury in rats, and this could serve as a model of human idiosyncratic liver injury. The hemostatic system is activated by SLD/LPS cotreatrnent and plays an important role in the development of SLD/LPS-induced liver injury. 42 2.2 Introduction Previous studies suggested that mild inflammation induced by bacterial lipopolysaccharide (LPS) could potentiate hepatotoxicity in rodents from IADR- associated drugs such as chlorpromazine (Buchweitz et al., 2002), ranitidine (Luyendyk et al., 2003) and trovafloxacin (Shaw et al., 2007). In one of these LPS/drug interaction models, the hemostatic system proved to be important in liver pathogenesis (Luyendyk et al., 2005). This system comprises coagulation and fibrinolytic components. In coagulation, thrombin plays a critical role by cleaving fibrinogen to fibrin that can form occlusive clots in sinusoids. Meanwhile, plasminogen activator inhibitor-1 (PAl-1) can inhibit fibrin clearance by the fibrinolytic system by inhibiting the generation of plasmin from plasminogen. To evaluate the utility of LPS/drug interaction models to study mechanism(s) of idiosyncratic liver injury, a broad range of drugs needs to be tested. Therefore, the purpose of this study was to test the hypothesis that modest inflammation induced by LPS interacts with SLD to cause liver injury in rats. When the results demonstrated a hepatotoxic interaction, the role of the hemostatic system was explored. 43 2.3 Materials and methods 2.3.1 Materials Unless otherwise noted, all chemicals were purchased from Sigma-Aldrich (St. Louis, MO). The activity of lipopolysaccharide (Lot 075K4038) derived from Eschen'cia coli serotype O55:B5 was 3.3 X 106 endotoxin units (EU)/mg as determined by a Limulus amebocyte lysate endpoint assay kit purchased from Cambrex Corp. (Kit 50-650U; East Rutherford, NJ). The reagents for alanine aminotransferase (ALT), aspartate aminotransferase (AST), y- glutamyltransferase (GGT) and total bilirubin were purchased from Thermo Corp. (Waltham, MA). Alkaline phosphatase (ALP) reagent was purchased from BioAssay Systems (Hayward, CA), and the kit for total bile acid determination was purchased from Diazyme Laboratories (Poway, CA). 2.3.2 Animals Male, Sprague-Dawley rats (Crl:CD(SD)lGS BR; Charles River, Portage, Ml) weighing 250 to 370 g or 150 to 200 g were used for in vivo or in vitro studies, respectively. Animals were allowed to acclimate for 1 week in a 12-hr light/dark cycle prior to use in experiments. They were fed standard chow (Rodent Chow/Tek 8640; Harlan Teklad, Madison, WI) and allowed access to water ad libitum. 2.3.3 Experimental protocol 44 Two administrations of SLD were used in the studies (Fig. 2.1). In a dose- response study, rats were given the first administration of SLD (10, 20, 50, 100 or 300 mglkg, p.o.) or its vehicle (0.5% methyl cellulose), and food was removed for 24 hr. 15.5 hr after the first administration of SLD, LPS (8.25X 105 EU/kg, i.v.) or its saline vehicle was administered. Half an hour later, a second administration of SLD (same dose) or its vehicle was given. Rats were anesthetized with isoflurane and killed at various times after the second administration of SLD. For subsequent studies, 50 mg/kg was chosen as the dose of SLD. For all studies, blood was drawn from the vena cava of anesthetized rats, and part was transferred into vacutainer tubes (Becton Dickinson) containing sodium citrate for preparation of plasma. The rest of the blood was allowed to clot at room temperature for preparation of serum. The exterior of the liver was rinsed with saline, and a portion of the left medial lobe was snap frozen in cooled methylbutane for immunohistochemistry. Three slices of the left lateral lobe about 3-4 mm thick were fixed in 10% buffered formalin for histological analysis. In experiments designed to evaluate the role of the hemostatic system, anticoagulant heparin (3000 Units/kg, so) or its saline vehicle was given to rats 0 and 6 hr after the second administration of SLD. 2.3.4 Evaluation of liver injury Hepatic parenchymal cell injury was assessed by measuring the activities of ALT and AST in serum. Cholestatic injury markers, including the activities of ALP and GGT, as well as the concentrations of total bilirubin and bile acids in serum, were 45 Fig. 2.1. Experimental protocol for animal treatment. Rats were given SLD or its vehicle (0.5% methyl cellulose) at —16 hr, and food was removed. At -0.5 hr 5 rats received LPS (8.25X10 EUIkg, i.v.) or its vehicle (saline), and 30 min later they were given a second administration (same dose as 1St administration) of SLD. 13t SLD 2"Cl SLD (10 - 300 mglkg, p.o.) (10 - 300 mglkg, p.o.) or Veh or Veh LPS Rats euthanized 5 . samples taken (8.25 x 10 EUIkg, i.v.) or Veh l w k V ‘ l Ur ' ‘ ) Hr -16 -0.5 0 24 46 also assessed (see above). Forrnalin-fixed liver slices were embedded in paraffin and cut into 6 um sections. Hematoxylin and eosin (H&E) staining was performed, and sections were examined under 100X magnification using a light microscope. Eight, randomly chosen microscope fields for each slide were evaluated for midzonal necrosis and assigned a score of 0-5. 0 represents no liver injury, and 1-5 represents lesions ranging from single cell necrosis (1) to necrotic area encompassing greater than 30% of the field (5). The average score was determined for each rat. 2.3.5 Evaluation of serum TNFa concentrations The concentration of TNFa in serum taken at 1 hr after the second administration of SLD was measured using an ELISA kit purchased from BD Biosciences (San Diego, CA). 2.3.6 Evaluation of hemostasis and fibrin deposition Thrombin-antithrombin dimer (TAT) concentration in plasma was used as a marker of thrombin activation and evaluated using an ELISA kit (catalog number OWMG15) purchased from Dade Behring, Inc. (Deerfield, Illinois). The concentration of the active form of plasminogen activator inhibitor-1 (active PAI- 1) was determined using a kit from Molecular Innovations, Inc. (Southfield, MI). The immunohistochemistry and quantification for cross-linked fibrin in liver were performed as described previously (Copple et al., 2002). Fibrin monomer is 47 solubilized in this protocol, and only cross-linked fibrin in liver is stained. To investigate whether fibrin deposition occurs before the onset of injury, livers were collected at 4 hr and fixed for immunohistochemistry. The fraction of positive pixels averaged from 10 randomly chosen microscope fields was determined for each animal. 2.3.7 Evaluation of liver hypoxia Liver hypoxia was evaluated by quantifying pimonidazole (PlM)-protein adducts. PIM is a hypoxia probe which is rapidly reduced under low p02 conditions to a reactive intermediate that forms PIM-protein adducts. PIM hydrochloride (Hypoxyprobe-1, 120 mg/kg; Chemicon International, Temecula, CA) was given to rats 2 hr before sacrifice. Four hr after the second administration of SLD, livers were collected and fixed for immunohistochemistry. The fraction of positive pixels averaged from 10 randomly chosen microscope fields was determined for each rat (Copple et al., 2004). 2.3.8 Hepatocyte (HPC) isolation and hepatocytotoxicity assessment in vitro HPCs were isolated from rat liver as previously described (Tukov et al., 2006). Isolated cells were suspended in Williams’ Medium E (Gibco BRL, Rockville, MD) with 10% fetal bovine serum, and cell viability was evaluated using trypan blue exclusion. The cell viability was always above 80%. The HPCs were suspended and plated randomly at a density of 2.5 x 105 cells/well in 12- 48 well plates (Corning lnc., Corning, NY). After 2.5 to 3 hr incubation which allowed HPCs to attach to the plate, serum-containing medium was removed, and serum-free medium was added. HPCs were treated with 60 uM sulindac sulfide or its vehicle (0.06% DMSO) and incubated in the presence of 20% or 5% O; (with 5% C02 and balance N2). After 8 hr incubation, the medium was collected, and the unattached cells were isolated by centrifugation. Both the remaining attached cells and unattached cells were lysed with 1% Triton X-100. ALT activity in the medium, attached cell lysate and unattached cell lysate was determined. Hepatocytotoxicity was assessed by calculating the ALT activity in the medium plus unattached cells as a percentage of the total ALT activity in the well (medium + unattached cell lysate + attached cell lysate). 2.3.9 Statistical analysis One way or two way analysis of variance (ANOVA) was used for data analysis, and Tukey’s test was employed as a post hoc test. For GGT activity and necrotic lesion score data, an ANOVA on ranks was performed, and Dunn’s test was used for multiple comparisons. P < 0.05 was set as the criterion for statistical significance. 49 2.4 Results 2.4.1 Dose-response and timecourse of liver injury SLD alone did not induce liver injury in rats at any of the doses given. SLD (2 administrations) at doses of 10 or 20 mg/kg did not cause hepatotoxicity in LPS-treated rats; however, rats had significant liver injury after cotreatrnent with 50, 100 or 300 mg/kg SLD plus LPS (Fig. 2.2). Fifty mg/kg was chosen as the SLD dose for further study. In a time course study, SLD or LPS given alone did not increase serum ALT activity at any time examined (Fig. 2.3A). In the SLD/LPS-cotreated group, ALT activity remained normal for 4 hr but began to increase by 8 hr after the second administration of SLD. By 12 hr, a significant increase in serum ALT activity was observed. By 24 hr, ALT activity had decreased to near normal. The activity of serum AST in rats also reached its peak at 12 hr and showed a pattern similar to ALT activity (Fig. 2.33). At 12 hr, the activities of ALP and GGT as well as the concentrations of total bilirubin and bile acids were also elevated significantly in the sera of SLD/LPS- cotreated rats compared to those of rats treated with SLD or LPS alone (Fig. 2.4). 2.4.2 Histopathological findings Hepatocellular lesions were not found in livers of rats treated with VehNeh, SLDNeh or Veh/LPS (Fig 2.5A, 2.5B and 25C). ln livers of SLD/LPS-cotreated rats, necrotic foci were present in the midzonal regions (Fig. 2.50). These were 50 Fig. 2.2. Sulindac dose-response in the absence and presence of LPS. Rats were treated with various doses of SLD (10, 20, 50, 100 or 300 mglkg, p.o.) or its vehicle (0.5% methyl cellulose) at -16 hr, and food was removed. At -0.5 hr rats received LPS (8.25X105 EUIkg, i.v.) or its vehicle (saline), and 30 min later they were given a second administration (same dose as 1st administration) of SLD. Blood samples were taken at 12 hr after the second administration of SLD, and ALT activity was measured. *significantly different from SLDNeh group at the same dose. P< 0.05, n=3. 2000 - -O— SLDNeh * -O— SLD/LPS 1500 ‘ ALT (UIL) 3 8 500 ‘ . . . . 4 O 50 100 150 200 250 300 SLD Dose (mglkg) 51 Fig. 2.3. Development of hepatocellular injury induced by SLD/LPS cotreatrnent. Rats were treated with SLD (50 mglkg, p.o.) or its vehicle (0.5% methyl cellulose) and LPS or its vehicle as described in Fig. 2.2. Blood samples were taken at various times (0, 1, 2, 4, 8, 12 or 24 hr), and ALT (A) and AST (B) activities in serum were measured. *significantly different from all other groups at the same time. #significantly different from SLD/LPS group at 0 hr. P<0.05, n=5- 10. 52 > —O— VehNeh . *# —o— SLDNeh 2000 —v— Veh/LPS A —¢v- SLD/LPS E |-— . _' 1000 < A 500 . 0 -\/‘\ _“—U——\I——————_—\r 0 4 8 12 16 20 24 Hr after 2nd Sulindac Administration * + VehNeh # —o— SLDNeh 200° ‘ + Veh/LPS —A— SLD/LPS 51500 - ‘ a |— . (D 1000 ‘ < 500 ‘ —— \ 1 ‘ I ~ ,. 7 ..h._. \ \ ’ o T V t 0 4 8 12 16 20 24 Hr after 2"d Sulindac Administration 53 Fig. 2.4. Markers of hepatic cholestasis induced by SLD/LPS cotreatrnent. Rats were treated as described in Fig. 2.3, and serum samples were collected at 12 hr. Activities of alkaline phosphatase (ALP) and v-glutamyltransferase (GGT) as well as concentrations of total bilirubin and bile acids in serum at 12 hr were evaluated. *significantly different from Veh/LPS group. #significantly different from SLDNeh group. asignificantly different from VehNeh group. p<0.05, n=4- 11. m "m l - ,l 8 I7- I: ,l||ll m I m “m .mMHU w m .m. m m w m o m m m .m. o :23 mEo< BE .53. 35v n_._< D *.I ..ll- . s — Veh Veh m m 1g III mlamwwsm w m m w o 33:00 :23 =5§=m .33 55 Fig. 2.5. Liver histopathology in SLDILPS-cotreated rats. Rats were treated with VehNeh (A), SLDNeh (B), Veh/LPS (C) or SLD/LPS (D) as described in Fig. 2.3. Liver sections were collected at 12 hr and were stained with hematoxylin and eosin. The pictures were taken under 200 X maginification, and a necrotic area is indicated with an arrow. ' I . '. ..‘. 1‘;° 1 I one. associated with hemorrhage and neutrophil infiltration. The numbers and sizes of necrotic foci in livers from SLD/LPS-treated rats progressed with time and were consistent with the elevated ALT and AST activities in rat serum (Table 2.1). Significant liver lesions were observed beginning at 8 hr after the second administration of SLD and persisted through 24 hr. 2.4.3 Effect of SLD/LPS cotreatrnent on serum TNFa concentration SLD alone had no effect on the concentration of TNFa in serum (Table 2.2). Treatment with LPS alone caused a significant increase in the serum TNFo concentration within 1.5 hr. SLD significantly enhanced the increase in serum TNFa induced by LPS. 2.4.4 Activation of the hemostatic system Thrombin functions as a vital activator of the coagulation system, whereas active PAl-1 is the major endogenous down-regulator of the fibrinolytic system. The concentrations of these two regulators were evaluated using ELISA at 1, 4 and 8 hr after the second administration of SLD. Both TAT and active PAl-1 concentrations were elevated by LPS alone at 1, 4 and 8 hr, whereas SLD was without effect by itself (Fig. 2.6). At 4 hr after the second administration of SLD, SLD significantly enhanced the LPS-induced increases in TAT and active PAI-1 in plasma. Compared to LPS alone, SLD/LPS cotreatrnent also tended to prolong the elevation in thrombin and active PAl-1 in plasma (Fig. 2.6A, 2.6B). 57 Table 2.1. Midzonal hepatic necrosis in livers of rats treated with SLD/LPS Liver sections from rats killed at 4, 8, 12 and 24 hr after the second administration of SLD were evaluated and assigned a score of 0-5 as described under Materials and Methods. Data are expressed as median score and 25th and 75th quartiles. *significantly different from VehNeh group at the same time. P<0.05, n=5-10. Treatment Time after 2nd SLD (hr) 4 8 12 24 VehNeh 0.50 (0.34-0.53) 0.50 (0.31-0.66) 0.44 (0.25-0.63) 0.63 (0.47-0.66) SLDNeh 0.38 (0.34-0.88) 0.88 (047-103) 0.75 (0.63-1.00) 0.75 (0.44-0.88) Veh/LPS 0.75 (047-097) 0.75 (0.56-1.19) 0.63 (0.44-1.09) 1.00 (0.81-1.26) SLD/LPS 0.88 (0.72-1.40) 1.88 (1 .63-359)" 58 3.37 (2.25-3.88)* 2.88 (2.25-3.38)‘ Table 2.2. Serum TNFa concentration in SLDILPS-treated Rats. Rats were treated with SLD (50 mglkg, p.o.) or its vehicle (0.5% methyl cellulose) at -16 hr, and food was removed. At -0.5 hr rats received LPS (8.25X105 EUIkg, i.v.) or its vehicle (saline), and 30 min later they were given a second administration (50 mglkg) of SLD. Serum samples were collected at 1 hr after the second administration of SLD. The concentration of TNFo (ng/mL) in serum was determined using ELISA. *significantly different from corresponding group not treated with LPS. #significantly different from Veh/LPS group. P<0.05, n=3-7. Treatment Veh SLD Veh 0.024 :1: 0.005 0.057 i 0.018 LPS 16.9 e 6.5* 50.2 s 10.7“ 59 Fig. 2.6. Activation of the hemostatic system. Rats were treated with SLD, LPS or their vehicles as described in Fig.2.3. They were killed 2, 4 or 8 hr after the second administration of SLD and plasma was collected. Concentrations of TAT (A) and active PAl-1 (B) in plasma were measured. *significantly different from VehNeh group at the same time. #significantly different from Veh/LPS group at the same time. P<0.05, n=3-5. 60 Active PAl-1 (nglmL) “3' TAT (ngImL) N 8 — VehNeh 1:21 SLDNeh — Veh/LPS 1:1 SLD/LPS 100 . * o . .l—A-l-l 2 4 8 Hr after 2"" SLD Administration 1000 ‘ — Veh/Veh El SLD/Veh goo . - Veh/LPS *# * :1 SLD/LPS I 600 1 * * 40° ‘ * L * 2m 4 0 . . . 2 4 8 Hr after 2'"I SLD Adrrl'nistration 61 Hepatic fibrin deposition was evaluated 4 hr after the second administration of SLD because that is a time just before the onset of liver injury as reflected by serum ALT and AST activities (Fig. 2.3). SLD alone and LPS alone were associated with small increases in fibrin that were not statistically significant. In contrast, SLD/LPS cotreatrnent led to a pronounced elevation in fibrin staining that was panlobular in distribution (Fig. 2.7 and 2.9A). As mentioned above, fibrin deposition can lead to hypoxia, which was evaluated by quantifying PIM-protein adducts in livers. PIM-protein adducts were at control levels in livers of SLD-treated rats. LPS treatment caused a modest increase, and SLD/LPS cotreatrnent led to a greater elevation of PIM-protein adducts, predominantly in midzonal regions of liver lobules by 4 hr after the second administration of SLD (Fig. 2.7 and 2.8B). 2.4.5 Effect of heparin on liver injury induced by SLD/LPS cotreatrnent Anticoagulant heparin (3000 Units/kg, s.c.), administered concurrently with the second administration of SLD, caused a marked decrease in fibrin deposition in liver as expected (Fig. 2.10A). PIM-protein adduct staining was also significantly reduced by heparin at 4 hr after the second SLD administration (Fig. 2.10B). To evaluate the role of hemostatic system in liver injury, heparin was given to rats concurrently with and 6 h after the second administration of SLD, and liver injury was evaluated at 12 hr. Heparin was without effect on ALT activity in VehNeh-treated rats but significantly attenuated the increase in ALT activity in 62 Fig. 2.7 Fibrin deposition in liver. Rats treated with VehNeh (A), SLDNeh (B), Veh/LPS (C) or SLD/LPS (D) as described in Fig. 2.4 were killed at 4 hr, and immunohistochemistry for fibrin was performed. 63 Fig. 2.8. Hypoxia staining in liver. Rats were treated with VehNeh (A), SLDNeh (B), Veh/LPS (C) or SLD/LPS (D) as described in Fig. 2.4 except that PIM hydrochloride (120 mg/kg) was administered 2 hr after the second administration of SLD. At 4 hr after the second administration of SLD, livers were collected and fixed for immunohistochemistry. 3A 8 100m of Fig. 2.9. Evaluation of fibrin deposition and hypoxia in liver. Rats were treated as described in Fig. 2.3. A) Animals were killed at 4 hr, and livers were processed for immunohistochemical determination of fibrin deposition. B) Rats received an additional treatment with PIM hydrochloride (120 mglkg) 2 hr after the second administration of SLD. They were then killed at 4 hrs, and livers were processed for immunohistochemical determination of hypoxia. In both panels the fraction of positive pixels averaged from 10 randomly chosen microscope fields (100 X) was determined for each animal. *slgnificantly different from Veh/LPS group. #significantly different from SLDNeh group. asignificantly different from VehNeh group. P<0.05, n=4-6. 65 i —Veh HELPS e. s u. e. m 0 o o 0 0 3.9.3 e>Eoon *0 .5362“: 55E Veh h S e P V L E 5m 5 m 5 0 1 1 0 0 0 D 0 n” 0 0 0 0 3.92.. 9:32. no .5568": £2.25 £39.95...“— SLD Veh 66 Fig. 2.10. Effect of heparin on liver fibrin and hypoxia. SLD and/or LPS were given to rats as described in Fig. 2.3. Heparin (3000 units/kg, so.) was given to rats at the same time as the second administration of SLD. Rats were killed at 4hr, and liver sections were fixed and stained immunohistochemically for fibrin (A) or PIM-protein adducts (B). The fraction of positive pixels was determined as described under Materials and Methods. *significantly different from VehNehNeh group. #significantly different from SLD/LPSNeh group. a significantly different from VehNeh/heparin group. P<0.05, n=4-6. 67 -Veh 1:3l-Iepan'n m o A m. 0 3.9.3 6552. he ESE m o cows—En: 6. 0 Veh Q03} e m .. .. s m m Q Q Q Q Q 3.3.3 02:3“. *0 ceases": 36328 5399.25 m 68 the sera of SLDILPS-treated rats (Fig. 2.11A). The SLD/LPS-induced elevation in total bile acid concentration in serum was also attenuated by heparin (Fig. 2.11B). Necrotic foci were observed in livers of rats treated with SLD/LPSNeh, but not in livers of rats treated with SLD/LPSIheparin (Fig. 2.12). 2.4.6 Effect of low oxygen on hepatocytotoxicity induced by SLD sulfide in vitro To assess whether hypoxia can affect the killing of hepatocytes by SLD, the active metabolite of SLD, SLD sulfide (60 uM) was administered to primary rat hepatocytes in vitro. Immediately after treatment, hepatocytes were incubated in oxygen replete (20% 02) or hypoxic (5% 02) atmospheres. The 5% 02 is equivalent to a nominal p02 of 30 mm Hg, which is the smallest reported oxygen concentration in blood around hepatic central vein in vivo (Jungerrnann and Kietzmann, 2000). This degree of hypoxia had no effect on the viability of hepatocytes after 8 hr of incubation. SLD sulfide alone increased ALT activity in the medium, indicating hepatocellular injury. When HPCs treated with SLD sulfide were exposed to 5% 02, cell death caused by SLD sulfide was significantly enhanced compared to incubation in 20% 02 (Fig. 2.13). 69 Fig. 2.11. Effect of heparin on SLDILPS-induced liver injury. Rats were treated with SLD/LPS as described in Fig. 2.3. Heparin (3000 units/kg, so) or its vehicle (saline) was given to rats at 0 and 6 h. Rats were killed at 12 hr, and ALT activity (A) and concentration of total bile acids (B) in serum was determined. *significantly different from VehNeh/heparin group. #signiflcantly different from SLDILPSNeh group. P<0.05, n=3-13. 7O ALT (UIL) 1200 4 1000 - 800 - 600 - 400 ‘ 200 - 3 Total Bile Acid (u_|\[l) O 3 3588 * - Veh [:3 Heparin # A [-3 F-Tl VehNeh SLD/LPS * _ Veh 1:3 Heparin i_jl:l Veh 71 Fig. 2.12. Effect of heparin on hepatic lesions induced by SLDILPS. Rats were treated with SLD/LPS as described in Fig. 2.4. Heparin (3000 units/kg, so.) or its vehicle (saline) was given to rats at 0 and 6 h. Liver sections were collected at 12 hr and stained with hematoxylin and eosin. Liver sections of rats treated with SLDILPSNeh (A) and SLD/LPS/heparin (B) were examined under 200X magnification. A necrotic area is indicated with an arrow. 72 Fig. 2.13. Effect of hypoxia on SLD sulfide-induced cytotoxicity. Rat primary hepatocytes were isolated as described under Materials and Methods. They were treated with 60uM SLD sulfide and kept in 20% or 5% oxygen. After 8 hr, ALT release was determined. *significantly different from Veh group at the same oxygen level. #significantly different from SLD sulfide treatment group at 20% oxygen. P<0.05, n=3. — 20%02 Veh SLD Sulfide 73 2.5 Discussion SLD is an NSAID that is used for the treatment of arthritis. All of the NSAle have been associated with hepatic IADRs in patients (O‘Connor et al., 2003). The average risk of serious hepatic injury for NSAle is approximately 1 case in 10,000 patient-years of use, and the risk from SLD is reported to be 5-10 fold greater than for NSAle as a class (Walker, 1997). The mechanism of NSAID- induced hepatic IADRs is not clearly understood, and several hypotheses have been proposed. It is commonly accepted that large amounts of active metabolites form after exposure to large doses of NSAIDs, and these might lead to protein adduct formation, oxidative stress and mitochondrial injury that could contribute to tissue damage (Boelsterli, 2002). However, evidence supporting these hypothses is mainly obtained from studies in vitro, and animal models of SLD- induced liver injury are lacking. Previous studies in rodents suggested that there may be a connection between inflammation and hepatic IADRs for at least some drugs (Buchweitz et al., 2002; Luyendyk et al., 2003; Roth et al., 2003; Shaw et al., 2007). In these drug-LPS interaction models, one administration of the drug was sufficient to produce a hepatotoxic interaction with LPS. In preliminary studies with SLD, we tried several single-administration regimens using various doses and times between SLD and LPS treatment. Although hepatotoxic signals (ie, increased serum ALT activity) were observed in some rats, these were inconsistent. Changing to a two-administration protocol in which the time between SLD administrations approximated 3 half lives (Hucker et al., 1973) provided relatively 74 consistent and statistically significant liver injury; moreover, this protocol corresponds to the twice per day treatment regimen typically used therapeutically in human patients. Using this protocol, SLD or LPS alone did not cause any lesions in the liver or increase the clinical chemical biomarkers of liver injury. However, in SLD/LPS-cotreated rats serum markers of both hepatocellular injury and cholestasis were increased significantly, and foci of necrotic parenchymal cells were found in livers. Interestingly, in a study of 91 cases reported to the US. Food and Drug Administration, SLD caused hepatocellular injury, cholestatic injury or mixed liver injury in human patients (Tarazi et al., 1993), consistent with observations presented here for rats treated with SLD/LPS. These results raise the possibility that a mild episode of inflammation might render human patients susceptible to SLD-induced liver injury. Judging by the timecourse of serum ALT and AST activities (Fig. 2.3) and histological changes (Table 2.1), the onset of liver injury was between 4 and 8 hr. The serum markers of hepatocellular injury rose until 12 hr and declined by 24 hr. Despite this decline, histologic evidence of liver injury persisted at 24 hr (Table 2.1). This decline in serum transaminases suggests that injury occurred in the first 12 hrs, and ALT and AST released from hepatocytes were cleared from the blood thereafter. The short initial half life of ALT/AST in rat plasma (approximately 5 hr) is consistent with this interpretation (Saheki et al., 1990). LPS has the potential to influence toxicity in a number of ways. For example, it is capable of downregulating the expression of several drug metabolizing enzymes (Morgan, 1989). SLD is bioactivated by hepatocytes to a 75 more toxic sulfide metabolite by methionine sulfoxide reductase (Kitamura et al., 1980; Kitamura and Tatsumi, 1982; Etienne et al., 2003a). It is not known if LPS downregulates the expression of this enzyme; however, in a preliminary study, LPS treatment did not increase the liver concentration of SLD sulfide (unpublished observation). Cytokines such as tumor necrosis factor-alpha (T NFo), interleukin-1 (IL-1) and interleukin-6 (IL-6) are upregulated after the activation of Toll-like receptor 4 on Kupffer cells by LPS (Luster et al., 1994; Su, 2002). Some of these changes have been linked to liver injury from drug/LPS interaction (Shaw et al., 2007; Tukov et al., 2007b). The observation that fibrin deposited in livers of rats cotreated with SLD and LPS led us to explore the hemostatic system in this study. Cytokines are potent modulators of the hemostatic system. For example, TNFd and lL-1 activate coagulation by upregulating tissue factor expression and by reducing the fibrinolytic activity of endothelial cells through an increase in PAI- 1 (Schleef et al., 1988; Salgado et al., 1994). The plasma concentrations of TAT and active PAl-1 increased rapidly in LPS-treated rats, confirming that LPS induces the activation of the coagulation system and provides conditions for inhibition of the fibrinolytic system (Fig. 2.6). Although SLD had no effect on these factors when it was given alone, it enhanced the LPS-mediated changes and tended to prolong the activation of the hemostatic system. This could be due to enhanced TNFa release by SLD (Table 2.2). In the liver injury induced by ranitidine/LPS cotreatrnent, ranitidine enhanced the LPS-induced increase in TNFa concentration through p38-dependent activation of TNFo converting 76 enzyme (T ACE), which cleaves membrane-bound pro-TNFd to form mature TNFa (Deng et al., 2008). Whether a similar mechanism is at play in SLD/LPS- cotreated rats is yet to be determined. SLD/LPS cotreatrnent significantly increased cross-linked fibrin in livers at 4 hr— Le, a time before the onset of liver injury. Although LPS activated the hemostatic system in rats, it alone was not sufficient to induce marked fibrin deposition in the liver (Fig. 2.9A). The hemostatic system is activated in endotoxemia-induced liver injury (Hewett and Roth, 1995), and one possible consequence is hypoxia in the liver resulting from disrupted blood flow in the sinusoids. PIM-protein adducts were slightly elevated in livers of rats treated with LPS alone, indicating that mild hypoxia occurred. In contrast, SLD/LPS led to a pronounced increase in PIM-protein adducts, suggesting marked hypoxia (Fig. 2.93). Unlike the distribution of fibrin which was panlobular, hypoxia occurred only in the midzonal regions of livers (Fig. 2.3, 2.4). Interestingly, necrotic foci were also present predominantly in this region (Fig.2.5). It has been reported that hypoxia causes hepatocelluar injury in isolated, perfused rat livers (Lemasters et al., 1981), and liver injury was induced in vivo in rats exposed for a brief period to a low concentration of oxygen (Fassoulaki et al., 1984). In studies presented here, the anticoagulant heparin reduced hepatic fibrin deposition and hypoxia induced by SLD/LPS cotreatrnent (Fig. 2.10). This suggests that hypoxia might be caused by fibrin clots in liver sinusoids. Moreover, both hepatocellular and bile ductular injury in SLD/LPS-treated rats 77 was significantly attenuated by heparin, supporting the hypothesis that hypoxia induced by fibrin clots plays an important role in the pathogenesis (Fig. 2.11). Previous studies suggested that hypoxia can potentiate the toxicity of some xenobiotics towards hepatocytes (Shen et al., 1982). Compared to SLD or its sulfone metabolite, SLD sulfide is more cytotoxic (Leite et al., 2006). Therefore, we treated rat primary hepatocytes with SLD sulfide. Hypoxia enhanced its ability to kill these cells (Fig. 2.13). SLD sulfide, but not SLD or SLD sulfone uncouples the mitochondria of HepGZ cells (Leite et al., 2006). Thus, hypoxia could exacerbate SLD sulfide-mediated mitochondrial dysfunction by diminishing the aerobic metabolism of hepatocytes. Moreover, hypoxia inducible factor 1 (HIF-1) accumulated in cells under hypoxic stress might induce the expression of proapoptotic proteins and cause the stabilization of p53, which increases permeability of the mitochondrial membrane and causes cell death (Greijer and van der Wall, 2004). In the SLD/LPS idiosyncratic liver injury model, hypoxia might enhance the mitochondrial toxicity of SLD, providing a synergistic effect on a mitochondrial pathway to cell death. Although further study is required to test this, SLD/LPS cotreatrnent in rats could be an animal model supporting the mitochondrial injury hypothesis of NSAID-induced IADRs described above. The mitochondrial pathway might not be the only contributor to liver injury. Hypoxia can also interplay with inflammatory factors. For example, when stimulated by LPS, neutrophils accumulate in sinusoids and transmigrate through blood vessels to liver parenchyma. At the site of inflammation, activated neutrophils release proteases that can kill rat hepatocytes (Ho et al., 1996), and 78 hypoxia enhanced neutrophil protease-mediated hepatocyte killing (Luyendyk et al,2005) Results with SLD are similar to those observed with diclofenac (DCLF), another NSAID that causes hepatic IADRs in people, in the sense that both drugs interacted with LPS to cause liver injury in rats. However, a large dose of DCLF (100 mglkg) caused liver injury by itself in rats in the absence of LPS- cotreatrnent. This was associated with intestinal injury and translocation of bacteria to the liver and was prevented by sterilization of the GI tract (Deng et al., 2006). These results suggested that GI irritation and translocation of bacteria or LPS caused by DCLF contribute to DCLF-induced hepatotoxicity in rats, and a similar mechanism might underlie DCLF IADRs in humans. In contrast, large doses of SLD (up to 300 mglkg) did not cause liver injury in rats by themselves (Fig. 2.2). This suggests that, at least in rats, SLD is less irritating to the intestine than DCLF, although the relative gastrointestinal toxicity of SLD and DCLF in humans is still controversial. One study indicated that SLD is less associated with gastrointestinal hospitalizations than DCLF (Garcia Rodriguez et al., 1992), whereas others suggested the opposite (Henry et al., 1993; Savage et al., 1993). The potential implication for human IADRs is that DCLF may be able to provide its own inflammatory stress through GI irritation, whereas SLD toxicity might require an additional inflammatory stress that arises independently of drug treatment. It seems possible that the latter could arise from the very condition that the drug is used to treat- e.g., an inflammatory flare of rheumatoid arthritis. Alternatively, an independently occurring inflammatory episode might interact 79 with SLD. The finding that viral hepatitis may predispose patients to NSAID hepatotoxicity supports this possibility (Tech and Farrell, 2003). In summary, SLD/LPS cotreatment caused liver injury in rats, which was not produced by SLD or LPS alone. The coagulation system was activated while the fibrinolytic system was inhibited in the cotreated rats. As a consequence, fibrin clots formed in sinusoids and hypoxia occurred selectively in livers of rats treated with SLD/LPS. The anticoagulant heparin protected rats against liver injury and also attenuated fibrin deposition and liver hypoxia. Hypoxia enhanced the cytotoxicity of SLD sulfide in vitro. The results support the hypothesis that NSAle that cause hepatic IADRs in humans interact with an inflammatory stress to cause liver injury in animals. Hypoxia may play an important role in this SLD/LPS idiosyncratic liver injury model through synergistic interplay with the toxic SLD sulfide metabolite. These observations do not exclude a role for other factors, such as cytokines, that are a focus of ongoing investigation. 80 CHAPTER 3 Zou W, Beggs KM, Sparkenbaugh EM, Jones AD, Younis HS, Roth RA and Ganey PE (2009). Sulindac metabolism and synergy with TNF in a drug- inflammation interaction model of idiosyncratic liVer injury. J Pharmacol Exp Ther. 81 3.1 Abstract Sulindac (SLD) is a nonsteroidal anti-inflammatory drug (NSAID) that has been associated with a greater incidence of idiosyncratic hepatotoxicity in human patients than other NSAIDs. In previous studies, cotreatrnent of rats with SLD and a modestly inflammatory dose of lipopolysaccharide (LPS) led to liver injury, whereas neither SLD nor LPS alone caused liver damage. In studies presented here, further investigation of this animal model revealed that the concentration of tumor necrosis factor-a (TNF) in plasma was significantly increased by LPS at 1 hr, and SLD enhanced this response. Etanercept, a soluble TNF receptor, reduced SLD/LPS-induced liver injury, suggesting a role for TNF. SLD metabolites in plasma and liver were determined by LC/MSIMS. Cotreatment with LPS did not increase the concentrations of SLD or its metabolites, excluding the possibility that LPS contributed to liver injury through enhanced exposure to SLD or its metabolites. The cytotoxicities of SLD and its sulfide and sulfone metabolites were compared in primary rat hepatocytes and HepG2 cells; SLD sulfide was more toxic in both types of cells than SLD or SLD sulfone. TNF augmented the cytotoxicity of SLD sulfide in primary hepatocytes and HepG2 cells. These results suggest that TNF can enhance SLD sulfide-induced hepatotoxicity, thereby contributing to liver injury in SLD/LPS-cotreated rats. 82 3.2 Introduction Several hypotheses have been put forward to explain the basis for IADRs; however, the modes of action are still unclear, in part because of the lack of animal models. One hypothesis is that inflammatory stress precipitates hepatic IADRs in humans (Roth et al., 2003; Ganey et al., 2004). In concert with this hypothesis, cotreatrnent of rats with lipopolysaccharide (LPS), which induces modest inflammation, and SLD resulted in liver necrosis, whereas neither LPS nor SLD was hepatotoxic alone (Zou et al., 2009b). In this study, we examined factors that could contribute to the pathogenesis of liver injury in rats cotreated with LPS and SLD. In vivo, SLD can be metabolized either irreversibly to SLD sulfone or reversibly to SLD sulfide, which is more cytotoxic than SLD itself. Since LPS can regulate drug metabolism (Renton, 2001), we tested whether LPS coexposure enhances bioactivation of SLD. Moreover, we determined the effect of SLD on LPS-induced tumor necrosis factor-a (T NF) production and its role in the development of liver injury. 83 3.3 Materials and methods 3.31 Materials Unless otherwise noted, all chemicals were purchased from Sigma-Aldrich (St. Louis, MO). LPS (Lot 075K4038) derived from Escherichia coli serotype 055235 with an activity of 3.3 X 106 endotoxin units (EU)/mg was used in experiments. Etanercept was purchased from Amgen Pharmaceuticals (Thousand Oaks, CA). HepGZ/C3A cells for in vitro studies were obtained from American Type Culture Collection (Manassas, VA). 3.3.2 Animals Male, Sprague-Dawley rats (Crl:CD(SD)lGS BR; Charles River, Portage, MI) weighing 250 to 370 g were used for studies in vivo (rats weighing 290 to 3009 were used to evaluate SLD and its metabolites in GI and feces), and rats weighing 150 to 200 g were used for primary hepatocyte isolation. Animals were fed standard chow (Rodent Chow/Tek 8640; Harlan Teklad, Madison, WI) and allowed access to spring water. They were allowed to acclimate for 1 week in a 12-hr light/dark cycle prior to use in experiments. 3.3.3 Experimental protocol As described in previous studies (Zou et al., 2009b), rats were given two administrations of SLD (50 mglkg, p.o.) or its vehicle (0.5% methyl cellulose) with a 16 hr interval, and food was removed after the first administration. Half an hour before the second administration of SLD, LPS (8.25X 105 EU/kg, i.v.) or its 84 vehicle (saline) was administered via a tail vein. Depending on the purpose of experiments, rats were anesthetized with isoflurane and euthanized at various times (0, 1, 2, 4, 8 and 12 hr) after the second administration of SLD. For the collection of plasma, a portion of blood drawn from anesthetized rats was transferred into vacutainer tubes (Becton Dickinson, Franklin Lakes, NJ) containing sodium citrate (final concentration 0.38%). The rest of the blood was allowed to clot at room temperature for preparation of serum. Collected plasma and serum were stored at — 80 0C until use. Three slices (3-4 mm thick) of the left lateral liver lobe were collected and fixed in 10% buffered formalin for histological analysis. A portion of the right medial lobe of the liver was flash-frozen in liquid nitrogen for pharrnacokinetic study of SLD and its metabolites. For determining drug concentration in the gastrointestinal (GI) tract and feces, each rat was housed in a separate cage after LPS or vehicle injection and euthanized at 2 hr. The entire GI tract and its contents were collected. Feces were retrieved from the cages and were homogenized with the GI tract and its contents for each rat. In the TNF inhibition study, rats were given etanercept (8mgfltg) or vehicle (sterile water) subcutaneously an hour before LPS (8.25X 105 EU/kg, i.v.) or its saline vehicle. We have demonstrated that etanercept inactivates TNF activity induced by LPS administration in rats using this treatment protocol (Tukov et al., 2007b). 3.3.4 Evaluation of liver injury The activity of alanine aminotransferase (ALT), a marker of hepatic parenchymal cell injury, was assessed in serum using a diagnostic kit from 85 Thermo Corp. (Waltham, MA). Liver slices fixed in 10% buffered formalin were embedded in paraffin, cut into 6 um sections and stained with hematoxylin and eosin (H&E) for histological evaluation. 3.3.5 Determination of TNF concentration in serum The concentration of TNF in serum collected at 0, 1, 2, 4 and 8 hr after the second administration of SLD was measured by ELISA (BD Biosciences; San Diego, CA). 3.3.6 LCIMSIMS analysis Plasma samples, liver homogenates, GI and fecal homogenates or HepGZ cells in culture medium were mixed with acetonitrile containing diclofenac as internal standard. After vortex and centrifugation, protein was removed, and the supernatant was diluted and transferred to ultra performance liquid chromatography (UPLC) sample vials for LC/MS/MS analysis. LC/MS/MS analysis was performed using a Waters ACQUITY UPLC System coupled to a Quattro Premier XE tandem quadrupole mass spectrometer. An extract volume of 2 uL was injected into the UPLC system and eluted with a gradient mixture (0-99%) of formic acid and acetonitrile. Electrospray ionization in positive ion mode was performed for analyses of plasma samples, and the collision and source cone voltages were optimized independently for each analyte. Multiple reaction monitoring (MRM) of the following m/z transitions was used for the quantitative analysis of diclofenac (296.2->214.2), SLD (357.2- 86 >333.2), SLD sulfone (373.2->233.2), SLD sulfide (341.2->234.2), SLD acyl glucuronide (533.1->339.1), SLD sulfone acyl glucuronide (549.1->355.1) and SLD sulfide acyl glucuronide (517.1->323.1). For samples other than plasma, electrospray ionization was performed in negative ion mode, and metabolite concentrations were determined by the MRM of transition of diclofenac (294.2->250.0), SLD (311.2->296.2), SLD sulfone (327.2->264.2), SLD sulfide (295.2->280.2), SLD acyl glucuronide (531.1- >355.1), SLD sulfone acyl glucuronide (547.1->371.1) and SLD sulfide acyl glucuronide (515.1->339.1). The LCIMS/MS method achieved low limits of quantification (LLOQ) of 30 ng/mL or less for all three forms of sulindac (sulfoxide, sulfide, and sulfone) using both positive and negative ion modes. Analytical reproducibility was judged to be :I: 12% in the middle of the calibrated range of concentrations. 3.3.7 Evaluation of cytotoxicity of SLD and its metabolites in vitro HepGZ cells were plated at a density of 4 x 104 cells/well in 96-well plates. After overnight incubation in Dulbecco's modified Eagle’s medium (DMEM) with 10% fetal bovine serum (FBS), medium was renewed and SLD, SLD sulfone, SLD sulfide (0- 500 (M) or its vehicle (0.5% dimethyl sulfoxide [DMSO]) was added to the wells. After a 24-hr incubation, lactate dehydrogenase (LDH) released into the medium and total cellular LDH were evaluated using a kit from Promega Corporation (Madison, WI). Cytotoxicity was assessed as the 87 percentage of LDH released into the medium relative to the total LDH in the well (medium plus lysed cells). For primary rat hepatocytes, isolation was performed as described previously (Tukov et al., 2006). Briefly, rat liver was first perfused in situ through the portal vein and then digested with Liver Digest Medium (lnvitrogen Corp, Carlsbad, CA). The digested liver was combed gently, and hepatocytes were obtained after centrifugation (100 x g, 30 s). Hepatocytes were suspended in Williams’ Medium E (lnvitrogen Corp, Carlsbad, CA) with 10% FBS, and the cell viability was always above 80%. Hepatocytes were plated at a density of 2.5 x 105 cells/well in 12-well plates and incubated for 3 hr to attach to the plate. Serum-containing medium was replaced by serum-free medium, and SLD, SLD sulfone, SLD sulfide (0- 120 (M) or vehicle (0.1% DMSO) was added to the culture wells. After 8 hr incubation, cytotoxicity was assessed by calculating the ALT activity in the medium plus unattached cells as a percentage of the total ALT activity in the well as described previously (Zou et al., 2009b). 3.3.8 Cytotoxicity from TNF and SLD metabolites SLD sulfide or its vehicle (0.5% DMSO) was administrated to HepG2 cells with recombinant human TNF (200 ng/mL) or its vehicle (medium). After 24 hr incubation, the percentage of LDH released was evaluated. To determine the remaining concentration of SLD sulfide in each well, HepG2 cells were scraped, 88 and acetonitrile was added to precipitate protein. After centrifugation, the concentration of SLD sulfide in supernatant was determined using LC/MS/MS. To assess further whether TNF can affect the cytotoxicity of SLD metabolites, isolated primary rat hepatocytes were treated with SLD sulfide (60 uM) in the presence or absence of recombinant rat TNF (2 pg/mL), and the percentage of ALT released was evaluated 8 hr later. 3.3.9 Statistical analysis Results are expressed as means 1 SEM. One-way or two-way analysis of variance (ANOVA) was applied for data analysis as appropriate, and Tukey’s test was employed as a post hoc test. Student’s t-test was performed when only two groups were compared. For all studies, P < 0.05 was considered as the criterion for statistical significance. 89 3.4 Results 3.4.1 Timecourse of TNF concentration in plasma Rats were treated with LPS and two administrations of SLD or their vehicles as described in Methods, and TNF concentration in serum was evaluated at various times up to 8 hr after the second administration of SLD. SLD had no effect on serum TNF concentration in rats. LPS alone led to a significant increase in TNF serum concentration at 0 and 1 hr (ie, 0.5 and 1.5 hr after LPS). The elevation of TNF concentration induced by LPS was significantly increased by SLD at 1 hr after the second administration of the drug (Fig. 3.1). 3.4.2 Effect of TNF inhibition on liver injury Etanercept is a soluble TNF receptor that neutralizes the biological activity of TNF. To investigate the role of TNF in liver injury, rats were treated with etanercept 1 hr before LPS administration. This treatment protocol inhibits the activity of TNF in rats (Geier et al., 2003). We have reported previously that neither LPS nor SLD produces liver injury when given alone at the doses used in these studies (Zou et al., 2009b). Also consistent with our previous report, SLD/LPS cotreatrnent increased serum ALT activity significantly (Fig. 3.2). Etanercept significantly attenuated this increase, whereas etanercept alone had no effect on serum ALT activity. Histological examination of H&E-stained livers of rats revealed a pattern consistent with the ALT activity. That is, midzonal necrotic foci were present in livers of rats treated with SLD/LPS but were found infrequently in livers of rats treated with etanercept/SLDILPS. 90 Fig. 3 with fl cellulo SLD. L vein. T 2. 4 or VehNe the sar SLD/L TNFa (pg/mL) Fig. 3.1. Timecourse of TNF concentration in rat serum. Rats were treated with two administrations of SLD (50 mglkg, p.o.) or its vehicle (0.5% methyl cellulose) with a 16 hr interval. Half an hour before the second administration of SLD, LPS (8.25X 105 EUIkg, i.v.) or its saline vehicle was administered via a tail vein. TNF was evaluated by ELISA in serum samples obtained from rats at 0, 1, 2, 4 or 8 hr after the second administration of SLD. *significantly different from VehNeh group at the same time. #significantly different from Veh/LPS group at the same time. P<0.05, n=4-5 for all points except 8 hr group (n=3), Veh/LPS and SLD/LPS at 1 hr (n=8 and 9, respectiveIY). and SLD/LPS at 0 hr (n=8). 7°°°° ‘ *# —o— VehNeh -”'- SLDNeh 3 6°°°° T + Veh/LPS E 50000 - - SLD/LPS 8’ ‘5' 40000 ”L *r :: 30000i I- 20000 10000 . 0 Hr after 2nd Sulindac Administration 91 Fig. 3.2. Effect of TNF inhibition on liver injury induced by SLD/LPS. Rats were treated with etanercept (8 mglkg, so.) or its vehicle 1 hr before LPS. SLD and LPS or their vehicles were administered to rats as described in Methods. ALT activity was determined at 12 hr (A). *significantly different from respective VehNeh group. #significantly different from Veh/ SLD/LPS group. P<0.05, n=4 for all groups except SLD/LPS/Etan (n=6). Liver sections from rats treated with VehNehNeh (B), EtanNehNeh (C), Veh/SLDILPS (D), and Etan/SLD/LPS (E) were examined. A necrotic area is indicated with an arrow. 92 > at- zooo — Veh r:1 Etan 3 .— 1000I .J < 500 *# o . 93 Peta: Cecre Vt 3.4.3 Effect of LPS on SLD metabolism in rats SLD and its sulfone and sulfide metabolites were determined in rat plasma at various times after the second administration of SLD. Plasma SLD concentration reached a peak 1 hr after administration and decreased gradually over 8 hr (Fig. 3.3A). ln LPS-treated rats, plasma SLD concentration was significantly smaller. SLD treatment increased SLD sulfone concentration in plasma steadily between 2- 8 hr (Fig. 3.3B). This increase was not observed after SLD/LPS cotreatrnent, so that the plasma concentration of SLD sulfone was significantly less in SLD/LPS-cotreated rats by 8 hr. Plasma SLD sulfide concentration reached a peak within 4 hr in both groups, and LPS administration decreased the SLD sulfide concentration in plasma significantly at 1, 2, 4 and 8 hr compared to that of SLD/vehicle-treated rats (Fig. 3.3C) ln livers of rats treated with SLD alone, the concentrations of SLD and its metabolites showed trends similar to those in plasma. LPS cotreatrnent decreased SLD and SLD sulfide concentrations, but SLD sulfone concentration was unaffected (Fig. 3.4). LPS selectively lowered the SLD concentration in liver at 1 and 2 hr, and decreased SLD sulfide concentration in liver at 2 and 4 hr. To investigate further the effect of LPS on SLD metabolism, rats were euthanized at 2 hr and SLD metabolite concentrations were determined in the GI tract and feces collected between -0.5 and 2 hr. The concentrations of SLD and SLD sulfide in the GI tract and feces were significantly increased by LPS (Fig. 3.5). However, the SLD sulfone concentration was not affected by LPS. 94 Fig. 3.3. Effect of LPS on plasma concentrations of SLD, SLD sulfone and SLD sulfide. Rats were treated with-SLD and with LPS or its saline vehicle as described in Fig. 3.1. They were euthanized, and plasma was collected at 0, 1, 2, 4 and 8 hr after the second administration of SLD. The plasma concentrations of SLD, SLD sulfone or SLD sulfide were determined as described in Methods. *significantly different from SLDNeh group at the same time. P<0.05, n=5 for all groups except SLD/LPS at 4 hr (n=7). 95 SLD (uglmL) SLD sulfone (uglmL) SLD sulfide (uglmL) 200 —o— SLDNeh -O— SLD/LPS 150 100 * * * so 4 * 0 . . 0 2 4 6 8 Hr after 2nd Sulindac Administration 200 1 150 * 100 50 . . . . 0 2 4 6 8 Hr after 2nd Sulindac Administration 200 1 150 . 100 ~ * * * l, * 50 - I 0 0 2 4 6 8 Hr after 2nd Sulindac Administration 96 Fig. 3.4. Effect of LPS on liver concentrations of SLD, SLD sulfone and SLD sulfide. Rats were treated with SLD and with either LPS or its saline vehicle as described in Fig. 3.1. Liver concentrations of SLD, SLD sulfone and SLD sulfide were determined at 0, 1, 2, 4 and 8 hr after the second administration of SLD. *significantly different from SLDNeh group at the same time. P<0.05, n=5 for all groups except SLD/LPS at 4 hr (n=8). 97 > W SLD(ng/mg) «- 8 '8‘ 3’ g s f i E' SLD sulfone (nglmg) s 2 SLD sulfide (nglmg) G 3 ii uh J A 0 2 4 6 8 I-I'aferndeindacAd‘rinish'afion 0 2 4 6 8 Hr after 2nd Sulindac Administration 0 2 4 6 8 Hr after 2nd Sulindac Administration 98 135.00ncr rd feces Rat: tiescnbed in fer—.01. Two I r: fie whole I 6 “om-geniz ?: SLD sulfid #187“. group Fig. 3.5. Concentrations of SLD, SLD sulfone and SLD sulfide in GI tract and feces. Rats were treated with SLD and with either LPS or its saline vehicle as described in Fig. 3.1. Each rat was housed in a different cage after the LPS injection. Two hours after the second administration of SLD, feces in the cage and the whole GI tract and its contents were collected for each rat. The mixture was homogenized with acetonitrile, and the concentrations of SLD, SLD sulfone and SLD sulfide were determined by LC/MS/MS. *significantly different from SLDNeh group. P<0.05, n=4. 99 LPS 6 4 2 0 3:: 3w 1 1 m. n. e s. 1 o o o 3:: one...» Dam 0.00 . LPS Veh 5 7. 0 3.5 222.5 3m 0.50 1 5 2 0 1.00 i 0 0 0 Veh 100 The l SLD sulfa: 3.4.4 Effe SLD at 8 hr at metabolit 3.4.5 Cy1 hepatocy Neitl increase SLD 5qu HM. In cilotcxlc C8” deat The concentrations of acyl glucuronide conjugates of SLD, SLD sulfone and SLD sulfide were below the limit of detection in all of the samples measured. 3.4.4 Effect of etanercept on SLD metabolism in rats SLD and its sutfone and sulfide metabolites were determined in rat plasma at 8 hr after the second administration of SLD. Etanercept had no effect on SLD metabolite concentration in plasma of rats cotreated with SLD/LPS (Table 3.1). 3.4.5 Cytotoxicity of SLD and its metabolites in HepG2 cells and rat primary hepatocytes Neither SLD nor SLD sulfone at concentrations up to 500 pM led to an increase in released LDH when applied to HepG2 cells (Fig. 3.6A). In contrast, SLD sulfide induced significant LDH release at concentrations greater than 125 pM. In rat primary hepatocytes, SLD and SLD sulfone also produced no cytotoxicity at the concentrations examined (Fig. 3.68), but SLD sulfide caused cell death at concentrations as small as 30 uM. The cytotoxicity of SLD sulfide was concentration-dependent, and 120 pM SLD sulfide killed almost all of the hepatocytes. 3.4.6 Effect of TNF on cytotoxicity of SLD and its metabolites in HepG2 and rat primary hepatocytes TNF alone did not affect the release of LDH from HepG2 cells (Fig. 3.7). Neither SLD nor SLD sulfone was cytotoxic in the presence or absence of TNF. 101 Table 3.1. Effect of etanercept on SLD metabolism. Rats were killed and plasma was collected at 8 hr after the second SLD administration. Metabolite concentrations were determined as described under Methods. n= 4-5. Concentration in plasma (uglmL) Treatment SLD SLD sulfone SLD sulfide Veh/SLDILPS 59.6 s 12.2 139.3 s 16.7 62.2 1 12.1 Etan/SLD/LPS 52.8 a: 10.8 153.8 1 13.8 39.8 s 13.3 102 ‘- ..- I v.- Fig. 3.6. Evaluation of cytotoxiclty induced by SLD, SLD sulfone or SLD sulfide. SLD, SLD sulfone or SLD sulfide was administered at various concentrations to HepGZ cells (A). The percentage of LDH released in the medium after 24 hr was determined as a marker of cytotoxicity. (B) Rat primary hepatocytes were treated with SLD, SLD sulfone or SLD sulfide for 8 hr, and the percentage of ALT activity released into medium was determined as described in Methods. *significantly different from vehicle (0 concentration). #significantly different from SLD or SLD sulfone at the same concentration. P<0.05, n=3. 103 > 3 +SLD -O—SLDSquono +SLDSulfldo 3 LDH (% released) 8 20 I J\_ *0 0 100 200 300 400 500 600 Concentration (uM) B 100 ‘ —o— SLD *# *# —O— SLD sulfone A —v— SLD sulfide '5 80 ' 0 g *# _q_; 60‘ f.’ s: 40 '3 < 20 . 0 0 30 60 90 120 Concentration (uM) 104 t3]. Cytotoxicil itrere treated \l agree or absence 371180 after 24 988(1) concentra late same cor Fig. 3.7. Cytotoxicity induced by TNF and SLD or its metabolites. HepGZ cells were treated with SLD (A), SLD sulfone (B) or SLD sulfide (C) in the presence or absence of TNF (200ng/mL). The percentage of LDH released was determined after 24 hr as described in Methods. *significantly different from vehicle (0 concentration). #significantly different from value in the absence of TNF at the same concentration of SLD or metabolite. P<0.05, n=3. 105 8 8 S A LDH (% released) 0 LDH (% released) LDH (% released) 101 + Veh -O- TNFa ._.,// T . . . 0 100 150 200 250 SLD concentration (uM) + Veh -O— TNFa ‘-H//l . . . f 0 100 150 200 250 SLD sulfone concentration (uM) * # + Veh -O- TNFa * *# # 1.1-9C” . . . . 0 100 150 200 250 SLD sulfide concentration (uM) 106 Small cytotoxic a the cytotcr The r 1981). T0 cells. the hr after 5 (54:02 (5.32010 A p0 sulfide in the cultui Significa- were tree Smaller concentrations (150, 200 (M) of SLD sulfide, which were not cytotoxic alone, induced cell death in the presence of TNF. TNF also enhanced the cytotoxicity of a larger concentration of SLD sulfide (250 (M). The conversion of SLD to SLD sulfide is a reversible reaction (Duggan, 1981). To evaluate whether TNF affects metabolism of SLD sulfide in HepG2 cells, the amount of SLD sulfide in medium plus HepG2 cells was measured 24 hr after SLD sulfide application to the cells. This amount in wells with TNF (5.4102 pg) was not significantly different from wells that received vehicle (5.310.109). A potentiating effect of TNF was also observed on the cytotoxicity of SLD sulfide in primary hepatocytes. TNF alone did not affect ALT activity released into the culture medium compared to vehicle treatment. SLD sulfide alone caused significant release of ALT activity into the medium (Fig. 3.8). When hepatocytes were treated with SLD sulfide and TNF together, TNF significantly enhanced the cell injury induced by SLD sulfide. 107 fig.1 hepat prérhaf 08500 In. Fig. 3.8. Effect of TNF on SLD sulfide-induced injury to rat primary hepatocytes. SLD sulfide (60 0M) and/or TNF (2 ug/mL) was administered to rat primary hepatocytes. The percentage of LDH released was determined as described in Methods. *significantly different from corresponding vehicle group. #significantly different from SLD sulfide alone group. P<0.05, n=3. *# 80 - — Veh ALT (% released) Veh SLD Sulfide 108 3.5 Discussion As reported previously (Zou et al., 2009b), SLD/LPS cotreatrnent induced severe liver injury in rats. Pro-inflammatory cytokines, especially TNF, have proved to play a critical role in other drug/LPS-induced liver injury models (Shaw et al., 2007; Tukov et al., 2007b). Moreover, studies suggest that reactive drug metabolites produced in liver are critical for idiosyncratic hepatotoxicity from some drugs (Kaplowitz, 2005). Therefore, we focused in this study on the roles of TNF and the toxic metabolite of SLD as well as their interaction in SLD/LPS- induced liver injury. The concentration of TNF in serum was elevated in rats after exposure to LPS, and SLD significantly enhanced the LPS-mediated increase in TNF as early as 1 hr. Besides SLD, other drugs associated with idiosyncratic hepatotoxicity in humans, such as ranitidine and trovafloxacin, also had a synergistic effect on the LPS-mediated increase in TNF in rodents (Shaw et al., 2007; Tukov et al., 2007b). Sulindac and other NSAIDs enhanced TNF release from LPS-pretreated, macrophage-derived RAW264.7 cells at concentrations achieved clinically in humans (Cho, 2007). These findings suggest that enhancement of serum TNF concentration might be a common characteristic of drugs that induce idiosyncratic liver injury. The source of TNF and the mechanism by which SLD enhances TNF appearance are unknown. After LPS exposure, the increase in plasma TNF concentration is mirrored by elevated liver concentration (Femandez—Martinez et al., 2004). Therefore, the source of TNF after LPS exposure is likely liver. However, the source of enhanced TNF in serum after 109 p'etrei intibtl role fc Stficir fated likely i T Ueatrr SLD 1 exam tract I SLD flora l 9361“ SLD. mGtal SLD-cotreatment is not known. TNF-converting enzyme (T ACE), which is required for release of biologically active TNF, is a possible contributor, since some NSAle can enhance the activity of this enzyme (Gomez-Gaviro et al., 2002). It is also possible that SLD or its metabolites enhances TNF transcription or translation or interferes with TNF clearance. The importance of TNF in SLD/LPS hepatotoxicity was explored by pretreating rats with etanercept, a soluble receptor that neutralizes TNF. TNF inhibition protected against SLD/LPS-induced liver injury, suggesting a critical role for TNF in this model. However, elevation in TNF concentration alone is not sufficient to cause liver damage, since much larger TNF concentrations have failed to induced liver injury (Deng et al., 2008). Thus, additional factors are likely involved in liver toxicity in SLD/LPS-cotreated rats. The requirement for bioactivation of SLD raises the possibility that LPS treatment leads to liver injury in SLD-treated rats by increasing the conversion of SLD to a toxic metabolite. To study the effect of LPS on SLD metabolism, we examined the concentration of SLD and its metabolites in plasma, liver, and GI tract plus feces. According to previous studies, two enzymes are responsible for SLD metabolism; methionine sulfoxide reductase (MSR) in both liver and gut flora reduces SLD to SLD sulfide, and a flavin-containing monooxygenase (FMO) converts SLD to SLD sulfone and also catalyzes the conversion of SLD sulfide to SLD. SLD was maximally absorbed in 1 hr, and SLD as well as its sulfone metabolite accumulated in liver, a result consistent with previous findings (Duggan et al., 1980). LPS can significantly down-regulate the expression of 110 hepatic oonseq (Vattan metabc hr, res; SLD fr omeer signific EXPOSL feces. abscrp The exl Wiser hepatic FMO in mice (Zhang et al., 2008). Oxidative stress, a possible consequence of LPS exposure, can increase the expression of MSR in bacteria (Vattanaviboon et al., 2005). Therefore, LPS might have an effect on shifting the metabolism of SLD towards SLD sulfide by regulating the expression of these two enzymes. However, LPS decreased the concentrations of SLD and SLD sulfide in plasma after the second administration of SLD. The liver concentrations of SLD and SLD sulfide were also decreased by LPS at 1 and 2 hr and 2 and 4 hr, respectively. These results suggested that LPS might decrease absorption of SLD from the GI tract. To address this possibility, we measured metabolite concentrations in the GI tract and feces at 2 hr, a time at which we found a significant decrease in both SLD and SLD sulfide in plasma and liver after LPS exposure (Fig. 3.5). LPS increased the concentration of SLD in the GI tract and feces, suggesting that LPS decreased the bioavailability of SLD by reducing its absorption. This result does not rule out the possibility that LPS has an effect on the expression of enzymes that metabolize SLD. Moreover, the SLD metabolite concentrations in the plasma of cotreated rats were not changed at 8hr by etanercept pretreatment, suggesting that TNF does not play a role in the ability of LPS to reduce SLD absorption. The cytotoxicity of SLD and its metabolites were compared in both HepG2 cells and primary rat hepatocytes. SLD and SLD sulfone were not toxic to HepG2 cells even up to 500 uM, yet SLD sulfide showed significant toxicity. This result is consistent with previous findings, although different medium was used and a different cytotoxicity assay was performed (Leite et al., 2006). It also has been 111 Mddyre (Khieti SLD.H0 qhdcxk sensitive SLD as qmncxi Ah! mn de; essentl. sulfide nhghtl heDatc the IN SUlficle enhan SLD tc S GEVQK SYIIGrg and p widely reported that SLD sulfide can induce apoptosis of other cancer cell lines (Kim et al., 2005; Bock et al., 2007), which raised interest in treating cancer with SLD. However, in this study, we found that the active metabolite of SLD was also cytotoxic to primary hepatocytes and that primary rat hepatocytes were more sensitive than HepG2 cells (Fig. 3.6). This might have implications for the use of SLD as an anticancer agent if normal host cells are more sensitive to the cytotoxic effects of SLD than are cancer cells. Although the mechanisms of drug-induced idiosyncratic liver injury are still not clear, it is believed that accumulation of active metabolites in liver is an essential first step for many drugs (Watkins, 2005). Accordingly, excessive SLD sulfide in liver might be critical for SLD- induced idiosyncratic liver injury. This might be why two administrations of SLD were required in this model to effect hepatotoxicity. Interestingly, LPS decreased the concentration of SLD sulfide in the livers of rats, suggesting that SLD sulfide accumulation alone was not sufficient to induce liver injury, and that LPS might be activating pathways that enhance the toxicity of SLD sulfide, instead of increasing the concentration of SLD toxic metabolite. Since TNF and SLD or its metabolites are both indispensable for the development of SLD/LPS-induced liver injury, we explored whether TNF acted synergistically with SLD or its metabolites using an in vitro system. Both HepG2 and primary rat hepatocytes were resistant to TNF toxicity. Even a much greater concentration of TNF than we used failed to kill HepGZ cells and primary rat hepatocytes (Adamson and Billings, 1992). SLD or SLD sulfone in combination 112 with T SLD E synerg comblr HOWE). liver ll investi- activat particu inhibit. NF-kB Baftim. ‘Iepatc PlOSur effect, Stifide lead t( DTOGUC (Brady tai'élets SI hepatc nil-Dug! with TNF was not cytotoxic; in contrast, this cytokine enhanced the toxicity of SLD sulfide to both cell types. There is evidence that SLD and TNF act synergistically to kill tumor cells in mice, which raised the possibility of using this combination of agents as a new anticancer therapy (Hiroshi Yasui, 2003). However, our results suggest that this therapy might also increase the chance of liver injury. The mechanism of SLD sulfide and TNF interaction is under investigation. TNF can lead either to hepatocyte proliferation through NF-kappaB activation or to activation of cell death signaling (Wullaert et al., 2007). SLD, and particularly SLD sulfide, are potent inhibitors of the NF-kappaB pathway through inhibition of lKappa kinase activity (Yamamoto et al., 1999b). It was reported that NF-kB plays an essential role in preventing TNF- induced cell death (Beg and Baltimore, 1996). As a result, it is possible that SLD sulfide sensitizes hepatocytes to TNF-induced cell death through inhibition of NF-kappaB prosurvival signaling. Moreover, SLD sulfide and TNF share a common toxic effect, which may add to enhance cell death. It has been reported that SLD sulfide can induce reactive oxygen species (ROS) in vitro (Sun et al., 2009) and lead to mitochondrial uncoupling (Leite et al., 2006). TNF can also cause the production of ROS (Schwabe and Brenner, 2006) and mitochondrial injury (Bradham et al., 1998). Therefore, ROS and mitochondria are two potential targets of interaction of SLD and TNF. SLD sulfide and TNF are not the only mediators that contribute to hepatotoxicity in this model. Previously, we found that liver hypoxia is induced through the activation of the hemostatic system in SLD/LPS-cotreated rats and 113 that inhibition of coagulation protects from liver damage (Zou et al., 2009b). Hypoxia might contribute to liver injury through synergistic interplay with SLD sulfide. Furthermore, we cannot exclude the possible roles of other mediators. For example, proteases released from neutrophils are important in other drug/LPS models (Luyendyk et al., 2005; Deng et al., 2007a). The proinflammatory cytokine, interferon-gamma, has been shown to exacerbate TNF-induced cytotoxicity in hepatocytes (Adamson and Billings, 1993). These mediators might interact with SLD sulfide, TNF and/or hypoxia to promote liver injury. In summary, SLD and LPS interact to produce liver injury in rats. The LPS- stimulated increase in the concentration of TNF in rat serum was enhanced by SLD, and this cytokine plays a critical role in the pathogenesis. SLD sulfide was more toxic than SLD or SLD sulfone in vitro. Although LPS cotreatrnent reduced the bioavailability of SLD and the production of toxic SLD sulfide, the synergy of this toxic metabolite with TNF was sufficient to cause liver injury in rats. Such synergistic interactions might be a trigger for idiosyncratic liver injury from SLD in humans. 114 CHAPTER 4 Wei Zou, Robert A. Roth, Husam S. Younis, Ernst Malle, and Patricia E. Ganey. The critical role of tumor necrosis factor-a- and plasminogen activator inhibitor-1- mediated neutrophil activation in a sulindac/lipopolysaccharide-model of idiosyncratic liver injury in the rat. (Submitted) 115 4.1Abstr Prev: nonsteroi In this s investigal was grea LPS alor cotreated released liver injur PMN pro factorc l 01 liver il lPAl-i) ( deDOSlIIC fibrin de; or inhib, product}, in livers C'Olitribui indeDen. "‘1qu in prOmOtlr 4.1 Abstract Previous studies indicated that lipopolysaccharide (LPS) interacts with the nonsteroidal anti-inflammatory drug sulindac (SLD) to produce liver injury in rats. In this study, the mechanism of SLD/LPS-induced liver injury was further investigated. Accumulation of polymorphonuclear neutrophils (PMNs) in the liver was greater in SLDILPS-cotreated rats compared to those treated with SLD or LPS alone. In addition, PMN activation occurred specifically in livers of rats cotreated with SLD/LPS. We tested the hypothesis that PMNs and proteases released from them play critical roles in the hepatotoxicity. SLD/LPS-induced liver injury was attenuated by prior depletion of PMNs or by pretreatment with the PMN protease inhibitor, eglin C. Previous studies suggested that tumor necrosis factor-a (T NF) and the hemostatic system play critical roles in the pathogenesis of liver injury induced by SLD/LPS. TNF and plasminogen activator inhibitor-1 (PAl-1) can contribute to hepatotoxicity by affecting PMN activation and fibrin deposition. Therefore, we tested the role of TNF and PAl-1 in PMN activation and fibrin deposition in the SLD/LPS-induced liver injury model. Neutralization of TNF or inhibition of PAH attenuated PMN activation. TNF had no effect on PAl-1 production or fibrin deposition. In contrast, PAl-1 contributed to fibrin deposition in livers of rats treated with SLD/LPS. In summary, PMNs, TNF and PAl-1 contribute to the liver injury induced by SLD/LPS cotreatrnent. TNF and PAl-1 independently led to PMN activation, which is critical to the pathogenesis of liver injury in SLDILPS-treated rats. Moreover, PAl-1 contributed to liver injury by promoting fibrin deposition. 116 4.2 Introduction We reported previously that SLD enhanced the LPS-induced elevation of serum TNF and plasma PAl-1 in rats (Zou et al., 2009b). TNF neutralization protected against liver injury in this model, suggesting that TNF plays an important role in the pathogenesis (Zou et al., 2009a). Fibrin deposition in liver sinusoids resulted from cotreatrnent and contributed to SLD/LPS-induced hepatotoxicity (Zou et al., 2009b). TNF and PAI-1 participate in other liver injury models by causing PMN activation and fibrin deposition (Deng et al., 2008). Therefore, we investigated the role of TNF and PAl-1 in mediating PMN accumulation and activation as well as fibrin deposition in livers of SLD/LPS- treated rats. 117 4.3 Materials and methods 4.3.1 Materials LPS (Lot 075K4038) derived from Escherichia coli serotype 0552B5 with an activity of 3.3 X 106 endotoxin units (EU)/mg as well as SLD and its metabolites were purchased from Sigma-Aldrich (St. Louis, MO). Eglin C was provided by Novartis Phami (Basel, Switzerland). PA|039 was purchased from Axon Medchem BV (Groningen, Netherlands). 4.3.2 Animals Male, Sprague-Dawley rats (Crl:CD(SD)lGS BR; Charles River, Portage, MI) weighing 250 to 370 g were used. Animals were fed standard chow (Rodent Chow/Tek 8640; Harlan Teklad, Madison, WI) and allowed access to water ad libitum. They were allowed to acclimate for 1 week in a 12 hr light/dark cycle prior to use in experiments. All procedures were approved by the MSU Committee on Animal Use and Care and complied with “Guide for the Care and Use of Laboratory Animals” published by the National Academy of Sciences. 4.3.3 Animal model and sample collection The SLD/LPS-induced liver injury model was described previously (Zou et al., 2009b). Food was removed, and rats were given the first administration of SLD (50 mglkg, p.o.) or its vehicle (0.5% methyl cellulose) 16 hr before the second administration of the same dose. LPS (8.25X 105 EU/kg, i.v.) or its 118 vehicle (saline) was administered half an hour before the second administration of SLD. Rats were anesthetized at various times after the second administration of SLD. Serum and plasma was prepared from blood withdrawn from the vena cava. Liver tissue from the left lateral lobe was collected and fixed in 10% buffered formalin for PMN staining. A portion of the left medial lobe of the liver was flash-frozen in isopentane for determination of hypochlorous acid (HOCl)- protein adduct staining as well as for fibrin deposition analysis. 4.3.4 Anti-PMN serum, eglin C, PAI039 and etanercept treatment protocols In PMN depletion experiments, rabbit anti-PMN serum or normal rabbit serum control was diluted 1:1 in sterile saline and given to rats (0.5 ml per rat, i.v.) half an hour before the first administration of SLD. The efficacy of the anti- PMN serum in depleting PMNs has been demonstrated in previous studies (Deng et al., 2007b). A PMN protease inhibitor, eglin C (8 mglkg, i.v.; kindly provided by Novartis Pharrn AG, Basel, Switzerland) or its saline vehicle, was administered to rats 4, 6 and 8 h after the second administration of SLD. A PAl-1 inhibitor, PAI039 [{1-benzyl-5-[4-(trifluoromethoxy)phenyl]-1H-indoI-3- yl}(oxo)acetic acid] (6 mglkg, p.o.) or its vehicle (0.5% methyl cellulose) was administered to rats 1 hr after the second administration of SLD. Etanercept (8 mglkg) or vehicle (sterile water) was given to rats subcutaneously one hour before LPS or its saline vehicle. 4.3.5 Evaluation of hepatotoxicity 119 The activity of alanine aminotransferase (ALT) in serum was used as a marker to assess injury to hepatic parenchymal cells. The assay was performed using a diagnostic kit from Thermo Fisher Scientific (Waltham, MA). 4.3.6 Determination of ClNC-1, MlP-1a and PAl-1 concentrations in plasma The concentrations of cytokine-induced neutrophil chemoattractant-1 (ClNC- 1) and macrophage inflammatory protein-1o (MlP-1d) in plasma were estimated by multiplex ELISA. Specific antibody-coupled beads were purchased from Millipore Corp. (Billerica, MA). Functionally active PAl-1 was measured by ELISA using a commercially available test kit from Molecular Innovations, Inc (Southfield, MI). 4.3.7 Evaluation of liver PMN accumulation and activation Paraffin-embedded liver tissue was cut into 6 um-thick sections on which PMN immunohistochemistry was performed as described previously (Yee et al., 2003). Briefly, paraffin was removed with xylene, and liver sections were incubated with polyclonal rabbit anti-PMN lgG as first antibody, and then incubated with biotinylated goat anti-rabbit IgG, avidin-conjugated alkaline phosphatase, and Vector Red substrate to stain PMNs. The numbers of PMNs enumerated in 10 randomly selected, 400 X high power fields were averaged to assess PMN accumulation in the liver. The potent oxidant HOCI, generated from hydrogen peroxide by myeloperoxidase (MPO) in the presence of physiological choride concentrations, 120 EQCHSV used as lherhss fixed ir phosph dockec Probes whh a IV-IVI QC 3, 20l sides' antiboc Califcr bang averag 4£18,l Tl descn 4.39 I 1 F? fibfln‘ reacts with proteins to form chloramines. These HOCl-protein adducts can be used as fingerprints (Malle et al., 2006) to directly assess activation of PMNs in liver tissues (Hasegawa et al., 2005; Deng et al., 2007b). Frozen liver sections fixed in 4% formalin for 10 min at room temperature were washed with phosphate-buffered saline (PBS) 3 times for 5 min each. The sections were blocked for 1 hr at room temperature with 3% [vlv] goat serum (Molecular Probes, Carlsbad, CA) in PBS, and then incubated for 2 hr at room temperature with a monoclonal antibody (clone 2D1OG9, subtype lgG2bk; diluted 1:1 in 3% [vlv] goat serum) specific for HOCI-modified epitopes generated in vivo (Malle et al., 2006) and in vitro (Malle et al., 1995). After another 3 washes with PBS, slides were incubated with Alexa Fluor 488-labeled goat anti-mouse secondary antibody (diluted 1:500 in 3% [vlv] goat serum, Molecular Probes, Carlsbad, California). Ten pictures were taken of 200 X power, randomly selected fields using a fluorescence microscope, and the fraction of positive pixels was averaged for each slide (Deng et al., 2008). 4.3.8 Assessment of fibrin deposition in liver The immunohistochemistry for cross-linked fibrin in liver was performed as described previously (Zou et al., 2009b). 4.3.9 Statistical analyses Results are presented as means 1 SEM. Student’s t-test was performed on fibrin deposition data. For the rest of the studies, one way or two way analysis of variance (Ah Newman-Ker for statistical variance (ANOVA) was applied for data analysis, as appropriate, and Student- Newman-Keuls test was used as a post hoc test to compare means. The criterion for statistical significance was P < 0.05. 4.4 Result 4.4.1Eval Liver i administra (Zou et a collected 1 SLD/LPS. (Fig. MA: with LPS. LPS and E HOCI- Chlorlde s} MPO of to “I (not sl 99510085 v with SLD l peroxide-t 01 rats treE “'2 Time Plasma PIaSrr and the C0 4.4 Results 4.4.1 Evaluation of PMN accumulation and activation in livers Liver injury induced by SLD/LPS occurs between 4 and 8 hr after the second administration of SLD, and ALT activity in rats significantly increases by 12 hr (Zou et al., 2009b). Accordingly, PMN accumulation was assessed in livers collected at 4 hr, a time before the onset of hepatocellular injury induced by SLD/LPS. SLD given alone had no significant effect on hepatic PMN number (Fig. 4.1A). An increase in PMN number was observed in livers of rats treated with LPS. PMN numbers were significantly greater in livers of rats cotreated with LPS and SLD compared to those treated with LPS alone. HOCl-protein adducts are generated by the MPO-hydrogen peroxide- chloride system of activated PMNs (Malle et al., 2006), cells containing up to 5% MP0 of total cell protein content. Adducts were not elevated in liver sections at 4 hr (not shown). However, at 8 hr pronounced formation of HOCl-modified epitopes was found in livers of rats treated with SLD/LPS, but not in rats treated with SLD or LPS alone (Fig. 4.13). This result indicates that the MPO-hydrogen peroxide-halide system of PMNs was activated between 4 and 8 hr in the livers of rats treated with SLD/LPS. 4.4.2 Time course of changes in CINC-1 and MlP-1d concentrations in plasma Plasma was collected from rats euthanized at various times (1, 4 and 12 hr), and the concentrations of ClNC-1 and MlP-1a were measured. LPS increased 123 Fig. 4.1. Evaluaton of PMN accumulation and activation in rat livers. Rats were treated with two administrations of SLD (50 mglkg, p.o.) or its vehicle (Veh, 0.5% methyl cellulose) with a 16 hr interval. LPS (8.25 X 105 EU/kg, i.v.) or its saline vehicle was administered half an hour before the second administration of SLD. (A) PMN staining was performed on livers collected 4 hr after the second administration of SLD. PMN number in 400 X high power fields (HPF) was counted to evaluate PMN accumulation. (B) HOCl-protein adduct staining was performed on slides of frozen liver collected at 8 hr. Ten random fields were photographed for every section, and the fraction of positive pixels was determined. *significantly different from respective group without LPS. #significantly different from Veh/LPS group. P<0.05, n=4-5. 124 *# 31:0 *# I 125 80‘ * Tlfi , m. V h S .- w. w. . . . . L. s d - . w m m 0 m m m w m m ..._n__._ XOQV \ u 25?. 3.9.5 o>Ewoa Co .5302“: 968% 5365.50... UNCA both ch onCHt 143E hduci $8 fUl'l seed grcu incn abo Fete ha: CINC-1 and MlP-1d concentrations at 1 and 4 hr (Fig. 4.2). The concentrations of both chemokines had returned to baseline by 12 hr. SLD treatment had no effect on ClNC-1 or MIP-1u concentrations in vehicle- or LPS-cotreated rats. 4.4.3 Effect of PMN depletion and PMN protease inhibition on SLDILPS- induced liver injury To assess the role of PMNs in SLD/LPS-induced liver injury, rabbit anti-PMN serum or normal serum was given to rats. In a previous study, anti-PMN serum selectively reduced PMNs without affecting other leukocyte numbers in blood (Deng et al., 2007b). Blood PMN number in the anti-PMN serum/SLDILPS group (499 t 23) was significantly smaller than that in the normal serum/SLDILPS group (2726 :1: 144) at 12 hr. Cotreatment with normal serum/SLDILPS led to increased serum ALT activity (Fig. 4.3). Pretreatment with anti-PMN serum abolished the SLD/LPS-induced increase in ALT activity. Eglin C is a potent and selective inhibitor of elastase and cathepsin G released by activated PMNs (Schnebli et al., 1985; Braun et al., 1987). Eglin C had no effect on ALT activity in serum of rats treated with vehicle but attenuated the elevation in serum ALT activity of rats treated with SLD/LPS (Fig. 4.4). 126 it 42. Cor =3. ’Mth S. :‘3 ‘2 hr aft Fifidtfaticn *7 15} ma Fig. 4.2. Concentrations of PMN chemokines in rat plasma. Rats were treated with SLD and. LPS or their vehicles (Veh) as described in Fig. 4.1. At 1, 4 and 12 hr after the second administration of SLD, plasma was collected and concentrations of (A) cytokine-induced neutrophil chemoattractant-1 (ClNC-1) and (B) macrophage inflammatory protein-1a (MlP-1or) were evaluated by multiplex ELISA. *significantly different from VehNeh group at the same time. P<0.05, n=5. 127 CINC-1 (pglmL) MlP-1a (ngImL) 2500 . 2000 . 1500 . 1000 . 500 - 120 ~ 100 . 80 . 60 . 40 . zol 1 hrs after 2nd dose of SLD — VehNeh I: SLDNeh * — Veh/LPS 1:1 SLD/LPS 4 12 1 4 12 hrs after 2nd dose of SLD 128 Fig. 4.3. Effect of PMN depletion on SLDILPS-induced liver injury. Rats were pretreated with either normal serum (NS) or rabbit anti-rat PMN serum (AS) half an hour before the first administration of SLD. Rats were euthanized 12 hr after the 2"d dose of SLD, and serum ALT activity was determined. Vehicle (Veh). *significantly different from VehNeh/NS group, #significantly different from SLD/LPS/NS group. P<0.05, n=3-6. 1000 * — NS [:3 AS 800 l 600 . 400 - 200- # o -———-—- [j VehNeh SLD/LPS ALT (UIL) 129 Fig.4.4. Effect of PMN protease inhibition on SLDILPS-induced liver injury. Eglin C or its vehicle (Veh) was administered to rats 4, 6, and 8 hr after the 2nd administration of SLD. Rats were euthanized at 12 hr, and ALT activity in serum was determined. *significantly different from VehNehNeh. #significantly different from VehNeh/Eglin C group. asignificantly different from SLDILPSNeh group. P<0.05, n=3-6. 2500 - - Veh l:l Eglin C * 2000 - 1500 # a 1000 - ALT (UIL) 500 ‘ VehNeh SLD/LPS 130 4.4.4 Effect of TNF on PMN accumulation and activation As noted above, cotreatrnent with SLD/LPS caused an increase in the number of PMNs in liver (Fig. 4.1A). Etanercept, which neutralizes TNF and inhibits its biological effects, did not affect PMN numbers in livers of rats treated with SLD/LPS (Fig. 4.5A). In contrast, etanercept prevented the elevation in SLD/LPS-induced formation of HOCl-protein adducts (Fig. 4.5B). These results suggest that TNF contributes to the release of cytotoxic factors from PMNs but not to PMN accumulation in the liver. 4.4.5 Role of PAH in liver injury and accumulation and activation of PMNs A previous study indicated that PAl-1 was selectively increased in the plasma of SLDILPS- cotreated rats (Zou et al., 2009b); however, its role in liver injury has not been investigated. The PAl-1 inhibitor, PAI039, greatly attenuated liver injury, as followed by ALT measurements (Fig. 4.6a) induced by SLD/LPS cotreatrnent. PAI039 also reduced PMN activation (Fig. 4.60) but not PMN accumulation at 8 hr (Fig. 4.6b). 4.4.6 Effect of TNF on plasma PAI-1 concentration In previous studies, TNF was significantly increased as early as 1 hr by SLD/LPS cotreatrnent, and PAl-1 was increased by 8 hr (Zou et al., 2009b). To evaluate whether TNF regulates the production of PAH, plasma concentration of PAH was evaluated in rats cotreated with etanercept. Etanercept given at a 131 Fig. 4.5. Effect of TNF inhibition on PMN accumulation and activation. Rats administered SLD/LPS were pretreated with etanercept or its vehicle (Veh) 1 hr before LPS. (A) PMN staining was performed on livers collected at 8 hr. The accumulation of PMNs in livers was evaluated by averaging PMN numbers in 10, randomly chosen, 400 X fields. (B) Quantification of HOCl-protein adducts in the livers of rats at 8 hr. *signiflcantly different from VehNehNeh group. #significantly different from Veh/SLDILPS group. P<0.05, n=3-6. 132 — VehNehNeh 1:1 VehlSLD/LPS — EtanISLD/LPS 1 . . _ . . m a ..u 5 m 5 ..... M m m w m o m m m m m. m m. 1 o o o 0 o o o "E: Xeov \ n st Egan e>Emoa .0 5:69”: 36:25 539.... -50: 133 i94.6. Effect 1111311011. Rat tithiPAIOE “ stered tc :‘aiized at mediation I 5370:3038 t Fig. 4.6. Effect of PAH inhibition on liver injury and PMN accumulation and activation. Rats were treated with SLD/LPS as described in Fig. 4.1. PAl-1 inhibitor, PAI039 (6 mglkg, p.o.), or its vehicle (Veh, 0.5% methyl cellulose) was administered to rats at 1 hr after the second administration of SLD. Rats were euthanized at 12 hr to measure ALT activity (A) or at 8 hr to assess PMN accumulation (B) and activation (C). *significantly different from VehNehNeh. #significantly different from SLDILPSNeh group. P<0.05, n=4-16. 134 — VehNehNeh CZ! SLDILPSNeh * _ SLD/LPS/PAl039 800 i o 6 0 o 4 2 “E: 531:“ 2:...— * o 5 o 5 o 5 o 3 2 2 1 1 W o h 0 o. h o . n o o o o o 0. 0 .282.— e>Euoa he 5:62.... 2268 522.. .80: 135 dose thal F increase in 4.4.7 Effec Fibrin result in h were redi According causingf caused b dose that protected against liver injury (Zou et al., 2009a) had no effect on the increase in plasma PAl-1 concentration caused by SLD/LPS (Fig. 4.7). 4.4.7 Effect of TNF and PAl-1 on fibrin deposition Fibrin clots form in the sinusoids of livers of SLD/LPS-cotreated rats and result in hepatic hypoxia (Zou et al., 2009b). Both fibrin deposition and hypoxia were reduced by anticoagulant treatment, which protected against liver injury. Accordingly, we evaluated whether TNF or PAI-1 exerts its toxic effect by causing fibrin deposition in the liver. Etanercept had no effect on fibrin deposition caused by SLD/LPS cotreatrnent, whereas PAI039 reduced it (Fig. 4.8). 136 Fig. 4.7. Effect of TNF inhibition on plasma PAl-1 concentration. Rats were treated with etanercept (Etan), SLD and LPS or their vehicles (Veh) as described in the legend to Fig. 4.5. Plasma active PAI-1 concentration was determined at 8 hr. * significantly different from VehNehNeh. P<0.05, n=4. — VehNehNeh C31 VehlSLD/LPS _ EtanlSLD/LPS 600 - * 400 « 200 i, N Plasma Active PAl-1 (ngImL) 137 Fig. 4.8. Effect of TNF or PAI-1 inhibition on fibrin deposition in liver. Rats were treated with SLD/LPS and etanercept (Etan, A) or PAl-1 inhibitor (B) respectively. Fibrin deposition was evaluated at 8 hr. * significantly different from SLD/LPSNehicle (Veh). P<0.05, n=4-7. 138 A 1. mmmmmmm . 0 a 0 o. 0 o. oni 0238a Co .5on“: 55¢ SLDILPS mmmmm o 0 0 0 0 3.9.5 0223.. he c232“: 55E Veh SLDILPS 139 4.5 Discussion PMNs are a double-edged sword in the innate immune response to microbial infection and tissue trauma (Butterfield et al., 2006). Stimulated by inflammatory signals, PMNs attach to endothelial cells via adhesion molecules and transmigrate to the site of infection/trauma, where they become activated to release cytotoxic factors. PMNs can be beneficial by removing invading organisms and stimulating tissue repair. However, excessive PMN activation causes tissue injury in many animal models (Jaeschke et al., 1990; Hewett et al., 1992). PMNs are involved in several models of drug-induced liver injury and in drug-LPS interaction models of idiosyncratic liver injury (Deng et al., 2006; Deng et al., 2007b; Ramaiah and Jaeschke, 2007; Shaw et al., 2009d). As has been reported, LPS administration causes PMNs to accumulate in liver. Although SLD mildly inhibited the adhesion of PMNs to nylon-wool columns in vitro (Venezio et al., 1985), it increased the LPS-induced PMN accumulation in liver before the onset of liver injury (Fig. 4.1A). Two PMN chemokines, MlP-1a and ClNC-1, are potent inducers of PMN recruitment and extravasation. A neutralizing antibody to either MlP-1a or ClNC-1 attenuated neutrophil sequestration in LPS-treated rodents (Standiford et al., 1995; Zhang et al., 1995). The concentrations of both chemokines were significantly increased in plasma by LPS, whereas SLD had no effect (Fig. 4.2). Thus, both chemokines might contribute to PMN accumulation in livers of LPS-treated rats; however, other factors must be involved after SLD/LPS cotreatment. The reason why SLD enhanced PMN accumulation is unknown, but some possibilities arise from 140 previous results. It is known that SLD/LPS cotreatrnent caused fibrin deposition in the liver (Zou et al., 2009b). It is possible that entrapment of PMNs in the meshwork of sinusoidal fibrin occurred. In addition, as a result of fibrin clots in sinusoids hypoxia occured in livers of cotreated rats (Zou et al., 2009b). Hypoxia can enhance the adherence of PMNs to human endothelial cells in vitro (Milhoan et al., 1992), and such an effect might further explain the SLD-induced increase in PMN accumulation. Generally, PMNs that sequester in the liver are not injurious unless extravasation of them into the parenchyma and activation occur (Chosay et al., 1997). Although PMNs accumulated in livers in rats treated only with a small dose of LPS (Fig. 4.1), staining for HOCI-modified proteins, specific markers for neutrophil-induced oxidant stress, suggested no activation of PMNs. SLD/LPS cotreatrnent, however, increased HOCI-protein adducts, indicative that the MPO- hydrogen peroxide-chloride system of PMNs becomes activated between 4 and 8 hr, when the onset of liver injury occurred. Our observations parallel recent findings in patients with steatohepatitis in which liver chemokine expression was higher in patients with MPO-mediated oxidation products and correlated with hepatic neutrophil sequestration (Rensen et al., 2009). The role of PMNs in SLDILPS-induced liver damage was further tested using anti-PMN serum, which markedly reduced PMNs in the circulation. The protection by anti-PMN serum shows that PMNs are critical to the development of SLDILPS-induced liver injury (Fig. 4.3). 141 Activated PMNs release various lysosomal hydrolases including serine proteases, among which elastase and cathepsin G have been identified as primary mediators in hepatocyte killing by PMNs in vitro (Ho et al., 1996). Eglin C is an inhibitor of elastase and cathepsin G. It attenuated liver injury (Fig. 4.4), suggesting that PMN proteases also play a role in the pathogenesis. Compared to the complete protection by PMN depletion, eglin C incompletely reduced SLDILPS-induced liver injury; thus, the proteases released from PMNs might not be the only PMN-derived mediators contributing to the pathogenesis. In numerous inflammatory liver injury models, antioxidants attenuated PMN- mediated liver injury in vivo (Liu et al., 1995; Jaeschke and Smith, 1997). Our results to date cannot rule out a role for reactive oxygen species other than HOCI, but this is a topic of current investigation. TNF neutralization significantly attenuated liver injury induced by SLD/LPS in vivo (Zou et al., 2009a). In addition, TNF directly interacted with SLD sulfide to kill hepatocytes in vitro. TNF can also activate endothelial cells to promote PMN migration (Smart and Casale, 1994). In results presented here, the number of PMNs sequestered in the liver was not affected by TNF neutralization, but TNF neutralization did reduce PMN activation in the liver (Fig. 4.5). These results suggest that TNF contributes to PMN activation, but not to hepatic accumulation of these cells. Like PMN depletion, anticoagulation using heparin abolished the hepatotoxicity induced by SLD/LPS cotreatrnent of rats (Zou et al., 2009b), which suggests that there is an interaction between PMNs and the hemostatic system in the pathogenesis. Hemostatic factors including thrombin and PAl-1 were increased in plasma by SLD/LPS cotreatrnent (Zou et al., 2009b). Interestingly, hemostatic factors can bind to PMNs and influence their accumulation and activation (Gillis et al., 1997). For example, thrombin can rapidly trigger lysozyme release from human PMNs and promote PMN activation in perfused rat liver after LPS exposure (Baranes et al., 1986; Copple et al., 2003). PAl-1 is an inhibitor of plasminogen activator and a key negative regulator of fibrinolysis. PAI039, a PAl-1 inhibitor, significantly attenuated SLDILPS-induced liver injury, suggesting that PAl-1 is a mediator of pathogenesis (Fig. 4.6A). PAI039 decreased fibrin deposition in livers of SLDILPS-treated rats (Fig. 4.83), which suggests that PAl-1 contributes to fibrin deposition in the SLD/LPS model. In addition to inhibiting fibrinolysis, PAI-1 can regulate PMN migration and potentiate LPS-induced PMN activation through a c-Jun N-tenninal kinase- mediated pathway (Kwak et al., 2006; Roelofs et al., 2009). Consistent with these findings, PAl-1 inhibition reduced HOCl-protein adduct staining in livers of SLDILPS-cotreated rats (Fig. 4.6C), suggesting that PAI-1 is involved in PMN activation. Therefore, PAI-1 contributed to both PMN activation and fibrin deposition. It can also play a proinflammatory role by stimulating the production of cytokines and chemokines. For example, in a murine model of trovafloxacin/LPS-induced liver injury, PAl-1 knockout markedly decreased the plasma concentrations of interleukin-1B, interleukin-10, keratinocyte chemoattractant and monocyte chemoattractant protein-1, respectively (Shaw et al., 2009c). Whether PAl-1 similarly regulates the production of chemokines in 143 SLD/LPS— treated rats and the role of these cytokines in PMN activation are topics for further investigation. PMNs can exacerbate fibrin deposition by releasing proteases (Deng et al., 2007b). For example, proteases from PMNs can release PAl-1 from endothelial cells and platelets and thereby inhibit fibrinolysis (Pintucci et al., 1992). Eglin C treatment significantly decreased active PAI-1 concentration and fibrin deposition in a model of ranitidine/LPS-induced liver injury (Deng et al., 2007b). SLD/LPS cotreatrnent led to fibrin deposition at 4 hr, before the activation of PMNs at 8 hr. Therefore, PMNs do not contribute to the initial formation of fibrin, but proteases released by activated PMNs might prolong fibrin deposition. The concentrations of PAH and TNF in blood were both significantly greater in SLDILPS-treated rats than in rats treated with either LPS or SLD alone. The peak of PAl-1 (4 hr) in plasma followed the peak of TNF production (i.e.,1 hr; Zou et al., 2009b). Although it has been reported that both TNF and LPS lead to PAI- 1 release from endothelial cells in vitro (Riedo et al., 1990), inhibition of TNF did not decrease PAl-1 concentration in SLDILPS-cotreated rats (Fig. 4.7). This indicates that PAl-1 production does not depend on TNF in this model. Consistent with this result, TNF did not affect fibrin deposition in liver (Fig. 4.8). Thus, the activation of hemostatic system is likely a direct effect of LPS but not mediated through TNF in this model. In contrast, TNF does mediate hemostatic system activation in ranitidine/LPS- or trovafloxacin/LPS-induced liver injury (T ukov et al., 2007a; Shaw et al., 2009e). Therefore, these results suggest that 144 TNF does not contribute to liver injury through the same mechanism in all drug/LPS interaction models. From results of this and previous studies, we can summarize mechanisms of liver injury induced by SLDILPS cotreatrnent (Fig. 4.9). Various mediators including TNF, hypoxia caused by hemostatic system activation and PMNs play critical roles in the pathogenesis of SLDILPS-induced liver injury. SLD enhances TNF elevation induced by LPS. SLD/LPS cotreatrnent also leads to the production of hemostatic factors including thrombin and PAl—1, both of which contribute to fibrin clot formation in liver sinusoids (Zou et al., 2009b). As a result, the liver becomes hypoxic. Although the concentration of the toxic metabolite, SLD sulifide, is decreased by LPS in livers and plasma of rats, it synergistically kills hepatocytes in the presence of TNF and hypoxia (Zou et al., 2009a). PMNs are another critical player in SLDILPS-induced liver injury (Fig. 4.3). PMN accumulation in the liver was primarily induced by LPS and this effect was enhanced by SLD (Fig. 4.1A). Activation of PMNs was observed in livers of rats treated with SLD/LPS (Fig. 4.18). Both TNF and PAl-1 contribute to PMN activation independently (Fig. 4.5 to 4.7). When activated, PMNs release proteases which induce liver injury by interacting with hypoxia (Luyendyk et al., 2005). In summary, the studies presented here further our understanding of the roles of various mediators and their interaction in this SLDILPS-induced idiosyncratic liver injury model. 145 Fig. 4.9. Mechanisms of SLDILPS-induced liver injury. See text for details N PMN _.___.. PMN _... proteases accumulation activation Thrombin—9 Tfibrin ——-b hypoxia PAI-1 146 CHAPTER 5 Wei Zou, Robert A. Roth, Husam S. Younis, Lyle D. Burgoon, and Patricia E. Ganey. Oxidative stress is an important player in the pathogenesis of liver injury induced by sulindac and lipopolysaccharide cotreatrnent 147 5.1 Abstract Among all the nonsteroidal anti-inflammatory drugs, sulindac (SLD) is associated with the greatest incidence of idiosyncratic hepatotoxicity in humans. Previously, an animal model of SLD-induced idiosyncratic hepatotoxicity was developed by cotreating rats with a nonhepatotoxic dose of LPS. Tumor necrosis factor-alpha (T NF) was found to be critically important to the pathogenesis. In this study, we further explored the mechanism of liver injury induced by SLD/LPS cotreatment by analyzing gene expression in livers of rats before the onset of liver injury. The results suggested that oxidative stress might be a potential mediator. Moreover, protein carbonyls, products of oxidative stress, were elevated in liver mitochondria of SLDILPS-cotreated rats. Antioxidant treatment witheither ebselen or dimethyl sulfoxide attenuated SLDILPS-induced liver injury. The role of oxidative stress was further investigated in vitro. SLD sulfide, the toxic metabolite of SLD, enhanced TNF-induced cytotoxicity and caspase 3/7 activity in HepG2 cells. SLD sulfide increased dichlorofluorescein fluorescence in HepG2 cells, suggesting generation of reactive oxygen species (ROS). Hydrogen peroxide and TNF cotreatrnent caused greater cytotoxicity than either treatment alone. Either antioxidant tempol or a pancaspase inhibitor Z-VAD-FMK decreased HepG2 cell death as well as caspase 3/7 activity induced by SLD sulfide/1' NF coexposure. These results indicate that SLDILPS treatment causes oxidative stress in livers of rats and that reactive oxygen species are important in the cytotoxic interaction of SLD and TNF by activating caspase 3/7. 148 5.2 Introduction Reactive oxygen species (ROS) include oxygen free radicals and other nonradical but highly reactive molecules (e.g., hydrogen peroxide). Excessive generation of ROS tilts the balance between prooxidant and antioxidant influences in the cell and results in oxidative stress. ROS can directly oxidize proteins, DNA or membrane lipids in target cells, and such effects can result in cell death. One ROS-mediated pathway of cell death is through caspase- dependent intracellular apoptotic signaling initiated by oxidative stress (Jones et al., 2000). The oxidative stress in liver can be induced under various conditions that include consumption of ethanol or other drugs that cause inflammatory stress (Galati et al., 2002; Choi and Ou, 2006; Cederbaum et al., 2009). In this study, gene expression was analyzed in livers from rats treated with sulindac and/or LPS. The results of Ingenuity pathway analysis of SLD/LPS- specific gene expression profiles pointed to the occurrence of oxidative stress. We tested the hypothesis that oxidative stress plays a role in the pathogenesis of liver injury induced by SLD/LPS in vivo. In hepatocytes, we evaluated the ability of SLD and its metabolites to prompt ROS generation and explored its role in cytotoxicity. 149 5.3 Materials and methods 5.3.1 Materials Unless othenrvise noted, all chemicals were purchased from Sigma-Aldrich (St. Louis, MO). The LPS (Lot 075K4038) used in animal experiments was derived from Escherichia coli serotype O55:B5 and had an activity of 3.3 X 106 endotoxin units (EU)/mg. HepG2/C3A cells were obtained from American Type Culture Collection (Manassas, VA). 5.3.2 Animals Male, Sprague-Dawley rats (Crl:CD(SD)lGS BR; Charles River, Portage, MI) weighing 250 to 370 g were used in this study. They were allowed to acclimate for 1 week in a 12-hr light/dark cycle prior to use in experiments. Animals were fed standard chow (Rodent Chowfl’ek 8640; Harlan Teklad, Madison, WI) and allowed access to spring water ad libitum. Experimental procedures complied with “Guide for the Care and Use of Laboratory Animals” (National Academy of Sciences). 5.3.3 Design of experiments in vivo As described in a previous study (Zou et al., 2009b), rats were given the first administration of SLD (50 mglkg, p.o.) or its vehicle (0.5% methyl cellulose), and food was removed at this time. Sixteen hours later, SLD at the same dose or its vehicle was administered to rats. LPS (8.25 X 105 EU/kg, i.v.) or its vehicle 150 (saline) was administered via a tail vein half an hour before the second administration of SLD. As reported previously, this protocol results in liver injury in the SLDILPS-cotreated rats (Zou et al., 2009b). At various times (4, 8 or 12 hr) after the second administration of SLD, rats were anesthetized with isoflurane, and blood was drawn from the vena cava. Serum was prepared from clotted blood. A portion of the right medial lobe of the liver was flash-frozen in liquid nitrogen for RNA extraction. Another portion was collected and cooled in ice-cold isolation buffer for mitochondrial preparation. In experiments designed to evaluate the effect of antioxidants on liver injury, ebselen (50 mglkg, p.o.) or its vehicle (0.5% methyl cellulose) was given to rats 1.5 hr before the second administration of SLD; dimethyl sulfoxide (0.3 mL/g, i.p.) was given to rats at the same time as the administration of SLD. 5.3.4 Gene expression analysis Total RNA was extracted from frozen liver tissue collected at 4 hr using a kit purchased from Qiagen Inc. (Valencia, CA) as described previously (Younis et al., 2006). Microarray analysis was performed using the standard protocol provided by Affymetrix, Inc. (Santa Clara, CA). Total RNA (10 pg) was reverse transcribed into cDNA in the presence of oligo dT primer using a Superscript II Double-Strand cDNA synthesis kit (lnvitrogen, Carlsbad, CA). cDNA was purified, and biotin-labeled cDNA was synthesized using the Enzo RNA Transcript Labeling Kit (Affymetrix, Santa Clara, CA). After the labeled cDNA was purified and its quality was evaluated, cDNA was hybridized to a Rat Genome 230 2.0 151 Array (Affymetrix), which comprised more than 31,000 probe sets. The array was stained with streptavidin-phycoerythrin (Invitrogen, Carlsbad, CA) and scanned to generate signal intensity files. Data normalization was performed using SAS version 9.1 on scanned image files (Eckel et al., 2005). Empirical Bayes analysis was used to calculate posteriori probabilities (P1(t) value) (Eckel et al., 2004). Ratios of gene expression were calculated by comparing SLDNeh, Veh/LPS or SLD/LPS groups to the VehNeh group. 5.3.5 Evaluation of liver injury and protein carbonyls in mitochondria Liver injury was assessed by measuring the activity of alanine aminotransferase (ALT) in serum using a diagnostic kit from Thermo Corp (Waltham, MA). Mitochondria were isolated from livers of rats treated with SLD/LPS or vehicles at 8 hr using a mitochondrial isolation kit (Sigma, St. Louis, MO). Briefly, fresh livers (50 mg) were collected and homogenized in HEPES buffer containing 200 mM mannitol, 70 mM sucrose, and 1 mM EGTA. Liver homogenates were centrifuged at 600xg for 5 minutes. The supernatant fraction was transferred into a new tube and centrifuged at 11,000xg for 10 minutes. The pellets were washed and centrifuged at 11,000xg for 10 minutes again. The mitochondrial pellets were resuspended in HEPES for the evaluation of membrane potential or protein carbonyl concentration. Protein carbonyl concentration in liver mitochondria was 152 measured using a commercially available kit purchased from Cayman Chemical (Ann Arbor, MI). 5.3.6 Evaluation of mitochondrial membrane potential The effect of SLD and its metabolites on mitochondrial membrane potential was evaluated using JC-1 mitochondrial membrane potential assay kit from Cayman Chemical (Ann Arbor, MI). Mitochondria isolated from normal rats (2 ug protein) were incubated with various concentrations of SLD or its metabolites at a final volume of 200 uL. After 30 min, the JC-1 dye solution (0.2 uL) was added. Fluorescence was read for JC-1 agglomerates and monomers respectively. The ratio of JC-1 agglomerates (excitation/emission=560l595 nm) to monomers (excitation/emission= 485/535 nm) in mitochondria was calculated, and the data were expressed as a percentage of vehicle control (0.5% dimethyl sulfoxide). The decrease in ratio of JC-1 agglomerates to monomers was associated with a decrease in membrane potential (Reers et al., 1995). 5.3.7 Evaluation of reactive oxygen species in HepGZ cells ROS in HepG2 cells were assessed using 5-(and-6)-chloromethyl-2’,7”- dichlorodihydrofluorescein diacetate, acetyl ester (CM-H2DCFDA) purchased from lnvitrogen, Inc. (Carlsbad, CA). HepG2 cells (4 x 105 cells/mL) in suspension were incubated with 10uM CM-HZDCFDA in Dulbecco’s Modified Eagle’s medium (DMEM) for 45 min. The cells were washed twice with DMEM with 10% fetal bovine serum. Cells were plated in 96-well plates and treated with 153 250 UN absenc DCF flL 5.3.8 E Hi DMEM reneWI 08008! (200- vehicle percel et al., HepG 5.3.9 variar Keuls Critefi e”‘lpil 250 uM SLD sulfide or its dimethyl sulfoxide (DMSO) vehicle in the presence or absence of recombinant human TNF (200 ng/ml) dissolved in DMEM vehicle. DCF fluorescence intensity was read at 0, 0.5, 1 and 3 hr after treatment. 5.3.8 Evaluation of cytotoxicity and capase 3 activity HepGZ cells were plated in 96-well plates at a density of 4 x 105 cells/mL in DMEM with 10% fetal bovine serum. After overnight incubation, medium was renewed, and HepG2 cells were treated with tempol (0.2- 1 M) or with the pancaspase inhibitor, Z-VAD-FMK (Z-VAD,10uM). Half an hour later, SLD sulfide (200— 250 uM), hydrogen peroxide (0.2- 2 mM), TNF (200 ng/mL) or their vehicles were added depending on the purpose of the experiment. The percentage of LDH released was evaluated at 24 hr as described previously (Zou et al., 2009a). Caspase 3” activity was evaluated at 6 hr after treatment in HepGZ cells using a Caspase-Glo 3/7 assay kit (Promega, Madison, WI) 5.3.9 Statistical analyses Results are expressed as means i S.E.M. One-way or two-way analysis of variance was applied for data analysis as appropriate, and Student-Newman- Keuls test was used as a post hoc test to compare means. For all studies, the criterion for statistical significance was P < 0.05. For gene array analysis, empirical Bayes analysis was used and posterior probability (P1(t)- value) > 0.9 was set as the criterion for significance. 154 5.4 Results 5.4.1 Gene expression changes regulated by treatment with SLD, LPS or SLDILPS In previous studies, neither LPS nor SLD was hepatotoxic when given alone; however, cotreatrnent with SLD/LPS caused severe liver injury in rats (Zou et al., 2009b). SLDILPS-induced liver injury occurred between 4-8 hr after the second administration of SLD. Genes regulated by SLD/LPS at the onset of liver injury i.e., 4 hr, might be involved in the pathogenesis. Thus, livers were collected 4 hr after treatment with SLD and/or LPS or their vehicles, and gene expression was analyzed. To compare the number of gene expression changes (relative to VehNeh) caused by treatment with SLDNeh, Veh/LPS or SLD/LPS, a Venn diagram was generated (Fig. 5.1). A large number of genes (1476) were changed by LPS administration. In contrast, SLD only caused a small number of genes expression changes (79). SLD/LPS cotreatrnent led to expression changes in 2040 genes. Not surprisingly, most of the gene expression changes caused by LPS (1309/1476) were represented in the genes regulated by SLD/LPS cotreatrnent. However, there were 721 genes regulated by SLD/LPS that were not affected by either SLD or LPS treatment alone (presented in appendix). 5.4.2 Gene expression changes specifically regulated by SLDILPS point to oxidative stress SLD/LPS was the only treatment that resulted in liver injury (Zou et al., 2009b). Accordingly, some of the 721 genes selectively regulated by SLD/LPS 155 F151. Venn d 101%. Rats w tiehlcle (0.5% r :e second admin csadministered fer the second stression was 8‘ I-‘ite sets Chang silthehNeh as iailment, and : linent groups 1113160 083 Fig. 5.1. Venn diagram of probe sets regulated by SLDNeh, Veh/LPS or SLDILPS. Rats were treated with two administrations of SLD (50 mglkg, p.o.) or its vehicle (0.5% methyl cellulose) with a 16 hr interval (n=5). Half an hour before the second administration of SLD, LPS (8.25X 105 EUIkg, i.v.) or its saline vehicle was administered via a tail vein. Livers were collected from rats euthanized at 4 hr after the second administration of SLD. RNA was isolated from liver, and gene expression was evaluated as described in Materials and Methods. The numbers of probe sets changed by SLDNeh, Veh/LPS or SLD/LPS cotreatrnent was derived using VehNeh as baseline. S indicates SLDNeh treatment, L indicates Veh/LPS treatment, and SL indicates SLD/LPS cotreatrnent. The intersection (A) of treatment groups represents the gene expression changed after both or all three of the indicated treatments. 156 are most were furll the genes by SLD/L ddennhh metabolis pathway 5.4.3 Oxi P101l treated I SQHmCar ht Sugg cotreatm Pret 53). The etheret 5.4.4 En MnOChm Sulfide 0 “TIM dic are most likely to be involved in the pathogenesis of liver injury. These genes were further subjected to Ingenuity Pathway Analysis, which annotated 576 of the genes, including 83 that were upregulated and 493 that were downregulated by SLDILPS. A list of toxicity pathways selectively affected by the 576 genes was determined (Table 5.1). Genes associated with pathway involved in fatty acid metabolism, LPS/IL—1-mediated inhibition of RXR function, NFkB signaling pathway and oxidative stress were highly impacted by SLD/LPS cotreatrnent. 5.4.3 Oxidative stress in SLDILPS-cotreated rats Protein carbonyl concentration was not affected in liver mitochondria of rats treated with SLD or LPS alone (Fig.5.2). In contrast, SLD/LPS cotreatrnent significantly increased protein carbonyls in mitochondria of livers collected at 8 hr, suggesting that hepatic oxidative stress was associated with SLD/LPS cotreatrnent. Pretreatment of rats with either ebselen or DMSO reduced liver injury (Fig. 5.3). That is, ALT activity at 12 hr was significantly increased by SLD/LPS, and either ebselen or DMSO markedly decreased serum ALT activity. 5.4.4 Effect of SLD sulfide on mitochondrial membrane potential Mitochondria isolated from livers of untreated rats were incubated with SLD, SLD sulfide or SLD sulfone. Compared to vehicle treatment, SLD or SLD sulfone up to 1mM did not change the ratio of JC-1 aggregates and monomers, a marker of 158 lable 5.1. F SLDILPS cotl SLDILPS. The Zhways was Cisewed nurr Either eXpe. “59101 ger Table 5.1. Pathways associated with genes specifically changed by SLDILPS cotreatrnent. Expression of 721 genes was changed specifically by SLD/LPS. These genes were imported into Ingenuity Pathway Analysis. A list of pathways was derived and ranked by p value, which indicates the deviation of observed number of genes for each pathway found in the imported list from the number expected to occur by chance. Ratio indicates the percentage of the number of genes affected in a particular pathway. 159 Fahy. Mehfl _._————-I PXFM —— LPSH lnmbi FunC —_ Xenc Meta —‘ Ann! Rece \- Hep; Idec Reg Pen Pun JiEf NFk Pah \ CYlt Pan 8F} 3+3 OXII \ PP) Act \ Toxicity Lists -Log(P) Ratio (%) Genes ALDH1A1,PECI,CYP4B1,A Fatty Acid 3 27 11 0 UH,ADH1C,CYP2D6, Metabolism ' ' CYP2J2, ACAD9,ADHFE1, HSD17B4,ACSL1,GCDH PRKACB,GSTM2,ALDH1A PXR/RXR Activation 2.95 12.7 1,AKT1,PRKACA,IL6,HNF4 A,RXRA ALDH1 L1 ,SLCO1A2,MAOB LPS/lL-1 Mediated ,GSTM2,ALDH1A1,MGST2, Inhibition of RXR 2.88 8.5 E6629219,XPO1,SULT1E1 Function ,LBP,RXRA,ACSL1,SULT1 B1 ,MGST3,ALDH6A1 GSTM2,MGST2,CYP4B1 ,A Xenobiotic 2 55 9 1 DH1C, Metabolism ' ' ALDH1 L1 ,CYP206,CYP2J2 ,ADHFE1, MGST3 lL1A,GSTM2,ALDH1A1,MG Aryl Hydrocarbon 2 43 8 6 ST2,MAPK1,CDK4,ALDH1L Receptor Signaling ' ' 1,NFlB,lL6,RXRA,ALDH6A 1,MGST3 PRKACB,IL1A,NFKBIA,PR . . KACA,SLCO1A2,LBP,IL6,H Hepatic Cholestasts 2.18 8.6 NF 4 A,RXRA, CYPBB 1 ,IRAK 2 Mechanism of Gene Regulation by PRKACB,IL1A,NFKBIA,MA Peroxisome 1 .66 8.60 PK1 ,PDGFA,ME1,RXRA,H Proliferators via SD17B4 PPARo - - PRKACB,TLR2,|L1A,AKT1, ngB s'gna'mg 1.53 7.6 GHR,NFKBIA,PRKACA,MA athway P3K8 Cytochrome P450 Panel - Substrate is a Fatty Acid 1.43 20.0 CYP4B1,CYP2J2 (Human) Oxidative Stress 1.28 8.93 ‘ng3'VCAM1'GSTM2'ME‘ PRKACB GHR NFKBIA MA PPARd/RXR ' ' ' Activation 1.26 6.37 PK1,PRKAA2,PRKACA,PL 160 CG1,|L6,ADIPOR2,RXRA Fig. Rats were isola diffel P<0. Fig. 5.2. Evaluation of protein carbonyl concentration in liver mitochondria. Rats were treated with SLD, LPS or their vehicles as described in Fig. 5.1. They were euthanized at 8 hr, and the livers were collected. Liver mitochondria were isolated, and protein carbonyl concentration was determined. *Significantly different from SLDNeh group. #Significantly different from Veh/LPS group. P<0.05, n=4-8. S" o *# N o- .N o ..s O P 01 Protein Carbonyl (nmollmg mitochondrial protein) 0| P o Veh SLD 161 fig. 5.3. Efi :ehylcellulc Serum ALT group. #Sigr Fig. 5.3. Effect of antioxidants on liver injury. (A) Ebselen, its vehcle (0.5% methylcellulose) or (8) DMSO was administered to rats treated with SLD/LPS. Serum ALT activity was evaluated at 12 hr. *Significantly different from VehNeh group. #Significantly different from SLD/LPS group. P<0.05, n=3-9. ALT (UIL) 1400 1200 ‘ 1000 ‘ 600 - ALT (UIL) 400 - 200 - 1 200 1000 800- 600‘ — VehNehNeh * E Veh/SLDILPS — EbselenlSLD/LPS - VehNeh 1:1 SLDILPS — DMSO/SLDILPS 163 me del 5.4 He SL ad 5y Th RC Ne 9‘1 DE C) di membrane potential (Fig. 5.4). SLD sulfide caused a concentration-related decrease in mitochondrial membrane potential. 5.4.5 Effect of SLD sulfide on the production of reactive oxygen species in HepGZ cells SLD sulfide increased DCF fluorescence in HepGZ cells at 0.5, 1 and 3 hr after addition (Fig.5.5), indicating that SLD sulfide induced ROS production. TNF plays a critical role in the pathogenesis of SLDILPS-induced liver injury in part by synergistically killing hepatocytes with SLD sulfide (Zou et al., 2009a). However, TNF alone had no effect on DCF fluorescence, and the effect of SLD sulfide on ROS generation was not influenced by TNF. 5.4.6 Cytotoxicity of hydrogen peroxide and TNF to HepGZ cells Neither TNF (200ng/mL) nor hydrogen peroxide (up to 1 mM) was toxic to HepGZ cells (Fig. 5.6). At 2 mM, hydrogen peroxide caused very modest cytotoxicity. When HepG2 cells were treated with TNF (200ng/mL) and hydrogen peroxide (1-2 mM) together, significant cytotoxicity occurred, as marked by a pronounced increase in LDH release. This indicated that TNF enhanced the cytotoxicity of hydrogen peroxide. 5.4.7 Effect of antioxidant treatment and caspase inhibition on cytotoxicity SLD sulfide (250 uM) killed HepG2 cells (Fig. 5.7). Tempol, which is a superoxide dismutase mimetic, reduced the cell death caused by SLD sulfide. As shown 164 Fig. 5.4. Effect of SLD and its metabolites on mitochondrial membrane potential. Mitochondria were isolated from the livers of normal rats and incubated with SLD or its metabolites, SLD sulfone or SLD sulfide, for 30 min. Mitochondiral membrane potential was evaluated. *Significantly different from any other group at the same concentration. #Significantly different from vehicle group. P<0.05, n=3. 140 1 -O- SLD -O- SLD sulfone 120 ‘ + SLD sulfide —L O O co 0 60- JC-1 aggregates/m onomers (% control) 4O . . . . 0.00 0.25 0.50 0.75 1.00 Concentration (mM) 165 Fig. spec in it spec eval the : Fig. 5.5. Effect of SLD sulfide and TNF on production of reactive oxygen species in Hesz cells. SLD sulfide (250 uM) was administered to HepGZ cells in the presence of TNF (200nglmL) or its medium vehicle. Reactive oxygen species generation at various times (0, 0.5, 1 and 3 hr) after treatment was evaluated using DCF fluorescence. *Significantly different from VehNeh group at the same time. P<0.05, n=4. + VehNeh —O— Sulfide/veh + Veh/T NF 80 w —A— Sulfide/1' NF 2: ._ * * g 75 * E 70 - * * g 65 « 5 o 60 ‘ m 2 55 - 3 E 50 - 3 45 . D 40 . . . . 0 0.5 1 3 Hr after treatment 166 Fig. 5.6. Cytotoxicity induced by hydrogen peroxide and TNF. Hydrogen peroxide was administered to HepG2 cells in the presence of TNF (200nglmL) or its medium vehicle. After 24 hr incubation, the percentage of LDH released from cells was evaluated as a marker of cell injury. *Significantly different from 0 concentration hydrogen peroxide group. #Significantly different from corresponding hydrogen peroxide/vehicle group. P<0.05, n=4. 80- +Veh *# .5 8 «I 60‘ 2 at h 40 ,,\° *# * E 20- ..l 0 ‘1 0.0 0.5 1.0 1.5 2.0 2.5 Hydrogen peroxide (mM) 167 Fig. 5 TNF. admin percel diffe re group Fig. 5.7. Effect of antioxidant on cytotoxicity induced by SLD sulfide and TNF. Tempol was administered to HepG2 cells half an hour before the administration of SLD sulfide (250 uM), TNF (200nglmL) or their vehicles. The percentage of LDH activity released was determined after 24 hr. *Significantly different from SLD sulfide group. #Significantly different from SLD sulfide/TNF group. P<0.05, n=5. or Q G O LDH (% released) .h O 20 1 0 SLD sulfide - - - + + + + + + TNF - - - - - - + + + Tempol - 0.2 1 - 0.2 1 - 0.2 1 168 previously (Zou et al., 2009a), a nontoxic concentration of TNF (see Fig. 5.6) significantly enhanced the cytotoxicity of SLD sulfide. Tempol decreases the cytotoxicity due to the interaction of SLD sulfide and TNF. 5.4.8 Effect of antioxidant treatment on caspase 3” activity induced by SLD sulfide and TNF cotreatrnent Either SLD sulfide or TNF alone at the concentrations used did not increase caspase 3/7 activity in HepGZ cells (Fig. 5.8). In contrast, coadministration of SLD sulfide and TNF activated caspase 3/7 activity at 6 hr. Pretreatment of HepGZ cells with antioxidant tempol or a pancaspase inhibitor Z-VAD abolished the increase in caspase 3/7 activity caused by SLD sulfide and TNF cotreatrnent. Z-VAD failed to protect HepG2 cells from the cytotoxic effect of SLD sulfide when it was given alone but reduced the cytotoxic effect of SLD sulfide/T NF coadministration (Fig. 5.9). 169 Fig. 5.8. Effect of antioxidant on caspase 3” activity. Tempol (200 uM), Z- VAD (10 uM) or their medium vehicle was administered to HepGZ cells. Half an hour later, SLD sulfide (250 uM) and/or TNF (200 ug/mL) was administered. Acitivity of caspase 3/7 in HepGZ cells was determined after 6 hr incubation. *Significantly different from any other group. P<0.05, n=4. 7. I— 6‘ >A 6315‘ (BC :2“ €00 09-5 3‘ 85 au- 2‘ mv :3 1- 0‘ SLDsquide - + - + + + TNF - - + + + + Tempol - - - - + - Z-VAD - - - - - 4. 170 Fig. 5.9. Effect of pan-caspase inhibitor on cytotoxicity induced by SLD sulfide and TNF. Z-VAD (10 uM), SLD sulfide, TNF or their vehicles were administered to HepG2 cells as described in Fig.5.8. The percentage of LDH activity released from HepG2 cells was determined after 24 hr. *Significantly different from corresponding SLD sulfide-free group. #Significantly different from corresponding SLD sulfide/T NF group in the absence of Z-VAD. P<0.05, n=3. -e— VehNeh * .3 80 , -O— TNFNeh a: + VehIZ-VAD # m + TNFIZ-VAD <0 . 2 60 Q) " 4o - °\° *# V :l: 20 i Q E _I 0 - / L. . . 0 150 200 250 SLD sulfide concentration (uM) 171 5.5 Discussion SLD/LPS cotreatrnent induced liver injury in rats, whereas the doses of either SLD or LPS employed were not hepatotoxic when given alone. To investigate the interaction between SLD and LPS, gene array analysis was performed on livers collected at 4 hr, a time before the onset of liver injury. Gene expression changes caused by SLD, LPS and SLD/LPS were compared. The results suggested that SLD alone had only a modest effect on gene expression (Fig. 5.1). As expected, LPS treatment caused the changes in the expression of numerous genes. Interestingly, SLD interacted with LPS to cause a large number of genes to be expressed specifically after SLD/LPS cotreatrnent. The pathway most impacted by SLDILPS is fatty acid metabolism (Table 5.1). All the 12 genes in that category are downregulated, and half of them (ACSL1, AUH, ACAD9, ADHFE1, HSD17B4, GCDH) locate in the mitochondria, which might indicate an impaired function of mitochondria. Several LPS related pathways (LPS/lL-1-mediated inhibition of RXR function, NFkB signaling pathway) are highly impacted by SLD/LPS cotreatrnent. These results suggest SLD has an effect on the signaling pathway driven by LPS. Ingenuity pathway analysis indicated that genes related to oxidative stress were influenced by cotreatrnent. Two important genes involved in detoxification of reactive oxygen species (glutathione peroxidase 3 and glutathione S-transferase mu 2) were significantly downregulated by SLD/LPS. Glutathione peroxidase partially determines the susceptibility of cells to oxidative stress (Yang et al., 2006), and downregulation of glutathione S-transferase mu 2 has been associated with 172 mane SUQQ befi> For repe asa m v 509 NS; eat an obs of ghi Dh 0m rat inj ek Ti increased levels of superoxide anion (Zhou et al., 2008). Accordingly, this result suggests that the cellular defense system against oxidative stress was impaired before the onset of liver injury. Oxidative stress has been associated with numerous models of liver injury. For example, ROS play a role in liver injury induced by alcohol and ischemia- reperfusion (Wiseman, 2006; Cederbaum et al., 2009). Hepatotoxic drugs such as acetaminophen can induce oxidative stress in mouse liver and in hepatocytes in vitro (Adamson and Harman, 1993; Lores Arnaiz et al., 1995). It has been suggested that ROS play a role in the idiosyncratic liver injury caused by NSAle (Boelsterli, 2002). SLD induces oxidative stress in cultured cell lines (Seo et al., 2007; Park et al., 2008). However, SLD has not been reported to cause oxidative stress in an animal model. In the model of SLD-LPS interaction, an increase in protein carbonyl concentration in isolated liver mitochondria was observed at 4 hr (Fig. 5.2), which suggests oxidative stress was induced in livers of rats cotreated with SLD/LPS before the onset of liver injury. Ebselen is a glutathione peroxidase mimic, and DMSO is a scavenger of ROS. Although DMSO can suppress conversion of the prodrug sulindac to its bioactive sulfide metabolite (Swanson et al., 1983), both of these agents decreased ALT activity in rats treated with SLD/LPS, suggesting that oxidative stress contributes to liver injury in this model. Injured mitochondria can be a major source of ROS arising from leakage of electrons from the electron transport chain (Zorov et al., 2006; Orrenius, 2007). There are numerous reports that NSAle lead to mitochondrial dysfunction by 173 acting as mitochondrial uncouplers or causing mitochondrial membrane permeability transition pore opening (Moreno-Sanchez et al., 1999; Al-Nasser, 2000; Boelsterli, 2002). In isolated rat mitochondria, SLD sulfide decreased mitochondrial membrane potential, whereas SLD or SLD sulfone showed no effect (Fig. 5.4). Consistent with this result, a previous study using the JC-1 assay also suggested that SLD sulfide may lead to dissipation of mitochondrial membrane potential in HepG2 cells (Leite et al., 2006). Decreased ATP synthesis can be a direct consequence of decreased mitochondrial membrane potential, which might explain why SLD sulfide is more hepatotoxic compared to SLD or SLD sulfone (Zou et al., 2009a). SLD sulfide also induced ROS generation in HepGZ cells (Fig. 5.5). However, exposure to SLD by itself failed to increase liver protein carbonyl concentration in rats; coexposure to LPS was necessary for this effect. Hypoxia, which occurs in the livers of rats cotreated with SLD/LPS as a result of hemostasis (Zou et al., 2009b), is a potential contributor of oxidative stress in vivo (Arteel et al., 1999). Moreover, PMNs accumulate and become activated in the livers of rats cotreated with SLD/LPS and contribute to liver injury at least in part by releasing cytotoxic proteases. During PMN activation that results in protease release, , NADPH oxidase assembles on the PMN plasma membrane and ROS are generated (Dahlgren and Karlsson, 1999). Thus, activated PMNs might contribute to ROS generation and consequent oxidative stress in this model. 174 A previous study revealed that TNF potentiated the cytotoxicity of SLD sulfide in HepG2 cells and primary hepatocytes in vitro (Zou et al., 2009a). However, TNF had no effect on ROS generation induced by SLD sulfide in HepGZ cells (Fig. 5.5), indicating that TNF does not contribute to cell death by enhancing oxidative stress. Interestingly, TNF enhanced the cytotoxicity of hydrogen peroxide in HepGZ cells (Fig. 5.6). This is consistent with previous observations that hydrogen peroxide and TNF synergistically kill primary mouse and rat hepatocytes in vitro (lmanishi et al., 1997; Han et al., 2006). These results might explain the synergistic killing by SLD sulfide and TNF; that is, although TNF does not enhance SLD sulfide-induced oxidative stress, it does render cells sensitive to ROS-mediated cell killing. This is supported by the observation that antioxidant tempol decreased the cytotoxicity of SLD sulfide and significantly reduced the cytotoxic interaction between TNF and SLD (Fig. 5.7). In vivo, it seems unlikely that SLD sulfide concentrations become great enough to cause ROS-mediated liver injury; rather, TNF production caused by LPS coadministration renders the liver more sensitive to otherwise noninjurious ROS generation. The gene expression results discussed above suggest that compromised antioxidant protective mechanisms could play a role in this heightened sensitivity. JNK is a common target of TNF and ROS and regulates apoptosis (Kanda and Miura, 2004; Schwabe and Brenner, 2006). TNF-induced JNK activation leads to caspase activation, which in turn leads to liver injury (Wang et al., 2006). Activation of caspase 3f7 was only observed in HepG2 cells cotreated with SLD 175 sulfide and TNF (Fig. 5.8), whereas TNF or toxic concentration of SLD sulfide alone had no effect. This indicates that the cytotoxicity of SLD sulfide alone in vitro depended on ROS but not on caspase activation. However, both ROS and caspase activation were critical in the synergistic killing induced by TNF and SLD sulfide (Figs. 5.7 and 5.9). The observation that tempol reduced the activation of caspase 3l7 (Fig. 5.8) suggests ROS contribute to caspase activation induced by SLD sulfide/T NF interaction. In summary, this study further revealed the mechanisms of SLD/LPS- induced liver injury in rats. According to the comparison of gene expression in the liver of rats treated with SLD and/or LPS or their vehicles, genes associated with oxidative stress were selectively regulated by SLD/LPS. SLD/LPS cotreatment led to an increase in protein carbonyl in the mitochondria of rat livers, and antioxidants protected against liver injury, which suggests oxidative stress is involved in SLDILPS-induced liver injury. SLD sulfide exerts its cytotoxicity through decreasing mitochondrial membrane potential and increasing the production of ROS in vitro. The synergistical interaction of SLD sulfide and TNF to kill HepG2 cells is dependent on the oxidative stress induced by SLD sulfide. Under oxidative stress TNF leads to the activation of caspase 3/7, which contributes to the cytotoxicity of SLD sulfide/T NF interaction. 176 CHAPTER 6 Summary and conclusions 177 6.1 Summary and conclusions The goal of this project was to test the hypothesis that LPS precipitates SLD- induced liver injury in rats. A model of SLDILPS-induced liver injury was developed, and the mechanisms were further investigated. These studies provide additional evidence that supports the hypothesis that inflammation is a susceptibility factor for idiosyncratic drug hepatotoxicity. This project also enhances our understanding of the mechanisms of drug-LPS interaction. First, to develop a liver injury model of SLD and LPS interaction, rats were treated with two administrations of SLD with a 16 hr interval. Two administrations of SLD were chosen because treatment with one administration of SLD with LPS was not sufficient to induce liver injury in rats. Half an hour before the second administration of SLD, a nonhepatotoxic dose of LPS was administered to rats via a tail vein. The ALT activity in rat serum increased at 8 hr and was significant at 12 hr (Fig. 2.3). Midzonal necrosis was also observed only in the livers of rats cotreated with SLD/LPS (Fig. 2.5). These results suggest that SLD interacts with the inflammatory stress induced by LPS, which leads to liver injury. Subsequently, several mediators potentially involved in the pathogenesis were investigated. Hemostatic factors, including PAH and thrombin, were elevated by LPS in rat plasma (Fig. 2.6) and were enhanced by SLD cotreatment. As a result, significant fibrin deposition was observed in liver sinusoids of rats cotreated with SLDILPS compared to those treated with SLD or LPS alone (Fig. 2.9). Presumably resulting from the impaired blood flow, hypoxia occurred in the liver 178 of rats treated with SLD/LPS. Anticoagulant heparin, which attenuated fibrin deposition and hypoxia in the liver, protected against liver injury induced by SLD/LPS (Fig. 2.10 and 2.11). These results suggest that the hemostatic system and hypoxia resulting from fibrin deposition are critical to SLDILPS-induced liver injury. TNF is another mediator involved in the pathogenesis of liver injury induced by inflammation or inflammation-xenobiotic interaction. LPS led to an increase in TNF concentration in serum, which was enhanced by SLD cotreatrnent (Fig. 3.1). TNF neutralization using etanercept decreased serum ALT activity in rats cotreated with SLD/LPS, which shows that TNF plays an important role in the pathogenesis (Fig. 3.2). The concentrations of SLD, SLD sulfone and SLD sulfide in plasma and livers of rats were decreased by LPS cotreatrnent (Fig. 3.3 and 3.4). In contrast, their concentrations in rat GI tract and feces were increased by LPS (Fig. 3.5). SLD is bioactivated to SLD sulfide, and this metabolite was much more cytotoxic to HepG2 cells and primary rat hepatocytes than SLD in vitro (Fig. 3.6). However, in rats the amount of SLD sulfide produced was insufficient to cause liver injury by itself. Presumably, it is this bioactivated metabolite that acts synergistically with LPS to precipitate liver injury in vivo. In vitro, SLD sulfide synergistically interacted with TNF to kill cells, whereas SLD or SLD sulfone had no interaction with TNF (Fig. 3.7 and 3.8). PMNs are a critical contributor to liver injury in other drug/LPS interaction models. LPS led to PMN accumulation in the liver, and SLD enhanced the 179 accumulation of PMNs induced by LPS (Fig. 4.1). However, HOCI-protein adducts were increased only in the livers of rats treated with SLD/LPS, suggesting that SLD/LPS cotreatrnent causes PMN activation in the liver. Either anti-PMN serum or the PMN protease inhibitor, eglin C, attenuated liver injury, which suggests that proteases released from activated PMNs participate in the pathogenesis (Fig. 4.3 and 4.4). Previous studies also showed that proteases synergistically interacted with hypoxia to kill hepatocytes (Luyendyk et al., 2005). This same interaction might contribute to SLDILPS-induced liver injury. Several mediators are involved in PMN activation which is a prerequisite for protease release. Inhibition of TNF or PAI-1, both of which are important mediators in liver injury induced by SLDILPS, decreased HOCl-protein adducts but had no effect on PMN numbers in the liver (Fig. 4.5 and 4.6). Thus, PMN activation but not accumulation is dependent on TNF and PAl-1. Neutralization of TNF had no effect on PAI-1 production, suggesting an independent relation between TNF and PAl-1 (Fig. 4.7). SLD/LPS cotreatrnent produced a specific gene expression profile in the liver compared to treatments with SLD alone or LPS alone. Analysis of the genes specifically changed by SLD/LPS at 4 hr suggested that a pathway associated with oxidative stress is influenced before the onset of liver injury (Table 5.1). Antioxidants protected against liver injury induced by SLD/LPS in rats and reduced cytotoxicity caused by SLD sulfide and TNF interaction in vitro. These results suggest oxidative stress is also involved in the pathogenesis of SLD/LPS- induced liver injury. Moreover, ROS not only kills hepatocytes itself but also 180 interacts with other mediators. A study in vitro showed that TNF enhanced the cytotoxicity of hydrogen peroxide in HepG2 cells (Fig. 5.6). Mechanisms of SLDILPS-induced liver injury are summarized in Fig. 6.1. TNF, the hemostatic system, hypoxia, PMNs as well as reactive oxygen species play critical roles in the pathogenesis of SLDILPS-induced liver injury. SLD enhances the activation of hemostatic factors, including thrombin and PAl-1, induced by LPS. An activated hemostatic system leads to fibrin clot formation in liver sinusoids. As a result, the liver becomes hypoxic which renders hepatocytes susceptible to injury. SLD also enhances the elevation in TNF induced by LPS. LPS decreased the bioavailability of SLD in rats, but the liver is nevertheless apparently able to produce enough SLD sulfide to precipitate a toxic interaction with LPS. SLD sulfide synergistically kills hepatocytes along with TNF and hypoxia in LPS-cotreated rats. PMNs are another critical player in SLDILPS- induced liver injury. PMN accumulation in the liver was primarily induced by LPS, and this effect was enhanced by SLD. Activation of PMNs was only observed in livers of rats cotreated with SLDILPS. TNF and PAI-1 contribute to PMN activation independently. Activated PMNs release proteases which induce liver injury by itself or by interacting with hypoxia. 181 Fig. 6.1. Proposed pathway in the pathogenesis of SLDILPS-induced liver injury. See text for details. W PMN _. PMN _. Proteases accumulation activation Thrombin——-> Fibrin ———-e Hypoxia PAI-1 6.2 Comm inflammatiO So far. hypothesis injury induc humans. Th (TVX), suli characteristi' to various e: PreVIOL inflammage been evalue 0f gram-n1 lipoteichoic routes of d liver injury the Ollly m to indUCe ; a‘dminlstra RAN, investigate acilVation 6.2 Commonality and differences among liver injury models of drug- inflammation interaction So far, the evidence in animal models supporting the inflammatory stress hypothesis has been accumulating. In animals, inflammation precipitates liver injury induced by several drugs associated with idiosyncratic liver injury in' humans. These drugs include ranitidine (RAN), diclofenac (DCLF), trovafloxacin (TVX), sulindac (SLD), halothane (HAL) and chlorpromazine (CPZ). The characteristics and mechanisms involved in the pathogenesis have been studied to various extents, and the results are summarized in table 6.1. Previous studies suggest that inflammation induced by different kinds of inflammagens precipitates drug-induced liver injury. The inflammagens that have been evaluated so far in drug interaction models include the cell wall components of gram-negative bacteria (LPS), Gram-positive stimuli (a peptidoglycan- lipoteichoic acid (PGN-LTA) mixture) and the viral RNA mimetic (polyl:C). The routes of drug administration are different among the models, which suggest that liver injury is not dependent on a specific route of drug administration. SLD is the only model in which the drug was administered twice; one dose of SLD failed to induce a significant liver injury in rats. Times between drug and inflammagen administration needed to elicit a toxic response vary among different models. RAN, TVX and SLD models are three that have been relatively well investigated using LPS as an inflammatory stimulus. Enhanced TNF production, activation of the hemostatic system and PMN accumulation in liver have been observed in these models. Neutralization or inhibition studies showed that TNF, 183 Table 6.1. Characteristics and underlying mechanisms of idiosyncratic liver injury models of drug-inflammation interaction. Y means the phenomenon or mechanism is observed in the model. N means the listed item is not true in the model. A blank entry means it has not been tested or reported. *indicates the inflammagen used in the mechanism studies. (See text for references) 184 RAN DCLF TVX SLD CPZ HAL LPS“ or Polyl:C* Inflammagen LPS LPS P GN+LT A LPS LPS or LPS Administration route of the i v p o i p p o i p i p drug . . . . . . . . . . . . ””"be' °f 9mg 1 1 1 2 1 1 administrations Time (hr) drug was given -15.5 - 2 —6 relative to LPS 2 2 3 and 0.5 Hr after the drug 12 ft administration when liver 6 6 12 a er 24 15 . . 2nd SLD Injury was observed Enhanced TNF production Y Y Y Y TNF Involved In Y Y Y Y pathogenesrs Hemostatic system activated Y Y Y Fibrin deposition and . . Y Y Y hypoxra involved PMN accumulation Y Y Y Y Y PMN activation Y Y PMNs involved In the Y Y Y Y pathogenesrs IFN / lL-18 dependent Y ROS involved Y Hemostasis affected by TNF Y Y N PMN accumulation N increased by TNF N N PMN accumulation affected . N by hemostasis PAl-1 increased by TNF Y Y N PMNs activated by PAH and Y Y TNF Kupffer cells and NK cells Y involved 185 hypoxia and PMNs are critical mediators in the pathogenesis of liver injury induced by the interaction with LPS for all three of these drugs. Their roles in DCLF, HAL and CPZ models need to be tested. These results suggest that TNF, hypoxia and PMNs are important players shared by many liver injury models of drug-inflammation interaction. These mediators are not independent but interact with each other. However, their interactions differ among models. Some mediators have been found to be critical in the pathogenesis in a single model but have not been tested in other models. For example, lFNy and lL-18 are both involved in TVX/LPS-induced liver injury. Oxidative stress proved to play a role in SLDILPS-induced liver injury. The roles of these mediators in other models need to be investigated. In summary, evidence is accumulating to support the hypothesis that inflammation precipitates or enhances drug-induced liver injury. TNF, the hemostatic system and PMNs are involved in the pathogenesis of all of the liver injury models of drug-inflammation interaction in which their roles have been tested. The roles of oxidative stress or proinflammatory cytokines (lFNy and IL- 18) and other potential mediators remain to be determined in other models. 186 6.3 Potential future studies This project generally clarified several important mediators involved in the liver injury induced by SLD/LPS coexposure. However, the mechanisms of SLD/LPS interaction remain incompletely understood. Potential directions for future studies of this idiosyncratic liver injury model are discussed below. Although it was observed that SLD enhances the appearance of TNF and hemostatic factors, the mechanisms have not been clarified. SLD has been reported to be toxic to the GI tract and to cause ulcers. A possible explanation for the effect of SLD on TNF, thrombin and PAI-1 is that SLD causes bacterial translocation to the liver resulting from gastrointestinal toxicity of SLD. In concert with this hypothesis, diclofenac at large dose exerts hepatotoxiticy to rats by causing bacterial translocation from the GI tract. Another possible mechanism is that SLD directly acts on the TLR4 signaling pathway. For example, ranitidine specifically enhanced the activation of p38, which led to enhanced TACE activation and TNF production. Therefore, the activity of signaling proteins in TLR4 pathway is worth evaluation. Activation of TACE is a potential target of SLD. The cellular sources of several mediators have not been identified. Kupffer cells are a major source of TNF in many inflammatory models. However, it is not known if they are the only source of TNF in the SLD/LPS model; hepatocytes and endothelial cells can also contribute to TNF production (Zhaowei et al., 2007). Kupffer cell depletion is a feasible approach to investigate the role of these cells in this model. The mechanisms by which the ROS are produced have not been 187 identified either. PMNs, Kupffer cells, hypoxia and TNF might all contribute to generation of ROS in the SLD/LPS model. Their roles can be investigated through their inhibition or depletion. The relation and interaction among these critical mediators are not fully understood. According to results in the ranitidine model, PMN proteases exacerbate fibrin deposition by upregulating PAl-1. ROS might contribute to the activation of PMNs (Jaeschke, 2006). In the TVX/LPS model, PAl-1 contributes to the production of proinflammatory cytokines. These interactions could be investigated in the SLD/LPS model. Despite of the discovery of various mediators including TNF, hypoxia, PMNs and ROS, there could be some participants yet to be discovered. IFNy, which was upregulated in plasma by SLD/LPS and plays a critical role in the TVX/LPS model, is a potential candidate. 188 APPENDICES Genes selectively regulated by SLDILPS cotretrnent. The number of gene expression changes (relative to VehNeh) caused by treatment with SLDNeh, Veh/LPS or SLDILPS were depicted using Venn diagram (Fig. 5.1). According to the diagram, there were 721 probe sets regulated by SLD/LPS that were not affected by either SLD or LPS treatment alone. The genes in this group were listed as below. ID :3: Symbol ID :23: Symbol 1 367589_at -2.674 A002 1 376009_at -2. 121 CTAGE5 1367614 J1 3.064 ANXA1 1376094_at -1.901 HINT3 1367636_at -1.696 IGF2R 1376105_at 4.540 COL14A1 1367672_at -1.542 HSD17B4 1376321_at 1.883 FAM38A 1367673_at -3.673 SELEN 8P1 1376337_at -1 .667 SMARCA2 1367695_at -1 .708 QDPR 1376569_at 1 .774 KLF2 1367721_at 1.563 3004 1376692_at 4.514 HIPK2 1367729_at -1 .814 OAT 1376706_at -2.293 TMEM47 1367750_at -2.102 PRPSAP1 1376715_at -1.671 CBARA1 1367771_at -2.808 TSC2203 1376727_at -1 .663 YIPF4 1367807_at -2.164 PLODl 1376758_at -2.106 ING1 136781 §_at -1.682 SLCSA6 1376771_at -1.797 PPM1 L 136781Lat -1.884 HDGF 1376796_at -1.664 RAB14 1367818_at -1.716 0003 1376862_at -2.386 UBE4B 1367857_at -4.297 FADS1 1376930_at -1.614 MRPL51 1 367869! at -2.537 OXR1 1377049_at -1.555 PNPLA7 1367874_at 3.210 RHOQ 1377060Aat -1.862 MCCC2 189 1367889_at -1.820 CAMK1 1377166_at -1 .522 ALSZ 1367896!at -2.700 CA3 1377209_at -1 .619 KLH L25 1367933_at -1 .658 AMD1 1377307_at 2.444 FAM89A 1367940_at 2.018 CXCR7 137760Lat -2.121 SNX24 1367998_at 2.628 SLPI 1377654_at -1 .605 FAM3A 1368021_at -1.676 ADH1 C 1377657_at -1.663 IBTK 1368057_at -2.056 ABCD3 1377745_at -1.751 LRRC40 1368067_gt -1.776 ZNF148 1377758_at -2.274 HSD17B13 136807 Lat -1 .775 MOSC2 1377810_at -1 .593 RALGPS2 1368085_at -1 .589 GCHFR 1377995_gt -1 .860 ITFGS 1368091_at -3.031 OPLAH 1378027_at -2.013 PVRL3 1368096_at -1 .693 RAB7L1 1378146_at -1 .741 T801 024 1368115_at -2.670 CLDN3 1 3781 8; at -1.556 NXT2 1368117_at -1.685 GPHN 1378394_at -1.819 MPPE1 1368122_at -2.453 RNF103 1378842_at -2.071 GABARAPL1 1 3681 31; at -1.598 MPDZ 1379044_at -1.894 LARP2 1368144_at 2.208 RG82 1379101!at -1.549 DHX36 1368183_at -1.787 PLCG1 1379315_at -1.664 RASSF7 1368215_at -2.300 TPP1 1379353_at -1.737 AASDHPPT 1368265!at -1 .802 CYP2T4 1 3793754 at 1 .601 PDGFA 1368272_at -3.178 GOT1 1379441_at -2.343 RNF160 1368277_at -2.073 PPP3CA 1379456_at ~2.233 MCART2 1 368378! at -1.699 ALDH1 L1 1379499_at 1 .965 LTB 1368387_at -1.990 BDH1 1379525_at -1.780 CRLS1 1368427_at -1.577 AKAP1 1 1379578_at -1.807 ZBTB20 1368435_at -4.049 CYP8B1 1379606__at -3.277 RAB30 1368437_at 2.305 CA4 1379645_at -1.859 PBRM1 190 1368446_at -2.661 SPINK1 1379784_at -1.796 PEX7 1368453Aat -2.931 FADSZ 1379794_at 2.159 GZMB 1368474_at 1.989 VCAM1 1379803_at 1 .675 LMO4 1368482_at 1.829 BCL2A1 1379850_at -2 . 093 PSMC6 1368509_at -1.541 3882 1379901_at -1.582 TBC1 D17 1368514_at -1.993 MAOB 1379909_at -1.968 GKAP1 1368519_at 4.112 SERPINE1 1379935_at 2.280 CCL7 1368545_at 1.614 CFLAR 1380063_at 3.086 CH25H 1368592_at 1 .688 1L1 A 1380229_at 1 .790 MAFF 1368657_at 3.211 MMP3 1381012_at -1.547 SERPINF1 1368702_at 1 .585 PAWR 1 381 193_at -1 .936 LPGAT1 136871 1_at -2.039 FOXA2 1381768_at -1 .523 MTHFS 1368733_at -1.784 SULT1 E1 1381973_at -1.569 SLC25A30 1368814_at -1 .592 ALDH6A1 1 382024_at 1 .906 DNAJ 36 13688603t 3.143 PHLDA1 1382101_at -1.722 HSZST1 1368862_at -1 .755 AKT1 1382150_at -1 .581 SLC25A46 1368869_at 3.134 AKAP12 1382200_at -2.381 CENPV 1368914_at 2.918 RUNX1 1382216_§t -1.574 GEMIN6 1368924_at -3.846 GHR 1 382274; at 1.703 RARRES1 1368960_at -1 .855 LGALSB 1382285_at -1 .656 NAGA 1369063_at -2.462 AN P32A 1382 325_at -1 .750 GCAT 1369069_at -2.219 AKAP1 1382332_at -1 .736 STAG2 1369070_at -1.549 PEX12 1382371_at -1 .538 DRAMZ 1369078_at -1.734 MAPK1 1382402_at -1 .961 ULK1 1369150_at -2.259 PDK4 1382496_at -1 .624 HN F4A 1369169_at -1.577 SLC23A1 1382602_at -1.696 UBR3 1369191_at 2.780 1L6 1382843_at -1.907 SGPL1 191 1369268_at 3.449 ATF3 1 382 935_at -2 . 046 KIAA0141 1369278_at -1.660 GNA12 1383004_at -1.550 AHCYL1 1369393_at 2.703 MAP3 K8 1383037_at -1 .693 POLDI P2 1369453_at -1.737 EPN1 1383050_at -2.286 CENPV 1369492_at -1.816 AADAC 1383118_at -1.901 TMEM209 1369654_at -2.490 PRKAA2 1383155_at -1.610 FAM1 17B 1369785_at -1.900 PPAT 1383159_at -1 .936 TOM1 L2 1369837_at -1.685 GULO 1383282_at -1 .689 THAP1 1 1369922_at -1.733 PLBDZ 1383358_at -2.712 AKAP1 1369926_at -1 .507 GPX3 1383359_at -1 .628 LNX2 1369931_at 1 .577 PKM2 1383395_at -1 .878 AGMAT 136993643t 1.687 CALM1 1383462_at -1 .678 RNF160 1 369942_at -1 .539 ACTN4 1 383463_at -1 .565 ZFP91 1369950_at -2.125 CDK4 1 383474_at 2.182 IRAK2 1369956_at -1.764 IFNGR1 1383732_at -3.490 BC021614 1369961Lat -1.593 FXYD1 1383863_at 1.529 LM02 1369982_at -1 .524 AP2A2 1383933_at -1 .607 KIAA0564 1369989_at -1.978 PNPO 1383960_at -1.712 PEX16 136999Lat -1 .577 DVL1 1384029_at -1 .646 XPA 1370029_at -1.643 CTBP1 1384131_at -2. 195 ATL2 1370036_at -2.348 SUOX 1384205_at -2. 590 NGLY1 1370047_at -1.679 EN PP1 1384254_at 2.056 OTUD1 1370067_at -2.173 ME1 1384293_at -1.962 CZOORF191 13701 12_at -1.865 PTEN 1384383_at -1 .700 AGPAT6 1370121_at -1.711 ADD1 1384628_at -1.722 IYD 137017Lat 2.146 PPP1R15A 1384903_at -2.443 GPT2 1370177_at 3. 345 PVR 1385160_at -1 .621 STAB2 192 1370190Lat 1.529 H3F3C 1385266!at -1.590 NLK 1370200_at -1.557 GLUD1 138556Lat 1.767 AKAP2 1370236_at -1.525 PPT1 1385690_at -1.948 MUT 1370249_at 2.047 TSPO 1385845_at -1.753 D730039F16R|K 1370285_at -1.690 CALCOCO1 1385889_at -1 .927 C20RF64 1370319_at -2.026 PPIF 1386280_at -1.574 METTL7B 1370322_at -1 .896 STK16 1 386764_at 1 .946 AKAP2 1370329_at -1 .957 CYPZDG 1386895_at -2. 147 MAGED1 1370334_at -1.877 PLEKHB1 138697Lat -3.225 CA3 1370359_at -1 .809 AMY2A 1387006_at -2.31 5 EG629219 1370360_at -1 .738 C3ORF34 1387018_at -1 .566 SORBSZ 1370375_at -2.340 GLSZ 138702Lat -1 .921 ALDH 1A1 1 370399_at -2.607 CYP4B1 1 387023_at -3. 142 GSTM2 1370501_at -1.843 UBEZG1 1387093_at -1.683 SLCO1 A2 1370516_at 1 .679 SLC15A3 1387094_at -2.694 SLCO1 A2 1370548_at -1.807 SLC16A10 1387186_at -1.732 RABQA 137080843t -2.317 CY85R3 1387188_at -1 .866 SLC17A1 1370814_at -1.501 DH RS4 1387190_at -1.542 DGKA 1370818_at -1.892 DECR2 1387209_at -2.259 SEC168 1370848_at 1.688 SLC2A1 1387214_at -1.997 E822 1370875_at 2.092 EZR 1387219_at 1 .927 ADM 1370881_at -1.504 TST 1387244_at -2.280 CGRRF1 1370891_at 1.770 CD48 1387296_at -2. 141 CYP2J2 1370905_at -1.728 DOCK9 1387314_at -1.763 SULT1B1 1370939!at -1.571 ACSL1 138737Lat -2.454 KHK 1371034_at -2.912 ONECUT1 138756Lat -1.863 SLCO1A1 137107Lat 1.819 ZBP1 1387652_at -1.638 IDE 193 1371317_at 4.606 L081 1367725_at -2041 GULO 1371322_at 1.956 LAMC1 1387782_at 4.676 DYNLL2 1371362_at 4.700 . KDELR3 1387786_at -1.610 MTPN 1371385_at 4.651 PSMG1 1387790_at -2.156 PAICS 1371388_at 4.576 PDHB 1387821_at 4.570 RAB3IP 1371400_at 2.932 THRSP 1387852_at 2.101 THRSP 1371404_at 4.935 EIF4B 1387857_at 4.707 srx7 1371405_at 4.671 CZORF64 1367661_at -1.693 AES 1371432_at 4.544 VAT1 1387864_at -1.936 KIDIN8220 1371443_at 4.626 C1ORF174 1387865_at 4.630 our 1371445_at 1.935 LRRC59 1387868_at 1.863 LBP 1371447_at 2.064 PLACB 1387900_at 4.926 CDIPT 1371460_at 4.646 C12ORF62 1387901_at -1.746 PTPRS 1371461_at 4.602 FAM548 1387920_at 4.505 MANZC1 1371464_at 4.542 ZFAND6 1387921_at 4.524 2031414 1371471_at 4.626 GLTSCR2 1387948_at 4.521 ICK 1371463_at 4.669 NNT 1387959_at -1.989 ASPG 1371493_at -2.337 AP2A2 1388136_at 4.522 TIMM9 1371525_at 4.666 SLC12A7 1388150_at 4.707 XPO1 1371527!at 1.766 EMP1 1388155_at 1.510 KRT18 1371531_at 4.564 LOC678880 1388167_at 4.991 NFIB 1371553_at 4.955 MRPL36 1388300_at -2.351 MGST3 1371560_at 4.656 IRF3 1388324_at -2.044 NIT1 1371571; at -2.016 MRPS36 1388338_Lat -2.086 PPP2R4 1371578_at 4.993 PRKACA 1388364 _at 4.576 NDUFS3 1371563_at 3.100 RBM3 1388382_at -1.605 C1ORF43 1371611_at 4.713 EXT2 1388404_at 4.673 GM12751 194 1371620_at 4.726 PRELID1 1388410_at 4.733 ucpz 1371634_at -2.053 TMEM126A 1388430_at 4.915 PTOV1 1371637_at 4.660 HP1BP3 1388441_at 4.920 LOC689574 1371668_at -3.060 RXRA 136647131 4.574 TCP11L2 1371697_at 4.641 PNPLA2 1388474_at 4.569 UBE2I 137171631 4.507 SMARCCZ 136646631 4.596 DPP8 137171731 4.751 MFN1 1388497_at 4.599 ACOT13 137174731 -2.748 PPDPF 136650331 -2.238 EID1 137179931 4.909 GAA 136650731 1.727 EIF6 1371835_at -2.283 PRKACB 136651631 4.902 FBXW5 137165331 4.631 MRPL42 1366533_a1 -2.024 crosm 137192031 4.604 POLDIP2 1388540_at 4.744 MAZ 1371948_at 4.972 PIGP 1388549_at 4.691 NCOA4 1371956_at -1.968 LOC683077 136656731 2072 THUMPD1 1371963_at 4.760 PCCA 1388617_at 4.766 BPHL 1371964_at -2291 GRSF1 1366636_a1 4.554 RNF167 1371982_at -2.173 DPY30 1388658_at 4.970 SURF2 1371983_at 4.763 JOSD1 136666631 1.796 ENC1 137199331 4.516 CPNE3 1388672_at -2.232 ZCCHC24 137207131 4.533 c0320 1388680_at 4.563 C1GALT1C1 137207331 4 .625 GATADZA 1388685_at 4.905 DGCR2 1372074_ at 4.752 NUDT3 1388725_at 4.622 LEPROT 1372085_at 4.959 ATL2 136673231 4.549 SLC35F5 137209931 4.663 FAM21A 136675531 4.966 SEC23A 137210231 4.667 C200RF191 136676331 -1.596 HMGB1 137212431 2491 EIF4B 1388809_at 4.757 SMPDL3A 137214931 4.664 AUH 136661531 4.933 SAPS1 195 1372158_at -1.531 MACROD1 1388817_at -1 .578 FAM63A 1372214_at -1.645 MRPS33 1388823_at -2.642 RABSB 1372217_at -1.727 TMEM199 1388831_at -1.735 SLCQA3R2 1372281_at -1.768 LYPLAL1 1388833Jt -1.548 POLE3 1372284_at -2.080 TRAPPC3 1388877_at -1 .718 MRPSS 1372286_at -1.558 TSPAN6 1388908_at -1.755 PECI 1372295_at -1.635 NARF 1388913_§t -1.978 PPAP2C 1372306_at -1.987 ETHE1 1388965_at -2.086 PPP2R5E 1372310_at -1 .667 |SOC1 1388976_at -1 .507 BOLA3 137237331 -1.894 CMBL 1388995_at -2.779 RNF14 1372389_at 2.456 IER2 1389072_at -1.532 MTMR4 1372394_at -2.382 HECTD1 1389128!at -2 .226 WDFY3 1372395_at -1 .632 MARCH6 1 389139Aat -2.060 TTC1 5 1372408_at -1 .731 GGA2 1389146_at -1 .547 FAM1 07B 1372409_at 1 .609 MADZL1 BP 1389167Aat -1 .643 MAPKAP1 1372421_at -1.772 AGA 1389176Aat -1.737 |NPP5F 1372426_at -2.115 ADAMTSL4 1389196_at -1.944 2310039H08R|K 1372459_at 1 .633 VASP 1389199_at -1 .647 CBORF58 1372463_at -2.216 FCH02 1389215_at -1.609 SEPHS1 1372469_at ~2.001 LOC68631 0 1 389253_at -2.605 VNN1 1372475_at -1.749 PINK1 1389329_at -1.533 LGALSB 1372507_at -1 .606 TCTA 1389338_at ~2.228 TMEM126B 137252Lat -2.098 FLCN 1389339_at -1.634 ARSA 1372562_at -1.682 C80RF82 1389351_at 1.854 LRRFIP1 1372571_at -1.793 MARCH2 1389358_at -1.931 LPGAT1 1372597_at -1.671 MRPL14 1389361L§t -1.829 ADI1 1372599_at -1.536 MGST2 1389386_at -1.585 CSORF23 196 1372612_at -2.414 DYNLL2 1389407_at -1 .626 DH RS1 1372624_at -1.590 AN06 1389538_at 2.149 NFKBIA 1372630_at -2.108 RADZ3A 1 389540_at -1 .747 LOC686590 1372650_at -1.765 DNMBP 1389548_at -1.613 ADHFE1 1372663_at -1 .616 PTDSSZ 1389567_at -2 . 071 SCAP 1 37272Q_ at -1.530 BTBD1 1389676_at -1.612 CCDC101 1 372723_ at -1 .588 IP09 1389738_at -1 .682 UNG 1372729_at 1 .727 PROCR 1389844_at -2.556 FKBP4 1372814_at -2.170 SFT2DZ 1389906_at -1 .775 FDFT1 1372819_at -1 .815 COG4 1389918_at -1 .726 LOC290704 1372828_at -1.733 MSRBZ 1389998_at -1 .543 NR2F2 1372835_at 1 .692 RHOJ 1390102_at -1 .910 DI RC2 1372845_at -1 .509 RPP21 1390189_at -1 .506 ZN F277 1372854_at -1 .653 TTC17 1390312_at 2.041 SAM 09L 1372860_at -1.576 LHPP 1390374_at -1 .650 FGFRL1 1372871_at -1 .877 C2ORF24 1390445_at -1 .669 LOC6881 33 1372888_at -1 .929 UBE4A 1390478_at -1 .942 ORC4 1372907_at -1 .912 ATP6VOE2 1 390526_at -1 .546 KLH L9 1372946_at 1 .654 CXORF4OA 1390591_at -1 .582 SLC1 7A3 1372947_at -1 .997 PLS3 1390699_at -1 .625 KIAA2026 1372996_at -2.171 L00684270 1390717_at -2.121 CRLS1 1373036_at -1.877 RGD1561455 1390819_at -1.696 TEF 1 373080; at -1.548 PAPOLA 1 390943_ at -2.030 C1 ORF63 1373145_at -1.640 VPS41 1390989_at -2.23O MOSPD2 1373157_at -1.817 USP47 139107Lat -1.707 RFC1 1373162_at -1.797 LOC681708 1391270_at -1.732 CNNM3 1373182_at -1.718 CLDN12 1391282_at -1.605 C60RF192 197 1373201_at -2.221 DBT 1391433_at -2.137 ACOT2 1373228_at -1.842 HGSNAT 1391483_at -1.992 CREB3L3 1373239_at -1 .879 SNX33 1391 507_at -2.042 ZN F467 1 373287_at -1 .829 ATOH8 1 391 527_at -1 .501 STAT6 1373305_at 1 .664 SNX4 1391702_at -1 .607 ZN F446 1 373389_at -1 .664 ACADQ 1 391 807_at -1 .756 TFCP2 1373409_at -1 .676 UBE3C 1392280_at 2.246 TLR2 1373426_at -1 .537 MAPK1 1392502_at -1 .940 AHCTF 1 1373450_at -1.847 USP38 1392534_at 1 .680 PMEPA1 1373469_at -2.332 RGD1565496 1392543_at -1 .903 RBBP4 1373492_at -1 .752 SDHAF2 1 392547_at 1 .538 C150RF48 1373502_at -1 .739 DYM 1392888_at 1 .624 GPC4 1373512_at -1.870 ILVBL 1392912Lat -1.800 CACYBP 1373523_at 2.919 FCGR3A 1392916_at -1 .827 MAP7 1373547_at -1.985 C7ORF25 1392929_at -1.642 CZOORF194 1373570_at -1.921 NPEPL1 1392955_at -1.793 SEL1 L 1373578_at -1.510 TRI M2 1 392975_ at -1 .765 LOC686393 1373625_at -1 .696 SHMT1 1 392979_at -1 .882 CACYBP 1373664_at -1.525 PIGC 1392984_at -1.544 CPN E3 1373686_at -1.864 SERPINA6 1393005_at -1 .500 SFT2 DZ 1373826_at -1.631 YPEL5 1393110_at -1.968 MPV17L 1373829_at 1.556 FGF R2 1393140!at 1.556 ZC3H12A 1373874_ at -1.871 SGPP1 1393171_at ~1.707 TMEM47 1373906_at -1.788 FAM173B 1393218_at -1.923 ATGZB 1373921_at -3.628 ECHDC3 1393351_at -1.635 RDH1O 1373923_at -1.736 RDH10 1393414_at -1.632 LOC679161 1373954_at -2.247 SU D83 1393615_at -1 .566 DEPDC6 198 1373984!at 1.500 SLC39A14 1393826_at -2.020 APON 1374039_at -1.558 CAR14 1393862_at -1.585 1700019617RIK 1374045_at -1.574 COGS 1394737_at -1.928 C90RF64 1374154_at -1.530 SFRSZIP 1395338_at -1.677 LRPPRC 1374303Lat -1.505 ALKBH2 1395565_at -1.691 COPS4 1374331_at 1.806 RQCD1 139561Lat -1.593 COPS4 1374396_at -1.733 ATP6V1C1 1396112_at -1.676 MTMR1O 1374467_at -1.621 TRAP1 1397268_at -1 .548 SLC17A4 1374487_at -1 .508 FAM96A 1397363_at -1 .514 PVR L3 1374554_at -1.718 C1 ORF128 1397419_at -1.633 MPP6 1374571_at -1.725 PIGX 1397519_at -1.562 ADI POR2 1374612_at -1 .966 PAPD5 1397526_at -1 .995 GCDH 1374669_at -1 .527 BRWD2 139752Lat -1 .512 DDX17 1375034_at -1.654 PLAZG15 1398249_at -1.764 SLCZ5A20 1375170_at 2.108 GM5068 1398282_at -3.201 KYNU 1375173_at -1.523 PLBD2 1398286_at -1.814 CSAD 1375298_at -1.664 PPPZRSC 1398295_at -1.525 SLC29A1 1375357_at -1.57O TOR1A 1398341_at -1.625 CISD3 1375429_at -1.864 ZH16 1398350_at 2.334 BASP1 1375431_at -1.752 C20RF69 1398472_at -1.651 C1 OORFZ 1375524_at -1.949 AR|D1A 1398514_at -2.151 HGD 1375536_at -1.919 NUMB 1398591_at 2.643 CCRL2 1375634_at -2.387 CCDC53 1398642_at -2.039 MTRR 1375638_§t -1.547 SDPR 1398808_at -1.999 IMPA1 1375869_at -2.210 ULK1 1398891_at -2.331 MRPL15 1375934_at -1.967 RNF128 1398902Aat -1.737 KIAA0664 1375951_at 1.799 THBD 1398976_at -1.956 C2OORF191 199 1375977_at -1.805 CETN2 , 139696131 -2.146 TRIAP1 200 BIBLIOGRAPHY Adachi M, Sakamoto H, Kawamura R, Wang W, lmai K and Shinomura Y (2007) Nonsteroidal anti-inflammatory drugs and oxidative stress in cancer cells. Histol Histopathol 22:437-442. Adamson GM and Billings RE (1992) Tumor necrosis factor induced oxidative stress in isolated mouse hepatocytes. Arch Biochem Biophys 294:223-229. Adamson GM and Billings RE (1993) Cytokine toxicity and induction of NO synthase activity in cultured mouse hepatocytes. Toxicol Appl Pharmacol 119:100-107. Adamson GM and Harman AW (1993) Oxidative stress in cultured hepatocytes exposed to acetaminophen. Biochem Pharmacol 45:2289-2294. Aithal GP (2004) Diclofenac-induced liver injury: a paradigm of idiosyncratic drug toxicity. Expert Opin Drug Saf 32519-523. Al-Nasser IA (2000) Ibuprofen-induced liver mitochondrial permeability transition. Toxicol Lett 1 1 12213-218. Arteel GE, Kadiiska MB, Rusyn 1, Bradford BU, Mason RP, Raleigh JA and Thurman RG (1999) Oxidative stress occurs in perfused rat liver at low oxygen tension by mechanisms involving peroxynitrite. Mol Pharmacol 55:708-715. Babior BM (1999) NADPH oxidase: an update. Blood 93:1464-1476. Baranes D, Matzner J and Razin E (1986) Thrombin-induced calcium- independent degranulation of human neutrophils. Inflammation 10:455-461. Barcia C, Fernandez Barreiro A, Poza M and Herrero MT (2003) Parkinson's disease and inflammatory changes. Neurotox Res 5:411-418. Barton CC, Barton EX, Ganey PE, Kunkel SL and Roth RA (2001) Bacterial lipopolysaccharide enhances aflatoxin B1 hepatotoxicity in rats by a mechanism that depends on tumor necrosis factor alpha. Hepatology 33:66-73. 201 Bedoucha M, Atzpodien E and Boelsterli UA (2001) Diabetic KKAy mice exhibit increased hepatic PPARgamma1 gene expression and develop hepatic steatosis upon chronic treatment with antidiabetic thiazolidinediones. J Hepatol 35:17-23. Beg AA and Baltimore D (1996) An essential role for NF-kappaB in preventing TNF-alpha-induced cell death. Science 274:782-784. Bjorge SM and Baillie TA (1991) Studies on the beta-oxidation of valproic acid in rat liver mitochondrial preparations. Drug Metab Dispos 19:823-829. Bock JM, Menon SG, Goswami PC, Sinclair LL, Bedford NS, Jackson RE and Trask DK (2007) Differential activity of sulindac metabolites against squamous cell carcinoma of the head and neck is mediated by p21waf1/cip1 induction and cell cycle inhibition. Cancer Biol Ther 6:30-39. Boelsterli UA (2002) Mechanisms of NSAID-induced hepatotoxicity: focus on nimesulide. Drug Saf 25:633—648. Bolder U, Trang NV, Hagey LR, Schteingart CD, Ton-Nu HT, Cerre C, Elferink RP and Hofmann AF (1999) Sulindac is excreted into bile by a canalicular bile salt pump and undergoes a cholehepatic circulation in rats. Gastmenterology 117:962-971. Boveris A and Cadenas E (1975) Mitochondrial production of superoxide anions and its relationship to the antimycin insensitive respiration. FEBS Lett 54:311- 314. Bradham CA, Qian T, Streetz K, Trautwein C, Brenner DA and Lemasters JJ (1998) The mitochondrial permeability transition is required for tumor necrosis factor alpha-mediated apoptosis and cytochrome c release. Mol Cell Biol 18:6353-6364. Braun NJ, Bodmer JL, Virca GD, Metz-Virca G, Maschler R, Bieth JG and Schnebli HP (1987) Kinetic studies on the interaction of eglin c with human leukocyte elastase and cathepsin G. Biol Chem Hoppe Seyler 368:299-308. Buchweitz JP, Ganey PE, Bursian SJ and Roth RA (2002) Underlying endotoxemia augments toxic responses to chlorpromazine: is there a relationship to drug idiosyncrasy? J Pharmacol Exp Ther 300:460-467. 202 Buck l, Morceau F, Grigorakaki C, Dicato M and Diederich M (2009) Linking anemia to inflammation and cancer: the crucial role of TNFalpha. Biochem Pharmacol 77:1 572-1579. Butterfield TA, Best TM and Merrick MA (2006) The dual roles of neutrophils and macrophages in inflammation: a critical balance between tissue damage and repair. J Athl Train 41:457-465. Canbay A, Feldstein AE, Higuchi H, Wemeburg N, Grambihler A, Bronk SF and Gores GJ (2003) Kupffer cell engulfment of apoptotic bodies stimulates death ligand and cytokine expression. Hepatology 38:1188-1198. Cederbaum Al, Lu Y and Wu D (2009) Role of oxidative stress in alcohol-induced liver injury. Arch Toxicol 83:519-548. Chen C, Edelstein LC and Gelinas C (2000) The Rel/NF-kappaB family directly activates expression of the apoptosis inhibitor Bcl-x(L). Mol Cell Biol 2022687- 2695. Cheng L, You O, Yin H, Holt M, Franklin C and Ju C (2009) Effect of polyl:C cotreatrnent on halothane-induced liver injury in mice. Hepatology 49:215-226. Cho JY (2007) lmmunomodulatory effect of nonsteroidal anti-inflammatory drugs (NSAIDs) at the clinically available doses. Arch Phann Res 30:64-74. Choi J and Cu JH (2006) Mechanisms of liver injury. I". Oxidative stress in the pathogenesis of hepatitis C virus. Am J Physiol Gastrointest Liver Physiol 290:G847-851 . Chojkier M (2005) Troglitazone and liver injury: in search of answers. Hepatology 41:237-246. Chosay JG, Essani NA, Dunn CJ and Jaeschke H (1997) Neutrophil margination and extravasation in sinusoids and venules of liver during endotoxin-induced injury. Am J Physiol272:G1195-1200. Clay KD, Hanson JS, Pope SD, Rissmiller RW, Purdum PP, 3rd and Banks PM (2006) Brief communication: severe hepatotoxicity of telithromycin: three case reports and literature review. Ann lntem Med 144:415-420. 203 Copple BL, Banes A, Ganey PE and Roth RA (2002) Endothelial cell injury and fibrin deposition in rat liver after monocrotaline exposure. Toxicol Sci 65:309-318. Copple BL, Moulin F, Hanumegowda UM, Ganey PE and Roth RA (2003) Thrombin and protease-activated receptor-1 agonists promote lipopolysaccharide-induced hepatocellular injury in perfused livers. J Pharmacol Exp Ther 305:417-425. Copple BL, Rondelli CM, Maddox JF, Hoglen NC, Ganey PE and Roth RA (2004) Modes of cell death in rat liver after monocrotaline exposure. Toxicol Sci 77:172- 182. Dahlgren C and Karlsson A (1999) Respiratory burst in human neutrophils. J lmmunol Methods 232:3-14. Daly AK, Aithal GP, Leathart JB, Swainsbury RA, Dang TS and Day CP (2007) Genetic susceptibility to diclofenac-induced hepatotoxicity: contribution of UGT287, CYP208, and ABCC2 genotypes. Gastroenterology 132:272-281. Davies NM and Watson MS (1997) Clinical pharmacokinetics of sulindac. A dynamic old drug. Clin Phannacokinet 32:437-459. De Marzo AM, Platz EA, Sutcliffe S, Xu J, Gronberg H, Drake CG, Nakai Y, lsaacs WB and Nelson WG (2007) Inflammation in prostate carcinogenesis. Nat Rev Cancer 7:256-269. Deng X, Lu J, Lehman-McKeeman LD, Malle E, Crandall DL, Ganey PE and Roth RA (2008) p38 mitogen-activated protein kinase-dependent tumor necrosis factor-alpha-converting enzyme is important for liver injury in hepatotoxic interaction between lipopolysaccharide and ranitidine. J Pharmacol Exp Ther 326:144-152. Deng X, Luyendyk JP, Ganey PE and Roth RA (2009) Inflammatory stress and idiosyncratic hepatotoxicity: hints from animal models. Pharmacol Rev 61 :262- 282. Deng X, Luyendyk JP, Zou W, Lu J, Malle E, Ganey PE and Roth RA (2007a) Neutrophil interaction with the hemostatic system contributes to liver injury in rats cotreated with lipopolysaccharide and ranitidine. J Pharmacol Exp Ther. 204 Deng X, Luyendyk JP, Zou W, Lu J, Malle E, Ganey PE and Roth RA (2007b) Neutrophil interaction with the hemostatic system contributes to liver injury in rats cotreated with lipopolysaccharide and ranitidine. J Pharmacol Exp Ther 3222852- 861. Deng X, Stachlewitz RF, Liguori MJ, Blomme EA, Waring JF, Luyendyk JP, Maddox JF, Ganey PE and Roth RA (2006) Modest inflammation enhances diclofenac hepatotoxicity in rats: role of neutrophils and bacterial translocation. J Pharmacol Exp Ther 319:1191-1199. Duggan DE (1981) Sulindac: therapeutic implications of the prodrug/pharmacophore equilibrium. Drug Metab Rev 12:325-337. Duggan DE, Hooke KF and Hwang SS (1980) Kinetics of the tissue distributions of sulindac and metabolites. Relevance to sites and rates of bioactivation. Drug Metab Dispos 8:241 -246. Eckel JE, Gennings C, Chinchilli VM, Burgoon LD and Zacharewski TR (2004) Empirical bayes gene screening tool for time-course or dose-response microarray data. J Biopharm Stat 14:647-670. Eckel JE, Gennings C, Themeau TM, Burgoon LD, Boverhof DR and Zacharewski TR (2005) Normalization of two-channel microarray experiments: a semiparametric approach. Bioinfonnatics 21 :1078-1083. Elmarakby AA and lmig JD (2009) Obesity is the major contributor to vascular dysfunction and inflammation in high fat diet hypertensive rats. Clin Sci (Lond). Etienne F, Resnick L, Sagher D, Brot N and Weissbach H (2003a) Reduction of Sulindac to its active metabolite, sulindac sulfide: assay and role of the methionine sulfoxide red uctase system. Biochemical and Biophysical Research Communications 31 2: 1005-1 01 0. Etienne F, Resnick L, Sagher D, Brot N and Weissbach H (2003b) Reduction of Sulindac to its active metabolite, sulindac sulfide: assay and role of the methionine sulfoxide reductase system. Biochem Biophys Res Commun 312:1005-1010. Fan M, Goodwin M, Vu T, Brantley-Finley C, Gaarde WA and Chambers TC (2000) Vinblastine-induced phosphorylation of Bel-2 and Bcl-XL is mediated by 205 JNK and occurs in parallel with inactivation of the Raf-1/MEKIERK cascade. J Biol Chem 275:29980-29985. Fan Y, Zhang W and Mulholland M (2005) Thrombin and PAR-1-AP increase proinflammatory cytokine expression in C6 cells. J Surg Res 129:196-201. Fassoulaki A, Eger El, 2nd, Johnson BH, Ferrell LD, Smuckler EA, Harper MH, Eger RR and Cahalan MK (1984) Brief periods of hypoxia can produce hepatic injury in rats. Anesth Analg 63:885-887. Fay WP, Garg N and Sunkar M (2007) Vascular Functions of the Plasminogen Activation System, in pp 1231-1237. Fernandez-Martinez E, Morales-Rios MS, Perez-Alvarez V and Muriel P (2004) lmmunomodulatory effects of thalidomide analogs on LPS-induced plasma and hepatic cytokines in the rat. Biochem Pharmacol 68:1321-1329. Galati G, Tafazoli S, Sabzevari O, Chan TS and O'Brien PJ (2002) Idiosyncratic NSAID drug induced oxidative stress. Chem Biol Interact 142225-41. Ganey PE, Luyendyk JP, Maddox JF and Roth RA (2004) Adverse hepatic drug reactions: inflammatory episodes as consequence and contributor. Chem Biol Interact 150235-51 . Garcia Rodriguez LA, Walker AM and Perez Gutthann S (1992) Nonsteroidal antilnflammatory drugs and gastrointestinal hospitalizations in Saskatchewan: a cohort study. Epidemiology 3:337-342. Garcia Rodriguez LA, Williams R, Derby LE, Dean AD and Jick H (1994) Acute liver injury associated with nonsteroidal anti-inflammatory drugs and the role of risk factors. Arch Intem Med 154:311-316. Geier A, Dietrich CG, Voigt S, Kim SK, Gerloff T, Kullak-Ublick GA, Lorenzen J, Matem S and Gartung C (2003) Effects of proinfiammatory cytokines on rat organic anion transporters during toxic liver injury and cholestasis. Hepatology 38:345-354. Gillis S, Furie BC and Furie B (1997) Interactions of neutrophils and coagulation proteins. Semin Hematol 34:336-342. 206 Gomez-Gaviro MV, Gonzalez-Alvaro I, Dominguez-Jimenez C, Peschon J, Black RA, Sanchez-Madrid F and Diaz-Gonzalez F (2002) Structure-function relationship and role of tumor necrosis factor-alpha-converting enzyme in the down-regulation of L-selectin by non-steroidal anti-inflammatory drugs. J Biol Chem 277:38212-38221. Gordon JR and Galli SJ (1990) Mast cells as a source of both preformed and immunologically inducible TNF-alpha/cachectin. Nature 346:274-276. Graham DJ, Green L, Senior JR and Nourjah P (2003) Troglitazone-induced liver failure: a case study. Am J Med 1 14:299-306. Granic l, Dolga AM, Nijholt IM, van Dijk G and Eisel UL (2009) Inflammation and NF-kappaB in Alzheimer's disease and diabetes. J Alzheimers Dis 16:809-821. Greaves RR, Aganlval A, Patch D, Davies SE, Sherman D, Reynolds N, Rolles K, Davidson BR and Burroughs AK (2001) Inadvertent diclofenac rechallenge from generic and non-generic prescribing, leading to liver transplantation for fulminant liver failure. Eur J Gastroenterol Hepatol 1 3:71 —73. Greijer AE and van der Wall E (2004) The role of hypoxia inducible factor 1 (HIF- 1) in hypoxia induced apoptosis. J Clin Pathol 57:1009-1014. Gujral JS, Hinson JA, Farhood A and Jaeschke H (2004) NADPH oxidase- derived oxidant stress is critical for neutrophil cytotoxicity during endotoxemia. Am J Physiol Gastrointest Liver Physiol 287:G243-252. Haasio K, Koponen A, Penttila KE and Nissinen E (2002) Effects of entacapone and tolcapone on mitochondrial membrane potential. Eur J Pharmacol 453221— 26. Han D, Hanawa N, Saberi B and Kaplowitz N (2006) Hydrogen peroxide and redox modulation sensitize primary mouse hepatocytes to TNF-induced apoptosis. Free Radic Biol Med 41 :627-639. Hasegawa T, Malle E, Farhood A and Jaeschke H (2005) Generation of hypochlorite-modified proteins by neutrophils during ischemia-reperfusion injury in rat liver: attenuation by ischemic preconditioning. Am J Physiol Gastrointest Liver Physiol 289:6760-767. 207 He K, Talaat RE, Pool WF, Reily MD, Reed JE, Bridges AJ and Woolf TF (2004) Metabolic activation of troglitazone: identification of a reactive metabolite and mechanisms involved. Drug Metab Dispos 32:639-646. Hehlgans T and Pfeffer K (2005) The intriguing biology of the tumour necrosis factor/tumour necrosis factor receptor superfamily: players, rules and the games. Immunology 11521-20. Henry D, Dobson A and Turner C (1993) Variability in the risk of major gastrointestinal complications from nonaspirin nonsteroidal anti-inflammatory drugs. Gastroenterology 1 05: 1 078-1088. Hewett JA and Roth RA (1995) The coagulation system, but not circulating fibrinogen, contributes to liver injury in rats exposed to lipopolysaccharide from gram-negative bacteria. J Pharmacol Exp Ther 272253-62. Hewett JA, Schultze AE, VanCise S and Roth RA (1992) Neutrophil depletion protects against liver injury from bacterial endotoxin. Lab Invest 66:347-361. Hiroshi Yasui MAKI (2003) Combination of tumor necrosis factor-alpha with sulindac augments its apoptotic potential and suppresses tumor growth of human carcinoma cells in nude mice. Cancer 97:1412-1420. Ho JS, Buchweitz JP, Roth RA and Ganey PE (1996) Identification of factors from rat neutrophils responsible for cytotoxicity to isolated hepatocytes. J Leukoc Biol 59:716-724. Hucker HB, Stauffer SC, White SD, Rhodes RE, Arison BH, Umbenhauer ER, Bower RJ and McMahon PG (1973) Physiologic disposition and metabolic fate of a new anti-inflammatory agent, cis-5-fluro-2-methyI-1-(p-(methylsulfinyl)- benzylidenyl)-indene-3-acetic acid in the rat, dog, rhesus monkey, and man. Drug Metab Dispos 12721-736. Imanishi H, Scales WE and Campbell DA, Jr. (1997) Tumor necrosis factor alpha alters the cytotoxic effect of hydrogen peroxide in cultured hepatocytes. Biochem Biophys Res Commun 230:120-124. lshisaka R, Sato N, Tanaka K, Takeshige T, lwata H, Klostergaard J and Utsumi T (1999) A part of the transmembrane domain of pro-TNF can function as a 208 cleavable signal sequence that generates a biologically active secretory form of TNF. J Biochem 126:413-420. Jaeschke H (2000) Reactive oxygen and mechanisms of inflammatory liver injury. J Gastroenterol Hepatol 15:718-724. Jaeschke H (2002) Neutrophil-mediated tissue injury in alcoholic hepatitis. Alcohol 27:23-27. Jaeschke H (2006) Mechanisms of Liver Injury. ll. Mechanisms of neutrophil- induced liver cell injury during hepatic ischemia-reperfusion and other acute inflammatory conditions. Am J Physiol Gastrointest Liver Physiol 290261083- 1088. Jaeschke H, Farhood A and Smith CW (1990) Neutrophils contribute to ischemia/reperfusion injury in rat liver in vivo. FASEB J 423355-3359. Jaeschke H, Fisher MA, Lawson JA, Simmons CA, Farhood A and Jones DA (1998) Activation of caspase 3 (CPP32)-like proteases is essential for TNF- aIpha-induced hepatic parenchymal cell apoptosis and neutrophil-mediated necrosis in a murine endotoxin shock model. J Immunol 160:3480-3486. Jaeschke H, Ho YS, Fisher MA, Lawson JA and Farhood A (1999) Glutathione peroxidase-deficient mice are more susceptible to neutrophil-mediated hepatic parenchymal cell injury during endotoxemia: importance of an intracellular oxidant stress. Hepatology 29:443-450. Jaeschke H and Liu J (2007) Neutrophil depletion protects against murine acetaminophen hepatotoxicity: another perspective. Hepatology 45:1588-1589; author reply 1589. Jaeschke H and Smith CW (1997) Mechanisms of neutrophil-induced parenchymal cell injury. J Leukoc Biol 61:647-653. Jones BE, Lo CR, Liu H, Pradhan Z, Garcia L, Srinivasan A, Valentino KL and Czaja MJ (2000) Role of caspases and NF-kappaB signaling in hydrogen peroxide- and superoxide-induced hepatocyte apoptosis. Am J Physiol Gastrointest Liver Physiol 278:G693-699. 209 Jungerrnann K and Kietzmann T (2000) Oxygen: modulator of metabolic zonation and disease of the liver. Hepatology 31:255-260. Kamata H, Honda S, Maeda S, Chang L, Hirata H and Karin M (2005) Reactive oxygen species promote TNFalpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases. Cal/1202649661. Kanda H and Miura M (2004) Regulatory roles of JNK in programmed cell death. J Biochem 136:1-6. Kaplowitz N (2005) Idiosyncratic drug hepatotoxicity. Nat Rev Drug Discov 4:489- 499. Kaplowitz N, Aw TY, Simon FR and Stolz A (1986) Drug-induced hepatotoxicity. Ann Intern Med 104:826-839. Kassahun K, Pearson PG, Tang W, McIntosh I, Leung K, Elmore C, Dean D, Wang R, Doss G and Baillie TA (2001) Studies on the metabolism of troglitazone to reactive intermediates in vitro and in vivo. Evidence for novel biotransfonnation pathways involving quinone methide formation and thiazolidinedione ring scission. Chem Res Toxicol14:62-70. Kim CH, Song KS, Kim KS, Kim JY, Lee BJ, Lee JG and Yoon JH (2005) Sulindac sulfide-induced apoptosis in sinonasal cancer cells. Acta Otolaryngol 125:201-206. Kitamura S and Tatsumi K (1982) In vitro metabolism of sulindac and sulindac sulfide: enzymatic formation of sulfoxide and sulfone. Jpn J Pharmacol 32:833- 838. Kitamura S, Tatsumi K and Yoshimura H (1980) Metabolism in vitro of sulindac. Sulfoxide-reducing enzyme systems in guinea pig liver. J Pharmacobiodyn 32290-298. Koch OR, Pani G, Borrello S, Colavitti R, Cravero A, Farre S and Galeotti T (2004) Oxidative stress and antioxidant defenses in ethanol-induced cell injury. Mol Aspects Med 25:191-198. 210 Krahenbuhl S, Brandner S, Kleinle S, Liechti S and Straumann D (2000) Mitochondrial diseases represent a risk factor for valproate-induced fulminant liver failure. Liver 20:346-348. Kujoth GC, Hiona A, Pugh TD, Someya S, Panzer K, Wohlgemuth SE, Hofer T, Seo AY, Sullivan R, Jobling WA, Morrow JD, Van Remmen H, Sedivy JM, Yamasoba T, Tanokura M, Weindruch R, Leeuwenburgh C and Prolla TA (2005) Mitochondrial DNA mutations, oxidative stress, and apoptosis in mammalian aging. Science 309:481-484. Kumashiro R, Kubota T, Koga Y, Tanaka M, Inada C, Kusaba N, Yoshida H, Hisamochi A, lde T, Tomita Y, Masumoto N, Tanikawa K, lga T and Sata M (2003) Association of troglitazone-induced liver injury with mutation of the cytochrome P450 2019 gene. Hepatol Res 26:337-342. Kwak SH, Wang XQ, He Q, Fang WF, Mitra S, Bdeir K, Ploplis VA, Xu 2, ldell S, Cines D and Abraham E (2006) Plasminogen activator inhibitor-1 potentiates LPS-induced neutrophil activation through a JNK-mediated pathway. Thmmb Haemost 95:829-835. Lasser KE, Allen PD, Woolhandler SJ, Himmelstein DU, Wolfe SM and Bor DH (2002) Timing of new black box warnings and withdrawals for prescription medications. Jama 287:2215-2220. Leite S, Martins NM, Dorta DJ, Curti C, Uyemura SA and Cardozo Dos Santos A (2006) Mitochondrial uncoupling by the sulindac metabolite, sulindac sulfide. Basic Clin Pharmacol Toxicol 99:294-299. Lemasters JJ, Ji S and Thurman RG (1981) Centrilobular injury following hypoxia in isolated, perfused rat liver. Science 213:661-663. Lenz A, Franklin GA and Cheadle WG (2007) Systemic inflammation after trauma. Injury 38: 1 336-1 345. Levi M, Keller TT, van Gorp E and ten Cate H (2003) Infection and inflammation and the coagulation system. Cardiovascular Research 60:26-39. Li AP (2002) A review of the common properties of drugs with idiosyncratic hepatotoxicity and the "multiple determinant hypothesis" for the manifestation of idiosyncratic drug toxicity. Chem Biol Interact 14227-23. 211 Lin JH, Yeh KC and Duggan DE (1985) Effect of uremia and anephric state on the pharmacokinetics of sulindac and its metabolites in rats. ll. Differential effects on the biliary excretion. Drug Metab Dispos 13:608-613. Linde A, Mosier D, Blecha F and Melgarejo T (2006) Innate immunity and inflammation - New frontiers in comparative cardiovascular pathology. Cardiovasc Res. Liu P, McGuire GM, Fisher MA, Farhood A, Smith CW and Jaeschke H (1995) Activation of Kupffer cells and neutrophils for reactive oxygen formation is responsible for endotoxin-enhanced liver injury after hepatic ischemia. Shock 3:56-62. Lores Arnaiz S, Llesuy S, Cutrin JC and Boveris A (1995) Oxidative stress by acute acetaminophen administration in mouse liver. Free Radic Biol Med 19:303- 310. Luster Ml, Germolec DR, Yoshida T, Kayama F and Thompson M (1994) Endotoxin-induced cytokine gene expression and excretion in the liver. Hepatology 1 92480-488. Luyendyk JP, Lehman-McKeeman LD, Nelson DM, Bhaskaran VM, Reilly TP, Car BD, Cantor GH, Deng X, Maddox JF, Ganey PE and Roth RA (2006) Coagulation-dependent gene expression and liver injury in rats given lipopolysaccharide with ranitidine but not with famotidine. J Pharmacol Exp Ther 317:635-643. Luyendyk JP, Maddox JF, Cosma GN, Ganey PE, Cockerell GL and Roth RA (2003) Ranitidine treatment during a modest inflammatory response precipitates idiosyncrasy-Iike liver injury in rats. J Pharmacol Exp Ther 30729-16. Luyendyk JP, Maddox JF, Green CD, Ganey PE and Roth RA (2004) Role of hepatic fibrin in idiosyncrasy—like liver injury from lipopolysaccharide-ranitidine coexposure in rats. Hepatology 40:1342-1351. Luyendyk JP, Shaw PJ, Green CD, Maddox JF, Ganey PE and Roth RA (2005) Coagulation-mediated hypoxia and neutrophil-dependent hepatic injury in rats given lipopolysaccharide and ranitidine. J Pharmacol Exp Ther 314:1023-1031. 212 Macher E and Chase MW (1969) Studies on the sensitization of animals with simple chemical compounds. XII. The influence of excision of allergenic depots on onset of delayed hypersensitivity and tolerance. J Exp Med 129:103-121. Malle E, Hazell L, Stocker R, Sattler W, Esterbauer H and Waeg G (1995) Immunologic detection and measurement of hypochlorite-modified LDL with specific monoclonal antibodies. Arterioscler Thromb Vasc Biol 15:982-989. Malle E, Marsche G, Panzenboeck U and Sattler W (2006) Myeloperoxidase- mediated oxidation of high-density Iipoproteins: fingerprints of newly recognized potential proatherogenic Iipoproteins. Arch Biochem Biophys 445:245-255. Maniratanachote R, Shibata A, Kaneko S, Yamamori I, Wakasugi T, Sawazaki T, Katoh K, Tokudome S, Nakajima M and Yokoi T (2005) Detection of autoantibody to aldolase B in sera from patients with troglitazone-induced liver dysfunction. Toxicology 216215-23. Matzinger P (1994) Tolerance, danger, and the extended family. Annu Rev Immunol122991-1045. Micheau O, Lens 8, Gaide O, Alevizopoulos K and Tschopp J (2001) NF-kappaB signals induce the expression of c-FLIP. Mol Cell Biol 21:5299-5305. Migita K, Abiru S, Nakamura M, Komori A, Yoshida Y, Yokoyama T, Daikoku M, Ueki T, Takii Y, Yano K, Yastuhashi H, Eguchi K and lshibashi H (2004) Lipopolysaccharide signaling induces serum amyloid A (SAA) synthesis in human hepatocytes in vitro. FEBS Lett 569:235-239. Milhoan KA, Lane TA and Bloor CM (1992) Hypoxia induces endothelial cells to increase their adherence for neutrophils: role of PAF. Am J Physiol 2632H956- 962. Mingatto FE, dos Santos AC, Rodrigues T, Pigoso AA, Uyemura SA and Curti C (2000) Effects of nimesulide and its reduced metabolite on mitochondria. Br J Pharmacol131:1154-1160. Moreno-Sanchez R, Bravo C, Vasquez C, Ayala G, Silveira LH and Martinez- Lavin M (1999) Inhibition and uncoupling of oxidative phosphorylation by nonsteroidal anti-inflammatory drugs: study in mitochondria, submitochondrial particles, cells, and whole heart. Biochem Pharmacol 57:743-752. 213 Morgan ET (1989) Suppression of constitutive cytochrome P-450 gene expression in livers of rats undergoing an acute phase response to endotoxin. Mol Pharmacol 36:699-707. Morgan MJ, Kim YS and Liu ZG (2008) TNFalpha and reactive oxygen species in necrotic cell death. Cell Res 18:343-349. Mosesson MW, Siebenlist KR and Meh DA (2001) The structure and biological features of fibrinogen and fibrin. Ann N YAcad Sci 936:1 1-30. Nguyen C, Rose NR and Njoku DB (2008) Trifluoroacetylated IgG4 antibodies in a child with idiosyncratic acute liver failure after first exposure to halothane. J Pediatr Gastroenterol Nutr 47: 1 99-202. O'Connor N, Dargan PI and Jones AL (2003) Hepatocellular damage from non- steroidal anti-inflammatory drugs. Qim 96:787-791. O'Neill LA (2008) The interleukin-1 receptor/'1' oII-Iike receptor superfamily: 10 years of progress. Immunol Rev 226210-18. Okaya T and Lentsch AB (2003) Cytokine cascades and the hepatic inflammatory response to ischemia and reperfusion. J Invest Surg 16:141-147. Olefsky JM (2009) lKKepsilon: a bridge between obesity and inflammation. Cell 138:834-836. Ong MM, Latchoumycandane C and Boelsterli UA (2007) Troglitazone-induced hepatic necrosis in an animal model of silent genetic mitochondrial abnormalities. Toxicol Sci 97:205-213. Ong MM, Wang AS, Leow KY, Khoo YM and Boelsterli UA (2006) Nimesulide- induced hepatic mitochondrial injury in heterozygous Sod2(+/-) mice. Free Radic Biol Med 40:420-429. Orrenius S (2007) Reactive oxygen species in mitochondria-mediated cell death. Drug Metab Rev 392443-455. 214 Ostapowicz G, Fontana RJ, Schiodt FV, Larson A, Davem TJ, Han SH, McCashland TM, Shakil AO, Hay JE, Hynan L, Crippin JS, Blei AT, Samuel G, Reisch J and Lee WM (2002) Results of a prospective study of acute liver failure at 17 tertiary care centers in the United States. Ann Intern Med 137:947-954. Park JH, Kim EJ, Jang HY, Shim H, Lee KK, Jo HJ, Kim HJ, Yang SH, Jeong ET and Kim HR (2008) Combination treatment with arsenic trioxide and sulindac enhances apoptotic cell death in lung cancer cells via activation of oxidative stress and mitogen-activated protein kinases. Oncol Rep 20:379-384. Paulus HE (1982) FDA Arthritis Advisory Committee meeting. Arthritis Rheum 25:595-596. Pintucci G, Iacoviello L, Amore C, Evangelista V, Cerletti C and Donati MB (1992) Cathepsin G, a polymorphonuclear cell protease, affects the fibrinolytic system by releasing PAl-1 from endothelial cells and platelets. Ann N YAcad Sci 667:286—288. Qu B, Li QT, Wong KP, Tan TM and Halliwell B (2001) Mechanism of clofibrate hepatotoxicity: mitochondrial damage and oxidative stress in hepatocytes. Free Radic Biol Med 31:659-669. Ramaiah SK and Jaeschke H (2007) Role of neutrophils in the pathogenesis of acute inflammatory liver injury. Toxicol Pathol 35:757-766. Reers M, Smiley ST, MottoIa-Hartshorn C, Chen A, Lin M and Chen LB (1995) Mitochondrial membrane potential monitored by JC-1 dye. Methods Enzymol 260:406-417. Rensen SS, Slaats Y, Nijhuis J, Jans A, Bieghs V, Driessen A, Malle E, Greve JW and Buunnan WA (2009) Increased hepatic myeloperoxidase activity in obese subjects with nonalcoholic steatohepatitis. Am J Pathol175:1473-1482. Renton KW (2001) Alteration of drug biotransformation and elimination during infection and inflammation. Pharmacol Ther 92:147-163. Riedo FX, Munford RS, Campbell WB, Reisch JS, Chien KR and Gerard RD (1990) Deacylated lipopolysaccharide inhibits plasminogen activator inhibitor-1, prostacyclin, and prostaglandin E2 induction by lipopolysaccharide but not by tumor necrosis factor-alpha. J Immunol 144:3506-3512. 215 Roelofs JJ, Teske GJ, Bonta Pl, de Vries CJ, Meijers JC, Weening JJ, van der Poll T and Florquin S (2009) Plasminogen activator inhibitor-1 regulates neutrophil influx during acute pyelonephritis. Kidney Int 75:52-59. Roth RA, Harkema JR, Pestka JP and Ganey PE (1997) Is exposure to bacterial endotoxin a determinant of susceptibility to intoxication from xenobiotic agents? Toxicol Appl Pharmacol 147:300-31 1 . Roth RA, Luyendyk JP, Maddox JF and Ganey PE (2003) Inflammation and drug idiosyncrasyuis there a connection? J Pharmacol Exp Ther 30721-8. Rothe M, Wong SC, Henzel WJ and Goeddel DV (1994) A novel family of putative signal transducers associated with the cytoplasmic domain of the 75 kDa tumor necrosis factor receptor. Cell 78:681-692. Rothwell C, McGuire EJ, Altrogge DM, Masuda H and de la lglesia FA (2002) Chronic toxicity in monkeys with the thiazolidinedione antidiabetic agent troglitazone. J Toxicol Sci 27:35-47. Saheki T, Komorizono K, Miura T, lchiki H, Yagi Y and Hashimoto S (1990) Clearance of argininosuccinate synthetase from the circulation in acute liver disease. Clinical Biochemistry 23:139-141. Sakon S, Xue X, Takekawa M, Sasazuki T, Okazaki T, Kojima Y, Piao JH, Yagita H, Okumura K, 001 T and Nakano H (2003) NF-kappaB inhibits TNF-induced accumulation of ROS that mediate prolonged MAPK activation and necrotic cell death. EMBO J 22:3898-3909. Salgado A, Boveda JL, Monasterio J, Segura RM, Mourelle M, Gomez-Jimenez J and Peracaula R (1994) Inflammatory mediators and their influence on haemostasis. Haemostasis 24: 1 32-1 38. Sallie RW, McKenzie T, Reed WD, Quinlan MF and Shilkin KB (1991) Diclofenac hepatitis. Aust N Z J Med 21:251-255. Savage RL, Moller PW, Ballantyne CL and Wells JE (1993) Variation in the risk of peptic ulcer complications with nonsteroidal antiinflammatory drug therapy. Arthritis Rheum 36:84-90. 216 Schild L and Reiser G (2005) Oxidative stress is involved in the permeabilization of the inner membrane of brain mitochondria exposed to hypoxia/reoxygenation and low micromolar Ca2+. Febs J 272:3593-3601. Schleef RR, Bevilacqua MP, Sawdey M, Gimbrone MA, Jr. and Loskutoff DJ (1988) Cytokine activation of vascular endothelium. Effects on tissue-type plasminogen activator and type 1 plasminogen activator inhibitor. J Biol Chem 263:5797-5803. Schnebli HP, Seemuller U, Fritz H, Maschler R, Liersch M, Virca GD, Bodmer JL, Snider GL, Lucey EC and Stone PG (1985) Eglin c, a pharrnacologically active elastase inhibitor. Eur J Respir Dis Suppl 1 39:66-70. Schwabe RF and Brenner DA (2006) Mechanisms of Liver Injury. 1. TNF-alpha- induced liver injury: role of IKK, JNK, and ROS pathways. Am J Physiol Gastrointest Liver Physiol 2902G583-589. Semenza GL (2004) Hydroxylation of HlF-12 oxygen sensing at the molecular level. Physiology (Bethesda) 1 92176-182. Seo SK, Lee HC, Woo SH, Jin HO, Yoo DH, Lee SJ, An S, Choe TB, Park MJ, Hong SI, Park IC and Rhee CH (2007) Sulindac-derived reactive oxygen species induce apoptosis of human multiple myeloma cells via p38 mitogen activated protein kinase-induced mitochondrial dysfunction. Apoptosis 12:195-209. Serteser M, Koken T, Kahraman A, Yilmaz K, Akbulut G and Dilek ON (2002) Changes in hepatic TNF-alpha levels, antioxidant status, and oxidation products after renal ischemia/reperfusion injury in mice. J Surg Res 107:234-240. Sevinir B, Meral A, Gunay U, Ozkan T, Ozuysal S and Sinirtas M (2003) Increased risk of chronic hepatitis in children with cancer. Med Pediatr Oncol 40:104-110. Shaw PJ, Beggs KM, Sparkenbaugh EM, Dugan CM, Ganey PE and Roth RA (2009a) Trovafloxacin enhances TNF-induced inflammatory stress and cell death signaling and reduces TNF clearance in a murine model of idiosyncratic hepatotoxicity. Toxicol Sci. Shaw PJ, Ditewig AC, Waring JF, Liguori MJ, Blomme EA, Ganey PE and Roth RA (2009b) Coexposure of Mice to Trovafloxacin and Lipopolysaccharide, a 217 Model of Idiosyncratic Hepatotoxicity, Results in a Unique Gene Expression Profile and Interferon Gamma-Dependent Liver Injury. Toxicol. Sci. 107:270-280. Shaw PJ, Fullerton AM, Scott MA, Ganey PE and Roth RA (2009c) The role of the hemostatic system in murine liver injury induced by coexposure to lipopolysaccharide and trovafloxacin, a drug with idiosyncratic liability. Toxicol Appl Pharmacol 236:293-300. Shaw PJ, Ganey PE and Roth RA (2009d) Trovafloxacin enhances the inflammatory response to a Gram-negative or a Gram-positive bacterial stimulus, resulting in neutrophil-dependent liver injury in mice. J Pharmacol Exp Ther 330272-78. Shaw PJ, Ganey PE and Roth RA (2009e) Tumor necrosis factor alpha is a proximal mediator of synergistic hepatotoxicity from trovafloxacin/lipopolysaccharide coexposure. J Pharmacol Exp Ther 328262-68. Shaw PJ, Hopfensperger MJ, Ganey PE and Roth RA (2007) Lipopolysaccharide and trovafloxacin coexposure in mice causes idiosyncrasy-like liver injury dependent on tumor necrosis factor-alpha. Toxicol Sci 1002259-266. Shen ES, Garry VF and Anders MW (1982) Effect of hypoxia on carbon tetrachloride hepatotoxicity. Biochem Pharmacol 31:3787-3793. Shenton JM, Chen J and Uetrecht JP (2004) Animal models of idiosyncratic drug reactions. Chem Biol Interact 150253-70. Shenton JM, Teranishi M, Abu-Asab MS, Yager JA and Uetrecht JP (2003) Characterization of a potential animal model of an idiosyncratic drug reaction: nevirapine-induced skin rash in the rat. Chem Res ToxicoI1621078-1089. Shibayama Y (1987) Enhanced hepatotoxicity of endotoxin by hypoxia. Pathol Res Pract1822390-395. Smart SJ and Casale TB (1994) TNF-alpha-induced transendothelial neutrophil migration is IL-8 dependent. Am J Physio/2661233245. 218 Sneed RA, Grimes SD, Schultze AE, Brown AP and Ganey PE (1997) Bacterial endotoxin enhances the hepatotoxicity of allyl alcohol. Toxicol Appl Pharrnaco! 144277-87. Solomon KA, Covington MB, DeCicco CP and Newton RC (1997) The fate of pro-TNF-alpha following inhibition of metalloprotease-dependent processing to soluble TNF-alpha in human monocytes. J lmmunol15924524-4531. Stahlmann R (2002) Clinical toxicological aspects of fluoroquinolones. Toxicol Lett 1 27:269-277. Standiford TJ, Kunkel SL, Lukacs NW, Greenberger MJ, Danforth JM, Kunkel RG and Strieter RM (1995) Macrophage inflammatory protein-1 alpha mediates lung leukocyte recruitment, lung capillary leak, and early mortality in murine endotoxemia. J Immunol 1 55: 1 51 5-1524. Su GL (2002) Lipopolysaccharides in liver injury: molecular mechanisms of Kupffer cell activation. Am J Physio! Gastrointest Liver Physiol 2832G256-265. Sun Y, Chen J and Rigas B (2009) Chemopreventive agents induce oxidative stress in cancer cells leading to COX-2 overexpression and COX-2-independent cell death. Carcinogenesis 30:93-100. Suntres ZE and Shek PN (1996) Treatment of LPS-induced tissue injury: role of liposomal antioxidants. Shock 6 Suppl 12857-64. Swanson BN, Mojaverian P and Boppana VK (1983) Inhibition of sulindac metabolism by dimethyl sulfoxide in the rat. J Toxicol Environ Health 12:213-222. Takeda K, Kaisho T and Akira S (2003) Toll-like receptors. Annu Rev Immunol 21:335-376. Tarazi EM, Harter JG, Zimmerman HJ, Ishak KG and Eaton RA (1993) Sulindac- associated hepatic injury: analysis of 91 cases reported to the Food and Drug Administration. Gastroenterology 104:569-574. Teoh NC and Farrell GC (2003) Hepatotoxicity associated with non-steroidal anti- inflammatory drugs. Clin Liver Dis 7:401-413. 219 Tettey JN, Maggs JL, Rapeport WG, Pinnohamed M and Park BK (2001) Enzyme-induction dependent bioactivation of troglitazone and troglitazone quinone in vivo. Chem Res Toxicol142965-974. Thirunavukkarasu C, Watkins SC and Gandhi CR (2006) Mechanisms of endotoxin-induced NO, IL-6, and TNF-alpha production in activated rat hepatic stellate cells: role of p38 MAPK. Hepatology 44:389-398. Toumade H, Pelletier L, Pasquier R, Vial MC, Mandet C and Druet P (1990) D- penicillamine-induced autoimmunity in Brown-Norway rats. Similarities with HgCl2-induced autoimmunity. J Immunol 14422985-2991. Trunkey DD (1988) Inflammation and trauma. Arch Surg 12321517. Tsung A, Sahai R, Tanaka H, Nakao A, Fink MP, Lotze MT, Yang H, Li J, Tracey KJ, Geller DA and Billiar TR (2005) The nuclear factor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion. J Exp Med 201:1135-1143. Tukov FF, Luyendyk JP, Ganey PE and Roth RA (2007a) The Role of Tumor Necrosis Factor alpha in Lipopolysaccharide/Ranitidine-induced Inflammatory Liver Injury. Toxicol Sci. Tukov FF, Luyendyk JP, Ganey PE and Roth RA (2007b) The role of tumor necrosis factor alpha in lipopolysaccharide/ranitidine-induced inflammatory liver injury. Toxicol Sci 100:267-280. Tukov FF, Maddox JF, Amacher DE, Bobrowski WF, Roth RA and Ganey PE (2006) Modeling inflammation-drug interactions in vitro: a rat Kupffer cell- hepatocyte coculture system. Toxicol In Vitro 20:1488-1499. Uetrecht J (2006) Idiosyncratic Drug Reactions: Current Understanding. Annu Rev Pharmacol Toxicol. Uetrecht J (2007) Idiosyncratic drug reactions: current understanding. Annu Rev Pharmacol Toxicol 47:513-539. Uetrecht JP (1999) New concepts in immunology relevant to idiosyncratic drug reactions: the "danger hypothesis" and innate immune system. Chem Res Toxicol 1 22387-395. 220 Vandenabeele P, Goossens V, Beyaert R, Declercq W, Grooten J, Vanhaesebroeck B, Van de Craen M, Vercammen D, Depuydt B, Denecker G and et al. (1994) Functional requirement of the two TNF receptors for induction of apoptosis in PC60 cells and the role of mitochondria in TNF-induced cytotoxicity. Circ Shock 44:196-200. Vattanaviboon P, Seeanukun C, Whangsuk W, Utamapongchai S and Mongkolsuk S (2005) Important role for methionine sulfoxide reductase in the oxidative stress response of Xanthomonas campestris pv. phaseoli. J Bacteria! 187:5831-5836. Venezio FR, DIVincenzo C, Pearlman F and Phair JP (1985) Effects of the newer nonsteroidal anti-inflammatory agents, ibuprofen, fenoprofen, and sulindac, on neutrophil adherence. J Infect Dis 152:690-694. Vial T, Goubier C, Bergeret A, Cabrera F, Evreux JC and Descotes J (1991) Side effects of ranitidine. Dmg Saf6294-117. Vignati L, Turlizzi E, Monaci S, Grossi P, Kanter R and Monshouwer M (2005) An in vitro approach to detect metabolite toxicity due to CYP3A4-dependent bioactivation of xenobiotics. Toxicology 2162154-167. Vrij AA, Rijken J, van Wersch JW and Stockbrugger RW (2003) Coagulation and fibrinolysis in inflammatory bowel disease and in giant cell arteritis. Pathophysio! Haemost Thromb 33:75-83. Walker AM (1997) Quantitative studies of the risk of serious hepatic injury in persons using nonsteroidal antiinflammatory drugs. Arthritis Rheum 40:201-208. Wallace KB and Starkov AA (2000) Mitochondrial targets of drug toxicity. Annu Rev Pharmacol Toxicol 40:353-388. Wang Y, Singh R, Lefkowitch JH, Rigoli RM and Czaja MJ (2006) Tumor necrosis factor-induced toxic liver injury results from JNK2-dependent activation of caspase-8 and the mitochondrial death pathway. J Biol Chem 281215258- 15267. Watkins PB (2005) Idiosyncratic liver injury: challenges and approaches. Toxicol Pathol 3321-5. 221 Westrick RJ and Eitzman DT (2007) Plasminogen activator inhibitor-1 in vascular thrombosis. Curr Drug Targets 82966-1002. \Mseman A (2006) Oxygen-induced reperfusion-injury is caused by ROS: Amelioration is possible by recombinant-DNA antioxidant enzymes and mimics in selected tissues. Med Hypotheses 662329-331. Wullaert A, van Loo G, Heyninck K and Beyaert R (2007) Hepatic tumor necrosis factor signaling and nuclear factor-kappaB: effects on liver homeostasis and beyond. Endocr Rev 282365-386. Yamamoto K, lchijo H and Korsmeyer SJ (1999a) BCL-2 is phosphorylated and inactivated by an ASK1/Jun N-terrninal protein kinase pathway normally activated at G(2)!M. Mol Cell Biol 1928469-8478. Yamamoto Y, Yin MJ, Lin KM and Gaynor RB (1999b) Sulindac inhibits activation of the NF-kappaB pathway. J Biol Chem 274:27307-27314. Yang MS, Chan HW and Yu LC (2006) Glutathione peroxidase and glutathione reductase activities are partially responsible for determining the susceptibility of cells to oxidative stress. Toxicology 226:126-130. Yee SB, Hanumegowda UM, Hotchkiss JA, Ganey PE and Roth RA (2003) Role of neutrophils in the synergistic liver injury from monocrotaline and bacterial lipopolysaccharide exposure. Toxicol Sci 72:43-56. Yee SB, Kinser S, Hill DA, Barton CC, Hotchkiss JA, Harkema JR, Ganey PE and Roth RA (2000) Synergistic hepatotoxicity from coexposure to bacterial endotoxin and the pyrrolizidine alkaloid monocrotaline. Toxicol App! Pharmacol 166:173-185. Younis HS, Vickers T, Levin AA and Henry SP (2006) CpG and Non-CpG Oligodeoxynucleotides Induce Differential Proinflammatory Gene Expression Profiles in Liver and Peripheral Blood Leukocytes in Mice. J Immunotoxico! 3:57- 68. Zhang J, Chaluvadi MR, Reddy R, Motika MS, Richardson TA, Cashman JR and Morgan ET (2008) Hepatic flavin-containing monooxygenase gene regulation in different mouse inflammation models. Drug Metab Dispos:dmd.108.025338. 222 Zhang P, Xie M, Zagorski J and Spitzer JA (1995) Attenuation of hepatic neutrophil sequestration by anti-CINC antibody in endotoxic rats. Shock 4:262- 268. Zhaowei J, D'Souza MJ and Oettinger CW (2007) Reversal of LPS induced endothelial cell TNF synthesis and increased permeability with microencapsulated antisense oligomers to NF-kappaB. J Microencapsu! 24:596- 607. Zhou SG, Wang P, Pi RB, Gao J, Fu JJ, Fang J, Qin J, Zhang HJ, Li RF, Chen SR, Tang FT and Liu PO (2008) Reduced expression of GSTM2 and increased oxidative stress in spontaneously hypertensive rat. Mol Cell Biochem 309299- 107. long WX, Edelstein LC, Chen C, Bash J and Gelinas C (1999) The prosurvival Bel-2 homolog Bfl-1/A1 is a direct transcriptional target of NF-kappaB that blocks TNFalpha-induced apoptosis. Genes Dev 13:382-387. Zorov DB, Juhaszova M and Sollott SJ (2006) Mitochondrial ROS-induced ROS release: an update and review. Biochim Biophys Acta 1757:509-517. Zou W, Beggs KM, Sparkenbaugh EM, Jones AD, Younis HS, Roth RA and Ganey PE (2009a) Sulindac metabolism and synergy with TNF in a drug- inflammation interaction model of idiosyncratic liver injury. J Pharmacol Exp Ther. Zou W, Devi SS, Sparkenbaugh E, Younis HS, Roth RA and Ganey PE (2009b) Hepatotoxic interaction of sulindac with lipopolysaccharide: role of the hemostatic system. Toxicol Sci 108:1 84-193. 223 111111111111 ll 63 1293 030 I u H N" u E“ “I H