a“ unhwa-tmrsh «5.1 ‘30» .. win. I. . ymfikfim: hvdwvfla. . of:§., ..~..!a.ruh. t: hvmfifiwmuh. A. 4.. 10, u. L 35,. afifiia. an»... tn§.ealh.flru (3. if... {Lia 5.7.3.1.? 34%..“ Am. . - r 5n! .1 in: sunniéflmm \. . {3513. {4... 1...! .411; 1'!) if. .f 200% LIBRARY Michigan State University This is to certify that the dissertation entitled INFLAMMATION AND IDIOSYNCRATIC DRUG REACTIONS: INFLAMMATORY MECHANISMS AND INTERACTIONS IN A MURINE MODEL OF TROVAFLOXACIN HEPATOTOXICITY presented by Patrick Joseph Shaw has been accepted towards fulfillment of the requirements for the Ph.D. degree in Pharmacology & Toxicology ”lbw A3231") Major Professor’s Sipnature ?.ob~©? 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 2/05 cJClR-C-lfiateouejndd-p. 15 H——_—.— INFLAMMATION AND IDIOSYNCRATIC DRUG REACTIONS: INFLAMMATORY MECHANISMS AND INTERACTIONS IN A MURINE MODEL OF TROVAFLOXACIN HEPATOTOXICITY BY Patrick Joseph Shaw A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Pharmacology and Toxicology 2008 ABSTRACT INFLAMMATION AND IDIOSYNCRATIC DRUG REACTIONS: INFLAMMATORY MECHANISMS AND INTERACTIONS IN A MURINE MODEL OF TROVAFLOXACIN HEPATOTOXICITY BY Patrick Joseph Shaw Drug-induced liver injury is the leading cause of acute liver failure in the United States and is a major concern for both public health and the pharmaceutical industry. Idiosyncratic adverse drug reactions (lADRs), a rare form of drug-induced liver injury, have been the reason for the majority of postrnarket regulatory actions on drugs. The liver is often a target of lADRs. lADRs are characterized by the toxicity being unrelated to the pharmacology of the drug and do not demonstrate obvious dose or time dependence. The erratic occurrence and lack of mechanistic evidence makes lADRs very difficult to predict. Hepatotoxicity induced by the fluoroquinolone antibiotic trovafloxacin (TVX) exhibited these characteristics. The mechanism underlying TVX-induced idiosyncratic hepatotoxicity is unknown. We and others have hypothesized that an inflammatory stress, commonplace and erratic in people, could alter the threshold for toxicity of certain drugs precipitating an lADR. This dissertation tested the hypothesis that an inflammatory stress could precipitate idiosyncrasy—like TVX hepatotoxicity in mice. Administration of a nonhepatotoxic dose of TVX 3 h before a nonhepatotoxic dose of either lipopolysaccharide (LPS) or peptidoglycan-lipoteichoic acid mixture caused significant heIDatocelIular necrosis and apoptosis. Levofloxacin (LVX), a fluoroquinolone antibiotic without lADR liability in humans, did not interact with LPS to cause hepatotoxicity. The remaining studies focused on understanding the mechanisms underlying TVX/LPS-induced liver injury. Gene expression analysis at a time before the onset of liver injury segregated mice to their respective treatment groups. Therefore, gene expression analysis was able to distinguish TVX/LPS-treated mice from all other treatment groups. Furthermore, LPS-induced increases in TNFa, lFNy, thrombin activation, PAH and VEGF were enhanced by TVX. The progression of TVX/LPS-induced liver injury was dependent on PMN activation, TNFa, IFNy, thrombin activation, PAH and VEGF. Based on this finding, mice were killed at a time near the onset of liver injury to explore how these mediators of inflammation interact with one another and the cascade of events which leads to TVX/LPS-induced hepatotoxicity. TNFa, lFNy, PAH and VEGF potentially interacted to form several cycles of dysregulated inflammation. These potential vicious cycles of inflammation might be involved in TVX/LPS-induced liver injury. In summary, novel proinflammatory properties and potential cycles of inflammation were identified which might be involved in various models of inflammatory tissue injury. Additionally, these studies support the possibility of predicting and identifying mechanisms underlying lADRs by utilization of a drug/LPS coexposure model. Copyright by PATRICK JOSEPH SHAW 2008 ACKNOWLEDGMENTS There are a lot of people who have helped me along the way and I would like to thank them for their contributions. No matter how many times I edit or rewrite this section I will not be able to express how grateful I truly am to everyone who has assisted me. First, and foremost, I would like to thank my mentor, Dr. Robert Roth. The decision to come to MSU and in turn, to join Bob's laboratory has been one of the best decisions I have ever made. As a result of working and interacting daily with Bob, I have developed into not only a better scientist, but also a better person. I have truly been lucky to have an advisor who has been an amazing mentor and a great friend. There are few things I will cherish more from MSU than Friday night happy hours for nachos and beer. I truly admire Bob’s passion for science; and only hope that I can carry this eagerness for science with me throughout my career. Dr. Patricia Ganey also deserves my gratitude. Patti played a significant role in my development as a scientist and my education in the laboratory. She has been a mentor to me throughout my graduate career. I have been extremely fortunate to have two truly amazing mentors to be there for me. There is really nothing I can say or do to truly express how much I appreciate what Bob and Patti have done for me as a scientist and a person. In addition to Bob and Patti, there have been several people at MSU who have helped me along the way. I owe a great deal of appreciation to my other committee members, Dr. Lapres and Dr. Kaminski, for their continued guidance and advice regarding my dissertation. Within the laboratory, Dr. Jane Maddox and former research assistant Sandra Newport helped me throughout my research. Jane and Sandy trained me and always were available for questions, even the really dumb ones. Dr. James Luyendyk, a former graduate student in the lab, has also played a significant role in my research. He helped train me in the laboratory when I first joined, and put up with me as I stumbled through the first couple months in the laboratory. Jim taught me a lot in the short time I worked with him. His approach to research shaped my graduate career, and I am lucky to have had the chance to work with him. I would also like to thank the people who have either rotated or worked with me to contribute to the research presented in this dissertation. Marie Hopfensperger, Susan Achberger and Aaron Fullerton have all contributed to the studies presented in this dissertation, and I appreciate their contributions. Additionally, a dissertation is a team effort and I want to thank all of the current and former members of the laboratory. Dr. Bryan Copple, Dr. Xiaomin Deng, Erica Sparkenbaugh, Dr. Francis Tukov, Wei Zou, Dr. Steve Bezdecny, Christine Dugan, Jingtao Lu, Theresa Eagle, Emilie Evenson, Dr. Sachin Devi, Nicole Crisp and Allen Macdonald have all been of assistance to me and I am grateful for their contributions. The Department of Pharmacology and Toxicology also deserves tremendous thanks. Dr. Stephanie Watts and Dr. Keith Lookingland have been great leaders of the graduate program at MSU. Additionally, the administrative staff within the department has been a great help and always available to assist me. The Center for Integrative Toxicology also deserves my gratitude. Amy Swagart and Carol Chvojka have always been helpful to me with financial and . travel problems. Beyond MSU, I have had the opportunity to collaborate with a number of exceptional scientists. l have worked closely with colleagues at Abbott Laboratories. I owe thanks to Dr. Eric Blomme, Dr. Jeff Waring, Michael Liguori and Amy Ditewig for their assistance and collaboration. Additionally, I owe a big thank you to Dr. Robert Stachlewitz who had me in his laboratory at Worcester, MA for a summer. Working with Rob at Abbott Laboratories was an amazing experience for which I am grateful. I am extremely grateful to the Society of Toxicology for everything it has offered me. Additionally, I owe a big thank you to the Michigan regional chapter of SOT. My experience within SOT has been a big part of my growth as a scientist. I have had the opportunity to work alongside exceptional scientists and people thanks to the SOT. I especially want to thank Betty Eidemiller, who assisted me in more ways than I can list during my time with SOT. Additionally, I would like to thank those who provided funding for my research. First off, thank you to Bob, who funded my studies through NIH grants. vi "“““l In addition, I would like to thank the Center for Integrative Toxicology, MSU, SOT and ASPET for the continued support in my graduate research and the attendance of scientific meetings. Finally, the only reason I was able to make this far was by the unwavering support and love from my family and friends. I owe a big thank you to some close friends: Brandon and Deirdre. Additionally, thank you to Sarah for being there for me during the writing of this dissertation, and understanding when I wasn’t in the best mood. I have an amazing family who deserve a lot of my appreciation. My parents have been there for me throughout, and I owe everything to them. I am eternally grateful. They have always dropped everything to help me, and I truly appreciate the sacrifices they made for me to get where I am today. Thank you Mom and Dad. Additionally, my step—parents, Greg and Carol, have always been supportive and willing to help [me out, and I am grateful. Furthermore, I would like to thank Eric, my brother and best friend. He has always been there to hear about the frustrations and encourage me. I wish I could put into words how much he has helped me. I don’t want to get all emotional, but I would not be anywhere near where I am today without my brother. I especially want to thank Eric, Mom and Dad, without whom none of this would have been possible. vii TABLE OF CONTENTS LIST OF TABLES .......................................................................... xiii LIST OF FIGURES ......................................................................... xiv LIST OF ABBREVIATIONS .............................................................. xviii CHAPTER 1 General Introduction and Specific Aims ....................................................... 1 1.1 Idiosyncratic adverse drug reactions ...................................... 2 1.1.1 Overview of idiosyncratic adverse drug reactions ....... 2 1.1.2 Hypothetical mechanisms of idiosyncratic adverse drug reactions .............................................................. 4 1.2 Inflammation ........................................................................... 20 1.2.1 Overview of inflammatory stress .................................. 20 1.2.2 Tumor necrosis factor a (TNFa) .................................. 27 1.2.3 Neutrophils ................................................................... 30 1.2.4 Interferon y (IFNy) ........................................................ 31 1.2.5 The hemostatic system ................................................ 34 1.2.6 Vascular endothelial growth factor (VEGF) ................. 38 1.3 Trovafloxacin-induced idiosyncratic liver injury ...................... 43 1.3.1 Overview of trovafloxacin ..................................... 43 1.3.2 Trovafloxacin-induced hepatotoxicity in people ..........44 1.3.3 Interaction between trovafloxacin and inflammation ...................................................... 46 1.4 Hypothesis and specific aims ......................................... 48 1.5 Overview and significance of dissertation .......................... 50 CHAPTER 2 Lipopolysaccharide and trovafloxacin coexposure in mice causes idiosyncrasy-Iike liver injury dependent on tumor necrosis factor-alpha ..... 52 2.1 Abstract ..................................................................... 53 2.2 Introduction ................................................................ 54 2.3 Materials and methods .................................................. 55 2.3.1 Materials ........................................................... 55 2.3.2 Animals ............................................................. 55 2.3.3 Experimental protocol .......................................... 56 2.3.4 Histopathology ................................................... 57 2.3.5 TNFa analysis ................................................... 57 2.3.6 Statistical Analyses ............................................. 57 2.4 Results ...................................................................... 58 2.4.1 Dose-response and timecourse of liver injury ............ 58 2.4.2 Comparison of trovafloxacin and levofloxacin ............ 58 2.4.3 Timecourse of TNFa concentration in plasma ........... 63 2.4.4 Pentoxifylline study .............................................. 68 viii 2.4.5 Etanercept inhibition of TNFa activity ...................... 73 2.5 Discussion .................................................................. 80 CHAPTER 3 Trovafloxacin enhances the inflammatory response to a gram- -negative or a gram- positive bacterial stimulus, resulting in CD18-dependent liver injury in mice ................................................................................. 86 3.1 Abstract ..................................................................... 87 3.2 Introduction ................................................................ 89 3.3 Materials and methods .................................................. 90 3.3.1 Materials ........................................................... 90 3.3.2 Animals ............................................................. 90 3.3.3 Experimental protocols ........................................ 90 3.3.4 Histopathology ................................................... 91 3.3.5 Cytokine measurements ....................................... 91 3.3.6 Neutrophil staining .............................................. 91 3.3.7 Statistical Analyses ............................................. 92 3.4 Results ...................................................................... 93 3.4.1 TVX coexposure with either LPS or PGN-LTA causes hepatotoxicity .......................................... 93 3.4.2 TVX enhances cytokine induction by either inflammatory stimulus .......................................... 93 3.4.3 Effect of TVX on microbial stimuli-induced hepatic neutrophil accumulation ........................................ 103 3.4.4 Effect of CD18 neutralization on TVX/LPS- and TVX/PGN-LTA-induced liver injury and inflammation ..103 3.5 Discussion .................................................................. 1 13 CHAPTER 4 TNFa acting at both p55 and p75 receptors is essential for synergistic hepatotoxicity from TVX/LPS coexposure ........................................... 119 4.1 Abstract ..................................................................... 120 4.2 Introduction ................................................................ 121 4.3 Materials and methods .................................................. 123 4.3.1 Materials ........................................................... 123 4.3.2 Animals ............................................................. 123 4.3.3 Experimental protocols ....................................... 123 4.3.4 Histopathology ................................................... 124 4.3.5 Cytokine measurements ....................................... 124 4.3.6 Neutrophil staining .............................................. 124 4.3.7 Hemostatic system measurements ......................... 124 4.3.8 Statistical analyses ............................................. 124 4.4 Results ...................................................................... 126 4.4.1 - p55"' and p75"' mice are protected from TVX/LPS- induced liver injury .............................................. 126 4.4.2 TNFa neutralization attenuates TVX/LPS-induced ix inflammatory cytokines and chemokines .................. 126 4. 4. 3 TVX/LPS-induced hepatic neutrophil accumulation is independent of TNFa ........................................ 131 4. 4. 4 TNFa neutralization attenuates TVX/LPS-induced hemostatic system activation ................................. 131 4.4.5 TVX and TNFa coexposure causes hepatotoxicity ..... 138 4.5 Discussion .................................................................. 143 CHAPTER 5 Coexposure of mice to trovafloxacin and lipopolysaccharide, a model of idiosyncratic hepatotoxicity, results in a unique gene expression profile and interferon gamma-dependent liver injury ....................................... 148 5.1 Abstract ..................................................................... 149 5.2 Introduction ................................................................ 150 5.3 Materials and methods .................................................. 152 5.3.1 Materials ........................................................... 152 5.3.2 Animals ............................................................. 152 5.3.3 Experimental protocols ......................................... 152 5.3.4 ALT activity and histopathology .............................. 153 5.3.5 RNA isolation ..................................................... 153 5.3.6 Gene array analysis ............................................ 153 5.3.7 Hepatic neutrophil accumulation ............................ 154 5.3.8 Plasma cytokine measurements ............................ 154 5.3.9 Statisitical methods ............................................. 155 5.4 Results ...................................................................... 156 5.4.1 Development of hepatocellular injury after TVX/LPS, but not LVX/LPS coexposure ................................. 156 5.4.2 Hepatic global gene expression changes before the onset of hepatotoxicity .......................................... 156 5.4.3 Gene expression changes within TVX/LPS, TVXNeh and Veh/LPS treatment groups ............................... 161 5.4.4 Several gene expression changes in TVX/LPS- treated mice are involved in interferon signaling ........ 161 5.4.5 Timecourse of plasma concentrations of IL-18 and IFNy ........................................................... 171 5.4.6 TVX/I7PS- induced hepatocellular Injury in lL-18"' mice ....................................................... 171 5.4.7 TVX/LPS- induced liver Injury in lFNy"' mice .............. 171 5.4.8 TVX/LPS-induced hepatic neutrophil accumulation and proinflammatory cytokines in IF Ny”' mice ........... 176 5.5 Discussion .................................................................. 183 CHAPTER 6 The role of the hemostatic system in a murine model of idiosyncratic liver injury induced by trovafloxacin and lipopolysaccharide coexposure ........... 200 6.1 Abstract ..................................................................... 201 6.2 6.3 6.4 6.5 CHAPTER 7 Introduction ................................................................ 203 Materials and methods .................................................. 205 6.3.1 Materials ........................................................... 205 6.3.2 Animals ............................................................. 205 6.3.3 Experimental protocols ......................................... 205 6.3.4 Heparin treatment ................................................ 205 6.3.5 Histopathology .. ............................................ 206 6.3.6 Hemostatic system measurements ......................... 206 6.3.7 Cytokine measurements ....................................... 206 6.3.8 Statisitical analyses ............................................. 206 Results ...................................................................... 207 6.4.1 Changes in the hemostatic system in TVX/LPS- induced liver injury ............................................... 207 6.4.2 Anticoagulant heparin protects from TVX/LPS- induced liver injury ............................................... 212 6.4.3 PAl-1"' mice are protected from TVX/LPS- induced liver injury ............................................... 219 6.4.4 Role of coagulation system activation and PAl-1 in TVX/LPS-induction of cytokines .............................. 219 Discussion .................................................................. 228 Vascular endothelial growth factor has a proinflammatory role which is critical to trovafloxacin and lipopolysaccharide coexposure- induced liver injury ......................................................................... 233 7.1 Abstract ..................................................................... 234 7.2 Introduction ................................................................ 235 7.3 Materials and methods .................................................. 236 7.3.1 Materials ........................................................... 236 7.3.2 Animals ............................................................. 236 7.3.3 Experimental protocols ......................................... 236 7.3.4 Histopathology .................................................... 237 7.3.5 Cytokine measurements ....................................... 237 7.3.6 Neutrophil staining ............................................... 237 7.3.7 Hemostatic system measurements ......................... 237 7.3.8 Statisitical analyses ............................................. 237 7.4 Results ...................................................................... 238 7.4.1 Timecourse of plasma concentration of VEGF ........238 7.4.2 Effect of VEGF neutralization on TVX/LPS- induced hepatotoxicity .......................................... 238 7.4.3 Effect of VEGF neutralization on plasma concentrations of cytokines ................................... 245 7.4.4 Effect of VEGF neutralization on TVX/LPS-induced . hepatic neutrophil accumulation .............................. 245 7.4.5 Effect of VEGF neutralization on the hemostatic system .............................................................. 245 xi 7.5 Discussion .................................................................. 255 CHAPTER 8 Summary and conclusions ............................................................... 259 8.1 Summary of research .................................................... 260 8.2 Proposed pathways of TVX/LPS-induced liver injury ........... 266 8.3 Major findings and implications ....................................... 273 8.4 Knowledge gaps and future studies ................................. 276 REFERENCE LIST ......................................................................... 278 xii Table 1.1 Table 1.2 Table 1.3 Table 1.4 Table 1.5 Table 5.1 Table 5.2 Table 5.3 Table 6.1 LIST OF TABLES Concordance of LPS/drug exposure in rats for lADR- causing drugs in humans ............................................... 19 The effects of TNFa on other mediators of liver injury .......... 29 The effects of IF Ny on other mediators of inflammatory liver injury .............................................................................. 33 The effects of the hemostatic system and hypoxia on mediators of inflammatory liver injury ............................... 37 The effects of VEGF on mediators of inflammatory liver injury ................................................................... 41 Pathways highly affected by the 142 functional genes selectively changed by TVX/LPS-treatment compared to all other treatment groups .............................................. 166 Genes selectively altered in expression by TVX/LPS that are regulated by interferons ..................................... 169 Supplemental table: Functional genes selectively changed by TVX/LPS-treatment compared to all other treatment groups .................................................. 192 Scorin of histopathology of livers from wild-type and PAl-1' ‘ mice cotreated with TVX/LPS ............................... 222 xiii LIST OF FIGURES Images in this dissertation are presented in color Fig. 1.1 Hypothetical relationship between inflammation and drug idiosyncrasy ......................................................... 1 7 Fig. 1.2 Toll-like receptor 4 signaling pathways ............................. 23 Fig. 1.3 A summary of the inflammatory process ........................... 26 Fig. 2.1 Dose response and development of liver injury from TVX/LPS cotreatment in mice ......................................... 60 Fig. 2.2 Comparison of TVX/LPS with LVX/LPS coadministration ...... 62 Fig. 2.3 Liver histopathology in mice cotreated with LPS and either TVX or LVX ........................................................ 65 Fig. 2.4 Timecourse of TNFa concentration in the plasma of treated mice ................................................................ 67 Fig. 2.5 The effect of pentoxifylline (PTX) on LPS-induced TNFa expression and TVX/LPS-induced liver injury ..................... 70 Fig. 2.6 Effect of PTX on TVX/LPS-induced liver pathology ............. 72 Fig. 2.7 The effect of etanercept on TVX/LPS-induced TNFa expression and liver injury ............................................. 75 Fig. 2.8 Effect of etanercept on TVX/LPS-induced liver pathology ..... 77 Fig. 2.9 Effect of etanercept treatment given at the peak of plasma TNFa on TVX/LPS-induced liver injury .................. 79 Fig. 3.1 Development of liver injury after TVX/LPS or TVX/PGN-LTA coexposure ............................................ 95 Fig. 3.2 Histopathology of livers from mice treated with TVX/LPS or TVX/PGN-LTA ......................................................... 97 Fig. 3.3 Effect of TVX pretreatment on LPS- and PGN-LTA- induced increases in cytokines ....................................... 99 Fig. 3.4 Effect of TVX pretreatment on LPS- and PGN-LTA- induced increases in chemokines .................................... 102 xiv Fig. 3.6 Fig. 3.? its. ”is. Fig.5. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. 3.5 3.6 3.7 3.8 4.1 4.2 4.3 4.4 4.5 4.6 4.7 5.1 5.2 5.3 5.4 5.5 Effect of TVX pretreatment on LPS- and PGN-LTA- induced hepatic neutrophil accumulation ........................... 105 Effect of CD18 neutralization on TVX/LPS- and TVX/PGN-LTA-ind uced liver injury .................................. 107 Effect of CD18 neutralization on TVX/LPS— and TVX/PGN-LTA-induced hepatic neutrophil accumulation ...... 109 Effect of CD18 neutralization on TVX/LPS- and TVX/PGN-LTA-induced cytokine increases ....................... 111 The role of TNF receptors in TVX/LPS-induced liver injury ................................................................... 128 Effect of TNFa inhibition on TVX/LPS-induced increases in plasma cytokines ........................................ 130 Effect of TNFa inhibition on TVX/LPS-induced increases in plasma chemokines ..................................... 133 Effect of TNFor inhibition on TVX/LPS-induced hepatic PMN accumulation ....................................................... 135 Effect of TNFa inhibition on hemostatic system dysregulation mediated by TVX/LPS coexposure ............... 137 TVX/TNFa coexposure-induced liver injury ........................ 140 Histopathology of TVX/TNFa-induced liver injury ................ 142 Development of TVX/LPS-induced liver injury .................... 158 Hierarchical clustering of hepatic gene expression profiles 160 Venn diagram depiction of probeset regulation of TVXNeh-, Veh/LPS- and TVX/LPS-treated mice relative to VehNeh controls ...................................................... 163 Timecourse of lL-18 and IF Ny plasma concentrations .......... 173 TVX/LPS-induced liver injury is attenuated in lL-18"' mice 175 lFNy"‘ mice are resistant to TVX/LPS-induced liver injury ..... 178 Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. Fig. 5.7 5.8 5.9 6.1 6.2 6.3 6.4 6.5 6.7 7.1 7.2 7.3 7.4 7.5 7.6 TVX/LPS-induced hepatic PMN accumulation is unchanged in IFNy"‘ mice ............................................... 180 Plasma concentrations of proinflammatory cytokines are reduced in lFNy"' mice ............................................. 182 Hypothesized role of lFNy in TVX/LPS-induced liver injury 188 Effect of fluoroquinolones and LPS on coagulation system activation ......................................................... 209 Effect of fluoroquinolones and LPS on plasma concentration of active PAl-1 .......................................... 211 Effect of fluoroquinolones and LPS on hepatic sinusoidal fibrin deposition ............................................................ 214 Effect of heparin treatment on TVX/LPS—induced hepatic fibrin deposition and liver injury ....................................... 216 Histopathology of heparin-treated and PAI-1"' mice treated with TVX/LPS ................................................... 218 Effect of PAH deficiency on TVX/LPS-induced hepatic fibrin deposition and liver injury ....................................... 221 Effect of heparin treatment or PAl-1 deficiency on TVX/LPS-induced increases in plasma cytokines ................ 225 Timecourse of VEGF plasma concentration ....................... 240 Effect of VEGF neutralization on TVX/LPS-induced plasma VEGF induction and liver injury ............................ 242 Protection from TVX/LPS-induced liver pathology by VEGF neutralization ..................................................... 244 Effect of VEGF neutralization on TVX/LPS-induced increase of cytokines .................................................... 247 VEGF neutralization did not affect TVX/LPS-induced increases in chemokines ............................................... 249 Effect of VEGF neutralization on TVX/LPS-induced hepatic neutrophil accumulation ...................................... 251 xvi Fig. 7.7 Fig. 8.1 Fig. 8.2 The role of VEGF in TVX/LPS-induced hemostatic system activation ......................................................... 254 Proposed pathways to TVX/LPS-induced liver injury ............ 268 Possible proinflammatory cycles induced by TVX/LPS coexposure ....... * ............................................ 272 xvii ALT ANOVA ATP cAMP CYP DAG DlLI DNA egr—1 Erk FAK FDA Flk-1 Flt-1 HIF-1or HPC lADR IFNy IFNGR1 IFNGR2 IKK-i IL-1 IL-4 IL—6 lL-1O lL-12 lL-15 IL-18 lP-1 0 IP3 IRAK IRF3 Jak1 JNK KC LBP LPS LTA LVX MAPK MOP-1 MHC MlP-1or MlP-2 LIST OF ABBREVIATIONS alanine aminotransferase analysis of variance adenine triphosphate cyclic adenosine monophosphate cytochrome P450 sn-1,2-diacylglycerol drug-induced liver injury deoxyribonucleic acid early growth response-1 extracellular regulated kinase focal adhesion kinase US. Food and Drug Administration VEGF receptor 2 VEGF receptor 1 hypoxia-inducible factor a hepatocyte idiosyncratic adverse drug reaction interferon y interferon 7 receptor 1 interferon y receptor 2 le kinase-i interleukin 1 interleukin 4 interleukin 6 interleukin 10 interleukin 12 interleukin 15 interleukin 18 lFNy-inducible protein 10 inositol (1 ,4,5)-triphosphate lL-1-receptor—associated kinase interferon regulatory factor 3 janus activated kinase 1 c-jun-N-terminal kinase keratinocyte chemoattractant LPS-binding protein lipopolysaccharide lipoteichoic acid levofloxacin mitogen-activated protein kinase monocyte chemoattractant protein-1 majOr histocompatibility complex macrophage inflammatory protein 1a macrophage inflammatory protein 2 xviii MPO MyDBB NFKB NK cells NKT cells p55 p75 PAl-1 PAR-1 PBMC PGN Pl PI3K PIP2 PKC PLCy PMN PTX RIP ROS Sck SODZ STAT1 TAB-2 TACE TAK1 TAT TBK1 TCR TIRAP TLR TNFa t-PA TRAF6 TRAM TRIF TVX u-PA VEGF VEGFR VRAP myeloperoxidase myeloid differentiation factor 88 nuclear factor-KB natural killer cells natural killer T cells TNF receptor 1 TNF receptor 2 plasminogen activator inhibitor 1 protease activated receptor-1 peripheral blood mononuclear cell peptidoglycan pharmacologic interaction phosphoinositide 3-kinase phosphatidylinositol (4,5)-biphosphate protein kinase C phospholipase C y neutrophil pentoxifylline receptor-interacting protein reactive oxygen species Shc-Iike protein superoxide dismutase 2 signal transducer and activator of transcription 1 TAK1 binding protein TNFoI-converting enzyme transforming growth factor-b-associated kinase thrombinzantithrombin TANK binding kinase 1 T cell receptor Toll/lL-1 R domain-containing adapters toll-like receptor tumor necrosis factor a tissue plasminogen activator tumor necrosis factor-associated factor 6 TRlF-related adapter molecule TlR-containing adapter molecule trovafloxacin urokinase plasminogen activator vascular endothelial growth factor VEGF receptor VEGF receptor-associated protein xix CHAPTER 1 General Introduction and Specific Aims 1.1 Idiosyncratic adverse drug reactions 1.1.1 Overview of idiosyncratic adverse drug reactions Adverse drug reactions are a serious problem for not only the public health, but also for pharmaceutical companies and drug-regulatory agencies. In a study in the United Kingdom, adverse drug reactions accounted for more than 6% of hospital admissions. Of these admissions due to adverse drug reactions, the mortality rate was 2% (1). In addition to the risk to public health, adverse drug reactions are a major issue for drug development. A significant amount of time and money is expended in the effort to predict the risk of adverse reactions from drug candidates. Despite comprehensive preclinical drug testing and clinical trials, over 10% of drugs approved during 1975-2000 were either withdrawn from the market or have been highly restricted in use (2). In 1998, the pharmaceutical industry spent over 20 billion dollars on drug discovery and development, with screening assays and toxicity testing accounting for about 20% of the total amount spent (3). Despite such extensive efforts, in 1999 over 258,000 post— marketing adverse events were reported in the United States, suggesting that this is a persistent major issue (4). Adverse drug reactions can occur in any number of tissues, but the liver is often the target organ. Of the 28 drugs removed from the US market between 1976 and 2005, 6 were withdrawn due to hepatotoxicity (5). Drug-induced liver injury (DILI) accounts for more than 50% of acute liver failure cases (6). It is associated with significant mortality; therefore, a number of drugs which have been associated with DILI have been removed from the market. For example, bromfenac (7), troglitazone (8, 9) and tienilic acid (10) have been completely removed from the market due to hepatotoxicity. In addition, hepatotoxicity of other drugs such as trovafloxacin (I'VX) (11, 12), nefazodone (13), and nevirapine (14, 15) has led to “black box” warnings limiting their use. DILI is the leading cause for the withdrawal of drugs from the market by either the US. Food and Drug Administration or pharmaceutical companies (16). An important subset of adverse drug reactions which cause DILI are idiosyncratic adverse drug reactions (lADRs), which account for 13-17% of all cases of acute liver failure (6, 17). lADRs typically occur in a small fraction of people (generally < 1%) within the range of doses used clinically. The exact mechanisms underlying lADRs are unknown but typically do not involve the pharmacological properties of the drug. In addition, lADRs lack an obvious dose- dependence, meaning that a dose which causes toxicity in some patients does not in others. Another characteristic of lADRs is that the onset of toxicity relative to the duration of drug therapy is variable. Finally, there is a wide range in the severity of the reactions depending on the drug and individuals. Despite extensive research, animal models do not exist which reproduce the hepatotoxicity caused by lADRs. The development of animal models is necessary to predict those drugs which cause lADRs and to decrease human suffering. A predictive animal model would be beneficial for several reasons. Prediction of drug candidates that could cause lADRs would prevent their development into marketed pharmaceuticals and thereby reduce risk to public health. In addition, it would prevent pharmaceutical companies from sending such candidates to clinical trials or to market and would thereby save money spent on clinical trials, marketing and potential lawsuits from patients affected by lADRs. Drugs which lead to lADRs are usually not identified in preclinical testing due to their typically rare occurrence and the use of relatively small numbers of animals in toxicity testing. The inability of animal tests to predict lADRs may be due, in part, to the reaction being idiosyncratic in animals as well as humans, and thus an extremely large number of animals would be needed to detect toxicity. It has been estimated that to predict an lADR confidently, toxicity testing would require 30,000 animals to be treated (18). In addition, the current animal testing paradigms might not include sufficient biological diversity to elucidate lADR toxicities. Since such large studies are not possible for drug candidates, it is critical that the modes of action of lADRs are better understood to develop predictive models. 1.1.2 Hypothesized mechanisms of idiosyncratic adverse drug reactions Despite extensive research, the mechanisms underlying lADRs remain poorly understood and incompletely characterized. There exist several obstacles to understanding lADRs. A substantial challenge is that an animal model for the early detection of hepatic lADRs is currently unavailable. In addition, the tissue from afflicted individuals is often difficult to obtain for research purposes, although the DILI network is trying to address this obstacle. Even when tissue from affected individuals is available, the tissue would have been harvested long after injury developed and is likely, therefore, to be of limited value for mechanistic studies. However, despite such limitations and difficulties, progress has been made in understanding ‘ lADRs. Such progress has led to the development of several diverse theories about lADR pathogenesis. To this point, none of the hypotheses to explain lADR pathogenesis have been proved or disproved. The prevalent hypotheses to explain lADR toxicity and supporting experimental evidence are described in more detail below. Reactive Intermediate Hypothesis One theory for the mechanism of lADRs is that a drug is metabolized into a reactive metabolite, which might bind with important cellular proteins, damage membrane integrity, alter calcium homeostasis or other intracellular signaling in ways which could lead to toxicity and that susceptible individuals have polymorphisms in the bioactivating enzyme(s) (19). Indeed, there are several cases in which a drug linked with lADRs has the ability to form a metabolite which is reactive (20). The reactive intermediate hypothesis can be closely associated with all of the hypotheses to be described, especially if a reactive metabolite and not the parent drug is the agent involved in the toxicity. Troglitazone is an antidiabetic drug which was linked with serious idiosyncratic hepatotoxicity (9). Research conducted after troglitazone was removed from the market showed that it is metabolized in the rat to five intermediates with the ability to form glutathione conjugates that appear in bile (21). In addition, metabolic activation by cytochrome P450 3A4 (CYP3A4) forms reactive metabolites which bind to proteins and nucleophiles (22). Whether these form protein adducts that play a role in toxicity is unknown. In addition, if the protein adducts are formed and involved in toxicity, the degree of protein adducts that constitute a threshold for troglitazone lADRs is unknown. Furthermore, several drugs which form reactive metabolites are not associated with an increased risk of lADRs (23). Moreover, one would expect an “intrinsic” (dose- related) toxicity picture in the absence of some metabolism-related sensitivity factor that renders a small fraction of patients susceptible to lADRs. Thus, although the reactive intermediate hypothesis is a reasonable one, a causal link between reactive metabolite generation and hepatotoxicity has not been established conclusively for drugs that cause lADRs. Genetic Polymorphism Hypothesis A related theory is that genetic polymorphisms among individuals can cause differences in the toxic responses of individuals to drugs. Many polymorphisms can lead to drug metabolism differences among individuals, leading to differences in pharmacokinetics and reactive intermediate formation (24). Human polymorphisms in genes encoding cytochrome P450 drug metabolizing enzymes have been identified and could lead to differences in drug metabolism and clearance that could render some individuals more susceptible to toxicity. In addition, it is possible that a polymorphism in drug metabolizing enzymes might lead to the formation of a reactive intermediate not seen in the majority genotype. Alternatively, a genetic polymorphism in a protective gene, such as an anti-inflammatory cytokine, might render individuals more susceptible to normally nontoxic doses of drugs, resulting in an lADR. For this hypothesis to explain lADRs, the genetic polymorphism of people on drug therapy would have to be as rare as the lADR itself or the lADR would have to be a result of a rare combination of several more common polymorphisms. Even if this explains the rarity of lADRs, the genetic polymorphism hypothesis does not explain other characteristics of lADRs such as the variability in the onset of toxicity. An example often referenced by supporters of the importance of polymorphisms is toxicity caused by isoniazid, a first-line drug used in the prevention and treatment of tuberculosis. Isoniazid has been linked to several cases of liver injury (25). The susceptibility of individuals to isoniazid-induced liver injury has been linked to a polymorphism resulting in a rapid acetylator phenotype (25, 26). It was hypothesized that the rapid acetylators produce more of a reactive metabolite which causes hepatocellular necrosis. However, several epidemiological studies failed to find an association between the rapid acetylation polymorphism and liver injury (27). Another example of this hypothesis is evident from a study in which individuals were treated with the idiosyncratic drug, diclofenac. It was found that individuals who developed a toxic response had a greater rate of polymorphisms in the interleukin 10 (IL-10) and interleukin 4 (IL-4) genes than the group of individuals who did not develop a toxic response to diclofenac (28). An association between lADRs and genetic polymorphisms does exist with some drugs; however, their roles remain uncertain, and it remains likely that other factors play a role in precipitating lADRs. Hapten Hypothesis A widely accepted theory to explain lADRs is that they result from an adaptive immune response. Some clinical characteristics of lADRs such as the delayed onset of toxicity, the lack of a simple dose-response relationship and eosinophilia have led some to postulate that lADRs are mediated by adaptive immunity (29). This has led to the formation of two related hypotheses. The hapten hypothesis states that a chemically reactive drug or a reactive metabolite binds to an endogenous protein. This protein adduct is then seen as a foreign antigen capable of initiating immunological recognition (30). According to this hypothesis, the drug-modified protein must be processed by antigen-presenting cells and presented to T cells. This results in sensitization of the T cells to the foreign antigen. The immune system develops memory to the foreign antigen, and upon subsequent exposure to the drug, robust immune system activation occurs, resulting in the formation of autoantibodies and/or the activation of cytotoxic T cells targeting self proteins (31). It is important to understand that both sensitizing and challenging exposures are required in this hypothetical mechanism. In support of the hapten hypothesis, the presence of autoantibodies has been detected in patients with hepatic lADRs after exposure to several drugs, including diclofenac, troglitazone, halothane and tienilic acid (28, 32). The study which found autoantibodies in the sera of patients who experienced diclofenac hepatotoxicity also reported the presence of autoantibodies in some patients treated with diclofenac who did not develop hepatotoxicity (28). Such a finding was also found in halothane-treated patients, in whom autoantibodies were found whether they developed toxicity or not (33). Thus, from these reports, a clear cause and effect relationship between autoantibodies and idiosyncratic hepatotoxicity is lacking. The clinical evidence supporting the role of the adaptive immune system may in some cases be explained by immune system activation occurring secondary to tissue damage. Efforts have been undertaken to show the involvement of the specific immune system in hepatotoxic lADRs; however, in all of the current animal models of drug immunogenicity, an adaptive immune response was detected in the absence of liver damage (34). Accordingly, experimental support for this hypothesis is incomplete, and an animal model of drug hepatotoxicity with an adaptive immune mechanism has not emerged so far. The danger hypothesis A theory closely related to the hapten hypothesis described above is the danger hypothesis, which proposes that a damaging immune system activation occurs only if the drug binds to a protein which causes some type a of a stress response, such as inflammation or cell death, resulting in a ‘danger’ signal (35). Thus, according to the danger hypothesis, the formation of a drug-protein adduct is insufficient to cause injury, a secondary signal during sensitization such as mild cell death or cytokine release then results in adaptive immune system activation and pathogenesis (36). It has been postulated that reactive drug metabolites themselves could cause this danger signal, and this is what determines which reactive metabolites lead to lADRs (37, 38). However, the ‘danger’ signal could be from a number of independent factors including an infection causing an innate immune response, resulting in an inflammatory stress. The pharmacological interaction (PI) hypothesis The PI hypothesis is closely related to the hapten and danger hypotheses, in that it suggests an active role for the adaptive immune system in the development of lADRs. The PI hypothesis proposes that drugs bind reversibly to the major histocompatibility complex (MHC) and T cell receptor (T CR) complex. It is hypothesized that the drug then acts like a superantigen to elicit an adaptive immune system response, precipitating an lADR (39). Much of the early work leading to the development of the Pl hypothesis was done with sulfamethoxazole; which caused proliferation of T cells isolated from sulfamethoxazole lADR patients (40). However, there is no evidence that an lADR drug binding to the MHC-TCR complex is capable of eliciting an immune response. The role as a possible superantigen to the MHC:TCR complex has not not been shown with any other drugs linked with hepatotoxic lADRs. In addition, evidence is also lacking in support of a causal link between an adaptive immune response and the precipitation of a hepatotoxic lADR. 1O Mitochondrial dysfunction hypothesis Another hypothesis for lADRs is that mitochondrial dysfunction and disturbances in mitochondrial integrity by oxidative stress are an underlying cause. Mitochondria play a critical role in providing the cell with energy, controlling the process of apoptosis and regulating intracellular oxidative stress. Mitochondrial dysfunction can encompass several changes such as decreased adenosine triphosphate (ATP) production, mitochondrial reactive oxygen species (ROS) production or depolarization of the mitochondrial membrane potential. One way in which mitochondrial dysfunction can occur is through DNA alteration. Mitochondrial DNA alterations which could result in dysfunction are rare but are seen in humans. It was found in a epidemiological study that >12 in 100,00 people either had mitochondrial DNA disease or were at risk to develop it; these results reflect the minimum prevalence of mtDNA disease and pathogenic mtDNA mutations (41). It is hypothesized that either a mitochondrial disease or polymorphism could alter mitochondrial function and render cells sensitive to a drug, resulting in idiosyncratic toxicity (42). It is also postulated that genetic or acquired mitochondrial abnormalities can lead to silent and gradually accumulating mitochondrial injury which reaches a threshold and abruptly triggers liver injury (43). There is extensive evidence linking lADR drugs with mitochondrial alterations. Troglitazone, tolcapone, diclofenac, valproic acid, and isoniazid are some of the drugs which cause lADRs and which have mitochondrial liability in hepatocytes (43-48). In addition, diclofenac and troglitazone are cytotoxic to 11 HepGZ cells through a mitochondrial mechanism (49, 50). In one study, superoxide dismutase 2 ($002) heterozygote mice, a model of silent mitochondrial abnormality, were chronically treated with troglitazone. This treatment had no effect on wild-type mice but resulted in hepatocellular necrosis in 8002*" mice (51). However, the hypothesis fails to explain the apparent lack of dose dependence that characterizes lADRs. In addition, there are several drugs that cause mitochondrial alterations in vitro but have not resulted in adverse drug reactions in people. It is of importance to note that mitochondrial dysfunctions can be induced by a number of independent factors such as xenobiotics which might be taken concurrently, hypoxia or inflammation. Therefore, it is possible that alterations in mitochondrial function play a role in other hypothesized mechanisms of lADRs. Failure to adapt hypothesis Another hypothesis of lADRs is that a small fraction of people develop minor liver toxicity in response to a drug. Most of these individuals “adapt” and experience a resolution of liver injury even in the continued presence of the drug. However, it is proposed that a small fraction of these people fail to “adapt”, and the injury progresses to overt toxicity (52). Reports of isoniazid hepatotoxicity seem to support this theory, inasmuch as 15% of patients taking isoniazid experience minor alanine aminotransferase (ALT) elevations, but less than 1% develop symptomatic hepatitis with continued treatment (53). 12 The mechanisms underlying the “adaptation” phenomenon are unknown. Adaptation may not be recognized in clinical trials because drug treatment is stopped when the serum ALT activity rises to greater than 3 times the upper limit of normal, making it impossible to diStinguish between patients who would and would not adapt. In addition, there are currently few animal models in which adaptation can be studied. However, future studies made possible by the DILI Network will attempt to address these issues and determine possible reasons for increased susceptibility of certain individuals to lADRs. It is also of importance to note that the ‘failure to adapt’ hypothesis does not discount other hypotheses of lADRs, as toxicity may be due to any number of mechanisms to which certain individuals cannot adapt and therefore experience an lADR. Multiple determinant hypothesis The multiple determinant hypothesis proposes that idiosyncratic reactions are the result of multiple, discrete but necessary factors or processes all occurring simultaneously (54). Each factor has an independent probability of occurring, but all of them are required to precipitate an lADR, thus accounting for the rare occurrence rate. According to the hypothesis, an idiosyncratic reaction would only occur in an individual if all the critical steps occur within an appropriate time. An equation for the probability of an idiosyncratic reaction is proposed below: PIADR = Pchem X Pexp X IDenv X Pgenei Where, 13 Pma is the probability of an lADR, Pchem is the probability contributed by chemical properties, Pexp is the probability determined by the drug exposure to the critical organ(s), Pam, represents probabilities determined by environmental factors (drug coexposure, inflammation, etc.) and Pgene is the probability related to genetic factors (54). The multiple determinant hypothesis is a rather general and encompassing hypothesis which takes into account the other hypotheses mentioned above. However, it is important to understand in more detail the mechanistic aspects of lADRs to develop predictive animal models. Inasmuch as environmental and genetic factors might play a role in the probability of a specific drug causing an lADR, it is important to determine which factors are important to toxicity and why. The hypothesis implies that an underlying factor has the potential to lower the toxicity threshold of a drug, rendering a normally therapeutic dose toxic. Several factors have the potential to affect the susceptibility of an individual to drug toxicity including age, gender, coexposure to other pharmacological agents, drug metabolism differences, and state of health. Inflammatory stress hypothesis In the multiple determinant hypothesis, one environmental factor that might render an individual sensitive to a normally nontoxic drug dose is inflammatory stress. This idea has led to the inflammatory stress hypothesis, 14 which states that an episode of inflammation has the potential to interact with concurrent drug therapy to precipitate an lADR. Inflammatory episodes are commonplace in people and occur erratically throughout life. Many are modest enough that they go unnoticed. A hypothetical relationship between inflammation and lADRs is illustrated in Fig. 1.1. For therapeutically useful drugs, the pharmacologic effect is seen at much smaller doses than signs of toxicity. Most drugs are developed so that the range between a therapeutic dose and the smallest toxic dose (ie., the therapeutic window) is as large as possible. As dose is increased, toxicity is seen (such as kidney toxicity in Fig. 1.1) and death ensues at large doses. Liver toxicity in this example is not observed because the toxicity threshold lies at doses higher than those that are lethal. The hypothesis is that a modest inflammatory stress can decrease the threshold for hepatic toxicity, thereby shrinking the therapeutic window and resulting in a toxic response at a normally safe and pharmacologically effective dose of the drug. In this case, an lADR would occur at a dose which is nontoxic to individuals not experiencing a concurrent inflammatory episode. The erratic nature of inflammatory episodes can explain the unpredictable nature of lADRs. Lipopolysaccharide (LPS), a component of gram-negative bacterial cell walls, is one agent that can induce an inflammatory stress as described in more detail below. Experimental models have been developed in which nontoxic doses of lADR-causing drugs are rendered hepatotoxic upon coexposure to a nontoxic dose of LPS. For example, rats became susceptible to hepatotoxicity from several drugs known to cause lADRs when they were concurrently exposed to a 15 Fig. 1.1. Hypothetical relationship between inflammation and drug idiosyncrasy. Drug A is a relatively safe and efficacious drug. The asterisk indicates the usual therapeutic dose. The safety margin between pharmacological effect and kidney toxicity is quite large. A modest inflammatory response shifts the threshold for liver toxicity and precipitates and idiosyncratic response (55). 16 < 9.5 no econ \ .xmhdozu . .6... 58m 355. esuodsau 17 nontoxic dose of LPS. Drugs known to cause lADRs in humans such as trovafloxacin, ranitidine, sulindac, chlorpromazine and diclofenac were all rendered hepatotoxic to rats when coupled with a nontoxic dose of LPS (56-60) (Table 1.1). Drugs in the same pharmacologic class which were not associated with lADRs in humans were used when available. These drugs not associated with human lADRs did not interact with inflammatory stress to cause hepatotoxicity in animal models (58, 59). Of the drugs tested, only the ones linked with lADRs in humans interacted with a concurrent inflammatory stress to cause hepatotoxicity in rats. This concordance suggested a potential role for inflammation in the mechanism of human lADRs. The results in animal models suggest that an inflammatory episode caused by LPS or other factors could render an individual susceptible to hepatotoxicity at normally nontoxic drug doses, thus causing an idiosyncratic reaction. A challenge still lies in understanding mechanisms of the hepatotoxicity observed with coexposure to LPS and an lADR-causing drug. The remainder of the Introduction and subsequent chapters of the thesis will explore inflammatory stress in greater detail and present work to develop and explore an inflammation/drug interaction model of TVX toxicity in mice. 18 Table 1.1. Concordance of LPS/drug coexposure model in rats for lADR- causing drugs in humans Linked to . . LPS/drug coexposure Drug hepatotoxrcIty in _ hepatotoxm to rats? humans? Trovafloxacin Yes Yes Levofloxacin No No Chlorpromazine Yes Yes Ranitidine Yes Yes Famotidine No No Sulindac Yes Yes Diclofenac Yes Yes 19 1.2 Inflammation 1 .2.1 Overview of inflammatory stress Inflammation is an innate immune system process critical for the host’s defense against infection and foreign substances. The inflammatory response is a complex process encompassing the recruitment of cells, release of cytokines and other biologically active mediators, vasodilation, hemostatic system activation and complement activation. The magnitude of an inflammatory response depends on the cause and varies from one individual to the next. Modest inflammatory episodes occur sporadically and are commonplace in people. Inflammation occurs in response to a number of stimuli including tissue injury, microbial pathogens and other foreign substances. As mentioned above, recognition of microorganisms by various cell types within the body induces an inflammatory response. Components of gram- negative bacteria have been measured In the plasma of individuals and are increased by conditions such as gastrointestinal disturbances, alcohol consumption, surgery, alterations in diet, etc. (55, 61). In turn, a great deal of inflammation research has focused on host responses to gram-negative bacterial cell wall constituents. Endotoxin is a component of gram-negative bacterial cell walls and is released when bacteria undergo cell division or are damaged by antibiotics (62). A major, biologically active component of endotoxin is LPS. Chapter 3 presents some studies exploring the interaction between TVX and gram-positive bacterial cell wall components peptidoglycan and lipoteichoic acid, 20 which can also induce inflammation. However, the majority of the work will explore in detail the interaction between TVX and LPS. The mechanism by which LPS induces an inflammatory response will described below. Toll-like receptors (TLRs) are conserved pattern recognition receptors that recognize bacterial components (63). The effects of LPS are elicited primarily through the activation of TLR4. LPS-binding protein and CD14 are required for presentation of LPS to TLR4; and the interaction of the co-receptor MD-2 with dimerized TLR4 is required to elicit activation of TLR4 by LPS (64, 65). After LPS activates TLR4, the resulting responses can be divided into those dependent on myeloid differentiation factor 88 (MyDBB) and those independent of MyD88. The signaling pathways activated by TLR4 activation by LPS are described in more detail below and summarized in Fig. 1.2. The activated TLR4 dimer recruits Toll/lL-1R domain-containing adapters (T IRAP), TlR-containing adapter molecule (TRlF) and TRIP-related adapter molecule (T RAM) (66). TIRAP recruitment and activation results in My088 recruitment. My088 is an adapter protein which activates inflammatory signaling pathways. The activation of My088 leads to the recruitment and phosphorylation of members of the lL-1-receptor-associated kinase (IRAK) family (67). Phosporylated IRAK then dissociates from My088 and interacts with tumor necrosis factor receptor-associated factor 6 (TRAF6) (68). Activated TRAF6 associates with TAK-1 binding protein-2 (TAB-2), causing activation of transforming growth factor-B-associated kinase 1 (TAK1). TAK1 is a mitogen- activated protein kinase (MAPK). At this point, TAK1 activates the p38 MAPK 21 Fig. 1.2. Toll-like receptor 4 signaling pathways. Schematic summary of TLR4 signaling following activation by LPS. LPS-binding protein (LBP) and soluble CD14 assist in the presentation of LPS to the TLR4 dimer. The TLR4 dimer is associated with MD-2 in the cell membrane. Upon LPS binding, Toll/lL-1R domain-containing adapter (T IRAP) and TRIF-related adapter molecule (T RAM) are recruited to the intracellular domain of TLR4. TIRAP recruitment and activation allows for myeloid differentiation factor 88 (MyD88) recruitment and activation. IL-1-receptor-associated kinase (IRAK) binds to the activated MyD88 and is in turn activated. IRAK activation allows for the binding and subsequent activation of tumor necrosis factor receptor-associated factor 6 (TRAF6). TRAF6 binds to a complex formed by TAK-1 binding protein-2 (T AB-2) and transforming growth factor-B-associated kinase 1 (T AK1). The binding of TRAF6 to the TAB- 2fl'AK1 complex results in TAK1 phosphorylation. TAK1 is a mitogen-activated protein kinase (MAPK) which in turn activates p38 and JNK MAPK pathways and NF-KB. The recruitment of TRAM also results in MyDBB-independent signaling. Toll/IL-1 R domain-containing-containing adapter molecule (TRIF) binds to TRAM and becomes activated. TRIF then serves as an adapter molecule for the activation of TRAF6, again leading to MAPK and NF-KB activation. TRIF binds to the TANK binding kinase1 (T BK1)/IKB kinase-l (lKK-i) complex which is believed to phosphorylate and activate TBK1. TBK1 phosphorylates and activates the transcription factor interferon regulatory factor 3 (IRF3), the activation of which results in interferon a and [3 (IFNa/B) expression. 22 W088 IRAK: TRAF6 TRAF6 2 IRF3 TAK1 NF-KB p38 MAPK JNK MAPK NF {B MAPKs IFNalp 23 pathways, c-jun-N-terminal kinase (JNK) MAPK pathways, and the inflammatory transcription factor, nuclear factor-KB (NF-KB) (69). NF-KB, p38 and JNK activation leads to gene/protein expression of several mediators of inflammation and cell death (70). In addition, LPS activation of TLR4 results in a MyD88-independent response via the recruitment of TRAM and TRIF (also known as TICAM1) (66). TRIF then can directly activate TRAF6 with the help of receptor-interacting protein (RIP), resulting in MAPK and NF-KB activation independent of My088. In addition, TRIF can bind to the TANK binding kinase 1 (T BK1)/II 3 § § § E ° ('luliedpogm 75 . -r . 4:: i “i ‘0'. L 4 a) at: m r . 5 . I f. > -. , , 1 ' . . a , f . e t i t .2— ° ti: 3 2 i. (10”ij g P." ('I lllle) 9"" 99 LPS PGN-LTA Veh LPS PGN-LTA Veh Fig. 3.3 (cont’d). - Veh F.“ .- IE 0- IS ash—P ...-I at .‘l 2 5 2 0. . _ ae+ 2 2 _I _l 8 .g s sass-38° §§§§§§° ('l ullfiu) I-dow (1w16d) 0l-'Il #0 l_. LPS PGN-LTA * LPS PGN-LTA Veh Veh s: g s s ° recess" (1 wlfidl :IOBA l1 wrfid) 1- UOJOIJGIUI 100 Fig. 3.4. Effect of TVX pretreatment on LPS- and PGN-LTA- induced increases in chemokines. Mice were treated with TVX or Veh and then LPS, PGN-LTA or Veh as described in Section 3.3.3. Mice were killed at 4.5 h, and plasma concentrations of KC, MlP-2 and MlP-1or were measured. n = 4-6 animals/group. * significantly different from VehNeh-treated group. " significantly different from TVXNeh-treated mice. ‘D significantly different from Veh-treated mice within the same treatment group. 101 m M _m Mil—ll. .... ... a .. . a . SE‘OEOV. "l? #¢ ”warm 3835 urn—=2 o Veh mmmmo 32:03 8.3:: LPS PGN-LTA Veh 102 3.4.3 Effect of TVX on microbial stimuli-induced hepatic neutrophil accumulation Hepatic neutrophil accumulation was evaluated at 4.5 h after LPS or PGN- LTA. Both LPS and PGN-LTA alone caused a significant increase in neutrophils present in the liver (Fig. 3.5). TVX alone did not cause hepatic neutrophil accumulation. In addition, TVX pre-treatment did not affect the number of neutrophils present in the liver after either inflammatory stimulus (Fig. 3.5). 3.4.4 Effect of CD18 neutralization on TVXILPS- and TVXIPGN-LTA-induced liver injury and inflammation CD18 antiserum was administered as described in Section 3.3.3. CD18 neutralization protected mice from TVXILPS- and TVX/PGN-LTA-induced liver injury as measured by plasma ALT activity (Fig. 3.6). 0018 antiserum administration significantly attenuated TVX/PGN-LTA- induced hepatic neutrophil accumulation (Fig. 3.7). In addition, CD18 neutralization reduced the TVX/PGN-LTA-induced increases in TNFa and MOP-1 (Fig. 3.8). In contrast, CD18 neutralization did not affect TVXILPS-induced hepatic neutrophil accumulation (Fig. 3.7). Similarly, CD18 neutralization did not affect any cytokines induced by TVX/LPS coexposure (Fig. 3.8). 103 Fig. 3.5. Effect of TVX pretreatment on LPS- and PGN-LTA- induced hepatic neutrophil accumulation. Mice were treated with TVX or Veh and then LPS, PGN-LTA or Veh as described in Section 3.3.3. Mice were killed at 4.5 h. Paraffin-embedded liver lobes were cut and stained for PMNs. n = 4-6 animals/group. * significantly different from VehNeh-treated group. ‘ significantly different from TVXNeh-treated mice. 104 “E: .8. czsd LPS PGN-LTA Veh 105 Fig. 3.6. Effect of CD18 neutralization on TVXILPS- and TVXIPGN-LTA- induced liver injury. Mice were treated with TVX/LPS or TVX/PGN-LTA and CD18 antiserum or control serum as described in Section 3.3.3. Mice were killed at 15 h and plasma ALT activity was measured. n = 6-10 animals/group. * significantly different from control serum treated mice within the same treatment group. 106 8000 . - Control Em CD18 antiserum 5 \\ 88 1000 - ALT activity (UIL) 88 8 TVX/LPS TVXIPGN-LTA 107 Fig. 3.7. Effect of CD18 neutralization on TVXILPS- and TVXIPGN-LTA- induced hepatic neutrophil accumulation. Mice were treated with TVX/LPS or TVX/PGN-LTA in addition to CD18 antiserum or control serum as described in Section 3.3.3. Mice were killed at 4.5 h. Paraffin-embedded liver lobes were stained for PMNs. n = 6-10 animals/group. 1‘ significantly different from control serum treated mice within the same treatment group. 108 Hepatic PMNs per HPF .5 O — Control — CD18 antiserum . . " J . , 'Ds 5“ - L'." . 35'; l . -I~ ., r. I I J .. TVX/LPS TVXIPGN-LTA 109 Fig. 3.8. Effect of CD18 neutralization on TVXILPS- and TVXIPGN-LTA- induced cytokine increases. Mice were treated with TVX/LPS or TVX/PGN- LTA in addition to CD18 antiserum or control serum as described in Section 3.3.3. Mice were killed at 4.5 h and plasma cytokine concentrations were measured. Control and CD18 antiserum control mice had equivalent baseline concentrations of IL-6 (0.05 1 0.005 ng/mL), lL-10 (36 1 6 pg/mL), TNFor (411 1 120 pg/mL), IL- 18 (75 1 15 pg/mL), KC (0.04 1 0.006 ng/mL), MIP-1a (541 1 56 pg/mL), lFNy (9 1 2 pg/mL) and MOP-1 (0.14 1 0.03 ng/mL). n = 6-10 animals/group. 1‘ significantly different from control serum treated mice within the same treatment group. 110 TVXILPS TVXIPGN-LTA gasses: ('I wrfld) Ol-"II (‘I wrfid) 9 VII TVXILPS TVXIPGN-LTA U r U U U V V c G O G O 0 O O O G 1' N 1- e- 1- (1 IIJIDU) 9-'u ('l wlfidl 1°le 140 a 120 111 Fig. 3.8 (cont’d). {:1 CD18 antiserum -Conlrol TVX/LPS TVXIPGN-LTA TVX/LPS TVXPGN-LTA ééé" itéééé (1wl5di 1ol-cllllil (1wr6u) l-dow TVXILPS TVXIPGN -LTA TVXILPS TVXIPGN-LTA geese:- estates (1wrlu)o)l (‘lullfidHNdl 112 3.5 Discussion Inflammatory episodes are commonplace and occur sporadically. We hypothesized that an inflammatory stress can decrease the toxicity threshold to certain drugs, precipitating an idiosyncratic toxicity. Previous studies showed that TVX enhanced the LPS-induced plasma TNFa increase and that TVX/LPS coexposure resulted in hepatotoxicity (216). The studies presented here examined whether TVX enhanced the LPS-induced increases in other cytokines. In addition, they explored whether TVX enhanced the cytokine response to gram- positive microbial stimuli, PGN and LTA, and if TVX/PGN-LTA coexposure was hepatotoxic to mice. Inflammation is a complex process which can be initiated by the host’s recognition of microbial products by toll-like receptors. Bacterial components can be measured in the plasma and are increased by a number of stressors including alcohol consumption, surgery and gastrointestinal disturbances (61). Bacterial components of both gram-positive bacteria (PGN and LTA) and gram-negative bacteria (LPS) have been measured in the plasma and activate toll-like receptors to induce inflammation. PGN and LTA activate TLR2 and induce NFch activation through My088-dependent mechanisms (217, 220-222). As described extensively in Section 1.2.1, LPS activates TLR4 and induces NFch activation through both My088-dependent and —independent mechanisms. A nontoxic dose of TVX was rendered hepatotoxic upon coexposure to a nontoxic dose of either LPS or PGN-LTA (Fig. 3.1). The timecourse of hepatotoxicity for TVX/LPS- and TVX/PGN-LTA-induced liver injury was similar, 113 inasmuch as plasma ALT activity increased as early as 4.5 h and continued to progress until 15 h. That TVX interacted with either TLR2- or TLR4-activating ligands to cause liver injury proves that the TVX/inflammation-induced liver injury shown previously (216) is not specific to TLR4 activation. Indeed, it suggests that TVX interacts with an inflammatory stress, irrespective of its source, to precipitate liver injury. The result suggests that inflammatory stress induced by either gram-positive or gram-negative bacteria might play a role in TVX hepatotoxicity. Despite a similar timecourse of liver injury, the histopathology differed between TVX/LPS- and TVX/PGN-LTA-induced liver injury (Fig. 3.2). TVX/LPS- treated mice developed lesions of hepatocellular necrosis and apoptosis primarily localized to midzonal regions, whereas TVXIPGN-LTA lesions of hepatocellular necrosis and apoptosis were primarily centrilobular regions. Such a difference in localization might be due to a difference in TLR2 and TLR4 expression in regions of the mouse liver, of which little is known. Another possibility is that the difference in the location of the lesions suggests a difference in the mechanism of pathology. Therefore, to examine possible mechanisms of pathogenesis, inflammatory cytokines were measured at 4.5 h, the onset of liver injury, to determine if TVX enhanced cytokine release in response to bacterial stimuli. TVX enhanced the LPS- and PGN-LTA-induced increases of several cytokines: IL-1B, lL-6, lL-18, VEGF, MCP-1 and lL-10. In contrast, only the LPS- induced increase of TNFa and IFNy was enhanced by TVX, and it did not affect the PGN-LTA induction of these cytokines (Fig. 3.3). These cytokines were only 114 measured at 4.5 h, therefore it is possible that TVX enhanced PGN-LTA-induced increases in TNFa and IFNy at other times. Indeed, TVX pretreatment caused a trend towards an increase in TNFa in PGN-LTA-treated mice, but the difference was not statistically significant at this time. lFNy induction by LPS, but not PGN- LTA, was enhanced by TVX, a result possibly related to the differences in TLR2- and TLR4-activation. TLR4, but not TLR2 activation induces dendritic cells to produce lL-12 (223, 224), which directly stimulates natural killer and T cells to produce IFNy (225). It is thus possible that TVX acts to enhance a number of the steps in this pathway of IFNy production induced by LPS which would not be activated by PGN-LTA. Similar to a number of cytokines, TVX enhanced the LPS- and PGN-LTA- induced increase in KC, MIP-2 and MlP-1a (Fig. 3.4), all of which can be upregulated as a result of NFIcB activation (226). It is thus likely that the common upregulation of cytokines by both stimuli was mediated through NFxB activation. KC, MIP-2 and MlP-10t all have chemotactic activity for neutrophils; therefore, hepatic PMN accumulation was quantified to determine if the TVX enhancement of chemokines was associated with an increase in LPS- and/or PGN-LTA- induced PMN accumulation. Despite an increase in several chemokines, TVX pretreatment did not affect LPS- or PGN-LTA-induced neutrophil accumulation in the liver (Fig. 3.5). After LPS/galactosamine treatment, neutrophils roll and adhere in hepatic postsinusoidal venules independently of KC or MlP-2, but extravasation of neutrophils into the parenchyma was significantly reduced by MlP-2 or KC neutralization (227). It is thus possible that TVX enhances LPS- and 115 PGN-LTA-induced PMN extravasation into the parenchyma and in turn PMN activation. This would not be detected by PMN staining. To determine if PMN activation is involved in TVX/LPS- or TVXIPGN-LTA- induced liver injury, mice were pretreated with a neutralizing antibody to CD18, a 82-integrin critical for PMN activation (102, 228). CD18 neutralization attenuated hepatotoxicity induced by either TVX/LPS or TVX/PGN-LTA coexposure (Fig. 3.6); therefore, PMN activation appears to be a common pathway required for the progression of liver injury. Neutrophil activation is required for TVX/Inflammation- induced liver injury in both rats and mice (58), which suggests that this requirement is not species-specific. It therefore might be an important pathway in TVX-induced hepatotoxicity in people. The mechanism of hepatic neutrophil accumulation after TVX/LPS or TVX/PGN-LTA coexposure is different, inasmuch as CD18 neutralization reduced TVX/PGN-LTA- but not TVX/LPS-induced PMN accumulation (Fig. 3.7). This finding that LPS-induced hepatic neutrophil accumulation is CD18- independent is consistent withprevious reports (229). However, that PGN-LTA- induced hepatic neutrophil accumulation is CD18-dependent has not been reported to our knowledge. Further studies are required to understand the difference in mechanisms of PMN accumulation between these two stimuli. TVX enhanced the induction of cytokines by either LPS or PGN-LTA, therefore cytokines were measured to determine if CD18 neutralization affected the induction of proinflammatory cytokines. CD18 neutralization did not significantly affect the TVX/LPS induction of lL-6, lL-10, TNFa, IL-1B, KC, MlP-1a, 116 lFNy or MCP-1 (Fig. 3.8). Previously, TNFa was found to be critical for TVXILPS- induced liver injury (216). The finding that CD18 neutralization did not affect TNFot induction suggests that PMN activation and TNFor activity either represent separate hepatotoxic pathways or that PMN activation is downstream of TNFa in a pathway involved in the pathogenesis in TVXILPS-cotreated mice. Similar to TVX/LPS, CD18 neutralization did not affect the TVX/PGN-LTA induction of lL-6, lL-10, lL-1B, KC, MlP-1a or IFNy. However, it did attenuate the TVX/PGN-LTA-induced increase in TNFa and MOP-1 (Fig. 3.8). Since lL-6, KC and lL-10 were unchanged by CD18 neutralization, Kupffer cell activation was probably not affected, and the attenuation of TNFor and MCP-1 may be neutrophil-dependent. Neutrophils can produce and release MCP-1, and the depletion of neutrophils significantly reduced MOP-1 production induced by the injection of apoptotic cells into the peritoneal cavity of mice (230, 231 ). Therefore, it is likely that the attenuation of MCP-1 induction by CD18 neutralization is due to the reduction in hepatic neutrophil accumulation. Similarly, the reduction in TNFa concentration by CD18 neutralization might also be due to the attenuation of hepatic neutrophil accumulation, inasmuch as neutrophils express TACE on their extracellular membrane. This enzyme is critical for TNFa cleavage and release (232). Whether TVX/PGN-LTA-induced liver injury requires TNFa is unknown, therefore protection by CD18 neutralization could be due to PMN inactivation or to reduced concentrations of TNFa. In summary, TVX synergized with a modest inflammatory stress induced by either a gram-negative or a gram-positive stimulus to cause liver injury in mice. 117 TVX enhanced the LPS- and PGN-LTA-induced increases in proinflammatory cytokines. However, TVX did not enhance the hepatic neutrophil accumulation driven by either of these stimuli. CD18 neutralization attenuated TVX/LPS- and TVX/PGN-LTA-induced liver injury, suggesting that PMN activation plays a critical role in injury progression in both models. CD18 neutralization attenuated TVX/PGN-LTA induced increases in hepatic neutrophil accumulation and TNFa and MOP-1 plasma concentrations. In contrast, it did not affect TVX/LPS-induced hepatic neutrophil accumulation or proinflammatory cytokine increases. The results suggest that inflammatory stress induced by either a gram-positive or gram-negative bacterial products could play a role in the development of TVX- induced hepatotoxicity and that the pathogenesis is CD18-dependent. 118 CHAPTER 4 Shaw, P.J., Ganey, PE. and Roth, R.A. (2008). TNFa acting at both p55 and p75 receptors is essential for synergistic hepatotoxicity from TVX/LPS coexposure. Submitted to JPE T 119 4.1 Abstract The use of trovafloxacin (TVX), a fluoroquinolone antibiotic, was severely restricted clue to an association of TVX therapy with idiosyncratic hepatotoxicity in patients. The mechanisms underlying idiosyncratic toxicity are unknown; however, one hypothesis is that an inflammatory stress can render an individual sensitive to the drug. Previously, we reported that treatment of mice with TVX and lipopolysaccharide (LPS) induced tumor necrosis factor a (TNFa)-dependent liver injury, whereas TVX or LPS treatment alone was nontoxic. The goal of this study was to elucidate the role of TNFa in TVX/LPS-induced liver injury. p55"' (TNFR1) and p754' (T NFR2) mice were protected from hepatotoxicity caused by TVX/LPS coexposure, suggesting that TVX/LPS-induced liver injury requires both TNF receptors. TNFor inhibition using etanercept significantly reduced the TVX/LPS-induced increases in the plasma concentrations of several cytokines around the time of onset of liver injury. However, despite the reduction in chemokines, etanercept treatment did not affect the TVX/LPS-induced hepatic accumulation of neutrophils. In addition, etanercept treatment attenuated TVX/LPS-induction of plasminogen activator inhibitor-1 (PAl-1), and this was associated with a reduction in hepatic fibrin deposition. Mice treated with TVX and a nontoxic dose of TNFa also developed liver injury. In summary, TNFa acts through p55 and p75 receptors to precipitate an innocuous inflammatory cascade. TVX enhances this cascade, converting it into one that results in hepatocellular injury. 120 4.2 Introduction Trovafloxacin (TVX), a fluoroquinolone antibiotic, is one example of a drug for which use was restricted severely due to lADRs. TVX was approved for use in the US. in 1997, and by 1999 its use was associated with 152 cases of serious hepatic events. Of these, 14 resulted in acute liver failure, 5 patients required liver transplants and 4 died (233). One hypothesis regarding the cause of lADRs is that inflammatory stress alters the toxicity threshold of an individual, rendering a normally therapeutic dose of a drug toxic (55, 73). In accordance with this hypothesis, nontoxic doses of TVX and bacterial lipopolysaccharide (LPS) synergized to cause acute liver injury in both rats and mice (58, 216). In this animal model, TVX pretreatment enhanced the LPS-induced peak in plasma TNFa concentration. In addition, TNFa neutralization completely protected mice from TVX/LPS-induced liver injury (216). TNFot is a pleiotropic cytokine that stimulates a number of cellular responses, including proliferation, production of inflammatory mediators, upregulation of adhesion molecules and programmed cell death. Large amounts of TNFa are produced in response to several microbial products, including LPS. TNFa is a key mediator of inflammatory responses, which can result in both tissue damage and host defense (74, 75). The main cellular source of TNFa is macrophages, but several other cell types produce TNFa including mast cells, hepatic stellate cells, endothelial cells, fibroblasts and neuronal cells (80, 81). 121 TNFa plays a critical role in several models of liver injury caused by viral hepatitis, ischemialreperfusion or hepatotoxic doses of LPS (76, 78, 79, 209). The biological effects of TNFa are elicited via two high affinity cell surface receptors, p55 (T NF-R1) andp75 (T NF-R2) (83). The role of each receptor has been evaluated in several models of liver injury. The p55 receptor has been studied more extensively and is important in hepatotoxicity caused by LPS, acetaminophen or carbon tetrachloride (77, 95, 96). In contrast, critical roles for both receptors have been shown only in a few models of hepatotoxicity, such as that induced by concanavalin A, Pseudomonas aeruginosa exotoxin A or adenovirus (234-236). The study presented here was designed to determine the importance of each receptor in TVXILPS-induced liver injury and to evaluate the influence of TNFor on other proinflammatory factors in this lADR model. 122 4.3 Materials and Methods 4.3.1 Materials Recombinant murine TNFa was purchased from R&D Systems (Minneapolis, MN). Please refer for Section 2.3.1 for additional information on this topic. 4.3.2 Animals p55"', p75"', and CS7/Bl6 wild-type controls were purchased from Jackson Laboratory (Bay Harbor, ME). Please refer to Section 2.3.2 for additional information on this topic. 4.3.3 Experimental protocols Mice fasted for 12 h were given TVX (150 mglkg) or Veh (saline) by oral gavage. They were then given LPS (2.0 x 106 EU/kg), TNFa (50 pg/kg)) or Veh (saline) by intraperitoneal injection 3 h later. Food was returned immediately after this closing. Mice were anesthetized with sodium pentobarbital (50 mglkg; i.p.) and killed at designated times after LPS, TNFa or Veh for various measurements. Blood was drawn from the vena cava into a syringe containing sodium citrate, resulting in a final concentration of 0.76%. The left lateral lobe was fixed in 10% neutral buffered formalin and paraffin blocked. 123 4.3.4 Histopathology Please refer to Section 2.3.4 for information on this topic. 4.3.5 Cytokine measurements Please refer to Section 3.3.5 for information on this topic. 4.3.6 Neutrophil staining Please refer to Section 3.3.6 for information on this topic. 4.3.7 Hemostatic system measurements Plasma thrombinzantithrombin lll (TAT) dimers were measured using the Enzygnost TAT ELISA kit purchased from Dade Behring (Marburg, Germany). Active PAl-1 plasma concentration was measured using an ELISA kit purchased from Molecular Innovations, Inc. (Novi, MI). Hepatic fibrin immunohistochemistry and estimation of deposition was done following the protocol described previously with a slight modification (237), ie, artifactual fibrin staining seen within vessel lumens in all treatment groups was removed from quantification calculations. 4.3.8 Statistical analyses Results are presented as mean 1 S.E.M. A student’s t-test or a 2-way analysis of variance (ANOVA) was used as appropriate after data normalization. 124 All pairwise comparisons were made using a Tukey test with the criterion for significance at p < 0.05. 125 4.4 Results 4.4.1 p55"' and p75"' mice are protected from TVXILPS-induced liver injury To determine the contribution of each TNF receptor to TVX/LPS-induced liver injury, p55"' and p75" mice were treated with TVX/LPS as described in Materials and Methods. TVX/LPS coexposure caused significant liver injury at 15 h in control (wild-type) mice. Both p55"' and p75"' mice were resistant to TVX/LPS-induced liver injury (Fig. 4.1). p75"' mice were completely protected from TVXILPS-induced liver injury and had significantly reduced plasma ALT activity compared to control and p55”' mice (Fig. 4.1). Histopathologic examination of livers corroborated this result, inasmuch as lesions of hepatocellular necrosis were decreased in p55"' compared to control mice, and were completely absent in p75"' mice (Fig. 4.1). 4.4.2 TNFa neutralization attenuates TVXILPS-induced inflammatory cytokines and chemokines In a previous study, treatment with etanercept, which is a mimic of the soluble p75 receptor, reduced TVX/LPS-induced increase in plasma TNFa concentration and protected mice from TVX/LPS-induced liver injury (216). TVX/LPS-treated mice were dosed with etanercept to determine the effects of TNFa on the induction of proinflammatory cytokines and chemokines at 4.5 h, a time near the onset of liver injury (see Fig. 3.1). TNFa inhibition attenuated the TVX/LPS—mediated induction of IFNy, IL-6, IL-10, MCP-1 and VEGF (Fig. 4.2). 126 Fig. 4.1. The role of TNF receptors in TVXILPS-induced liver injury. Wild- type, p55"' and p75"' mice were treated with TVX 3 h before LPS as described in Materials and Methods. Mice were killed 15 h after LPS, and plasma ALT activity was measured. Photomicrographs were taken of livers from representative mice from each group. n = 5-8 animals/group. *significantly different from wild-type group; # significantly different from p55"' group. 127 as I .8. I 0.5. \\ § ('III'I) 1.1V 128 Fig. 4.2. Effect of TNFa inhibition on TVXILPS-induced increases in plasma cytokines. Mice were treated with vehicles or with TVX/LPS in addition to etanercept or its vehicle as described in Materials and Methods. Mice were sacrificed 4.5 h after LPS administration. Plasma concentrations of lFNy, lL-6, IL- 10, IL-1B, MCP-1 and VEGF were measured as described in Materials and Methods. n = 4-6 animals/group. *significantly different from respective VehNeh; # significantly different from TVXILPSNeh group. 129 an ... ... m w w o 2 1 sees s... m m m m .. 353a. » caste... TVX/LPS VehNeh TVX/LPS VehNeh .Tm m m m m o seesaw... ..iT|.ll .i Tm . W m m m .. 38332.... TVX/LPS VehNeh TVX/LPS VehNeh *# TVX/LPS VehNeh -r‘1 m m m o 3.52.. mom; a» .TT N m w w o .. 3 £35 7&0! TVX/LPS VehNeh j Etanercept - Veh “...... 130 The increase in lL-1B plasma concentration following TVX/LPS treatment was not changed by etanercept treatment (Fig. 4.2). TNFa neutralization significantly reduced the TVX/LPS induction of chemokines MlP-2, KC and MlP-1a (Fig. 4.3). 4.4.3 TVXILPS-induced hepatic neutrophil accumulation is independent of TNFa Previous results pointed to a role for PMNs in the pathogenesis of TVX/LPS-induced liver injury (Chapter 3). Despite causing a reduction in chemokines (Fig. 4.3), etanercept treatment did not reduce hepatic neutrophil accumulation induced by TVX/LPS coexposure. In fact, etanercept treatment slightly increased neutrophil accumulation (Fig. 4.4). 4.4.4 TNFa neutralization attenuates TVXILPS-induced hemostatic system acfivafion The coagulation system plays an important role in TVX/LPS-induced pathogenesis (presented in Chapter 6). To determine if TNFa plays a role in TVX/LPS-induced coagulation system activation, TVX/LPS-treated mice were treated with etanercept and killed at 4.5 h. The dose of etanercept markedly reduced the TVX/LPS-induced release of TNFa in this model (216). Plasma thrombin-antithrombin (TAT) dimers, measured as a biomarker of coagulation system activation, were significantly increased in TVX/LPS-treated mice (Fig. 4.5A). Etanercept treatment caused a trend toward reduction in plasma TAT dimers, but this difference was not statistically significant. The plasma 131 Fig. 4.3. Effect of TNFa inhibition on TVXILPS-induced increases in plasma chemokines. Mice were treated with vehicles or with TVX/LPS in addition to etanercept or its vehicle as described in Materials and Methods. Mice were sacrificed 4.5 h after LPS administration. Plasma concentrations of MlP-2, KC and MIP-1or were measured as described in Materials and Methods. n = 4-6 # animals/group. *significantly different from respective VehNeh group; significantly different from TVXILPSNeh group. 132 KC (nglm L) MlP-2 (nglmL) MIP-1a (pglm L) ..L N O 90 ‘ l=l Etanercept 00 . * # 3o . 0 _— __ VehNeh TVX/LPS 400 . — Veh 300 l Etanercept | 200 - I a # 100 ‘ l a 0 __ —_ VehNeh TVX/LPS 600 500 l — Veh I=I Etanercept 400 *# 300 200 ..L O O O VehNeh TVX/LPS 133 Fig. 4.4. Effect of TNFa inhibition on TVXILPS-induced hepatic PMN accumulation. Mice were treated with vehicles or with TVX/LPS in addition to etanercept or its vehicle as described in Materials and Methods. Mice were sacrificed 4.5 h after LPS administration. Paraffin-embedded livers were stained for neutrophils, and the number of neutrophils was quantified as described in Materials and Methods. n 4-6 animals/group. *significantly different from respective VehNeh group; * significantly different from TVXILPSNeh group. 134 PMNs per HPF d N 03 h 0| 0’ N on O O O O O O O O O A; L - Veh a Etanercept VehNeh 135 TVX/LPS Fig. 4.5. Effect of TNFa inhibition on hemostatic system dysregulation mediated by TVX/LPS coexposure. Mice were treated with vehicles or with TVX/LPS in addition to etanercept or its vehicle as described in Materials and Methods. They were sacrificed 4.5 h after LPS administration. Plasma concentrations of (A) TAT dimers and (B) active PAl-1 were measured as described in Materials and Methods. (C) Hepatic fibrin deposition was stained immunohistochemically and quanitified as described in Materials and Methods. n = 4-6 animals/group. *significantly different from respective VehNeh group; 1"significantly different from TVXILPSNeh group. 136 300 250 ‘ 3 E 200 - l— 150- < l- 100 ‘ 50 ‘ ~ VehNeh TVXILPS 1200 - * 3 E 1000 ‘ _ Veh g 800 = Etanercept ‘7 600 E n. *# g 400 ‘6' 200i < o ‘* VehNeh TVXILPS C A o g 0014 IE- 0 * u .5 0012 « _ Veh a a a: Etanercept o g 0010- l 1: ._ o.- .E “a 0.008- 3 o t; :- 0.006 . # 7, o In c 0.0“ ‘ '5 .9 g '6 0002- 55, g 0.000 J VehNeh TVXILPS 137 concentration of active PAl-1, an inhibitor of the fibrinolytic system, was increased by TVX/LPS coexposure (Fig. 4.58). Etanercept significantly reduced the TVX/LPS induction of plasma active PAl-1 (Fig. 4.58). Fibrin deposition in tissue occurs if the rate of coagulation system activation exceeds the rate of fibrinolysis. TVX/LPS coexposure caused a significant increase in sinusoidal fibrin deposition in the liver at 4.5 h, which was significantly reduced by etanercept treatment (Fig. 4.5C). 4.4.5 TVX and TNFct coexposure causes hepatotoxicity TVXILPS-induced liver injury is dependent on TNFa (216), but to determine if TNFa alone could interact with TVX, mice were treated with TVX and recombinant murine TNFa as described in Materials and Methods. They were killed 15 h after TNFa treatment, the time of maximal plasma ALT activity in TVXILPS-treated mice. TVX or TNFot treatment alone did not cause an increase in plasma ALT activity (Fig. 4.6). However, TVXfTNFor coexposure increased plasma ALT activity. Histopathological evaluation of liver sections corroborated the lack of injury from TVX or TNFa alone (Fig. 4.7). In contrast, TVX/TNFa coexposure caused hepatocellular necrotic and apoptotic lesions primarily in centrilobular and midzonal regions of liver lobules, and these lesions extended to periportal regions in some severely affected mice. 138 Fig. 4.6. TVXfI’NFa coexposure-induced liver injury. Mice were treated with TVX 3 h before recombinant murine TNFa as described in Materials and Methods. Mice were killed 15 h after TNFa administration, and plasma ALT activity was measured. n - 4-5 animals/group. *significantly different from TVXNeh group; " significantly different from Veh/TNFa group. 139 ALT activity (UIL) 8 O O 2000 - 1000 . — Veh I: TVX Veh TNFa 140 Fig. 4.7. Histopathology of TVXITNFa-induced liver injury. Mice were treated with TVX 3 h before recombinant murine TNFa as described in Materials and Methods. Mice were killed 15 h after TNFor administration, and photomicrographs were taken of representative livers. 141 142 4.5 Discussion Previously, we reported that a nontoxic dose of TVX interacts with a nontoxic dose of LPS to cause TNFa-dependent liver injury in mice (216). The critical role of TNFa in TVXILPS-induced liver injury was based upon TNFa neutralization, however the role of each TNF receptor was not studied. Activation of the p55 receptor results in two main signals: NFch activation and activation of caspases leading to apoptosis (238, 239). Inasmuch as ligation of the p55 receptor can result in NFKB activation and cell death, it is not surprising that the p55 receptor is critical to several models of liver injury (77, 95, 96, 234-236). Similar to other models dependent on TNFor and p55, TVXILPS-induced liver injury was significantly attenuated in p554' mice. It is possible that p554' mice have reduced liver injury due to decreased plasma TNFoc concentrations, since TNFor induction by LPS was found to be attenuated in p55"‘ mice (96). However, p55 was not involved in the production of TNFa by galactosamine/LPS coexposure (240). The function of the p75 receptor is less understood compared to the p55 receptor. Similar to p55, activation of the p75 receptor causes NFch activation, but it does not result in caspase activation (241). The critical role of the p75 receptor in hepatotoxicity is unclear: it is not involved in some models of liver injury that are dependent on TNFa (96, 240) but is involved in others (234-236). Indeed, p75"' mice were completely protected from TVXILPS-induced liver injury and had significantly reduced hepatocellular injury compared to p554‘ mice. 143 Why p75"' mice are completely protected from TVXILPS-induced hepatotoxicity is unclear. It is possible that the p75 receptor is playing two roles in the progression of TVXILPS-induced hepatotoxicity. It has been suggested that the p75 receptor acts to bind TNFa and transfer it to the p55 receptor, resulting in p55 activation at lower concentrations of TNFa (242). Therefore, without the p75 receptor present, the threshold for TNFa—dependent liver injury might be higher than the concentration that is achieved. Additionally, the p75 receptor can cooperate with the p55 receptor to enhance necrotic cell death in response to TNFa (243). To account for the minor hepatotoxicity seen in p55”' mice, p75 activation must also cause minor hepatocellular injury independent of p55 following TVX/LPS coexposure. Therefore, it is possible that combined activation of p55 and p75 synergize to cause extensive hepatocellular necrosis which is not seen when either receptor is absent. In addition, the p75 receptor is involved in the LPS-induced production of TNFa (96). It is possible that the complete protection in p75”' mice resulted from a combination of these mechanisms, inculding a reduction in LPS-induced TNFa production. TNFa was involved in the TVXILPS-mediated increases in several inflammatory cytokines: lFNy, lL-6, MCP-l, VEGF, MlP-2, KC and MIP-10t. The attenuation of lL-6 and MlP-2 after TNFot neutralization was also seen in another model of drug/LPS coexposure-induced hepatotoxicity (244). The reduction of such a large number of cytokines might be due to a decrease in TNFor-driven NFKB activation mediated through p55 and p75 receptor activation (238, 241). 144 Several of the cytokines that required TNFa for their release have chemotactic properties. Therefore, we measured PMN accumulation in livers of these mice. The hepatic accumulation of PMNs induced by TVX/LPS was not decreased by TNFa inhibition. It is thus likely that the TVXILPS-induced hepatic PMN accumulation is mediated by selectins and other adhesion molecules as seen in endotoxemia or by TNFa-independent sinusoidal contraction (227, 245). It is possible that TNFa is not involved in PMN accumulation but is needed for PMN activation, since TNFa can promote neutrophil activation in vitro (246). If TNFor enhances PMN activation and degranulation, it might explain the slight Increase in hepatic PMN accumulation in TVXILPS/etanercept-treated mice, since when TNFa is present the accumulated PMNs might be activated, degranulate and undergo clearance from the tissue. In addition to being critical to TVX/LPS upregulation of cytokines, it is possible that TNFa is involved in the progression of liver injury by enhancing hemostasis. TVX/LPS coexposure caused fibrin deposition in liver sinusoids, and treatment with anticoagulant heparin significantly reduced TVXILPS-induced liver injury (presented in Chapter 6). TNFa has the potential to interact with the hemostatic system in several ways. It can induce tissue factor which activates the coagulation system, but it also increases PAI-1 expression which could depress fibrinolysis (244, 247, 248). Indeed, TVXILPS-induced increases in active PAl-1 and hepatic fibrin deposition were TNFa-dependent, whereas coagulation system activation showed a trend but was not significantly reduced following TNFa inhibition. The results suggest that if tissue factor induction 145 occurs during TVX/LPS coexposure, it is TNFa-independent, but that a slight reduction in coagulation system activation by etanercept along with a more pronounced reduction in active PAl-1 was able to prevent hepatic fibrin deposition. Based on the importance of TNFa in TVXILPS-induced liver injury, we examined whether TVX can interact with a dose of TNFot to induce similar hepatocellular damage. Indeed, TVX treatment prior to a nonhepatotoxic dose of recombinant murine TNFa resulted in significant liver injury. Other studies have shown that TNFa by itself does not cause liver injury in mice but can when administered with galactosamine or a DNA synthesis inhibitor (249, 250). It is unclear from these results whether TVX sensitized mice to TNFa-induced liver injury or vice versa. However, the hepatocellular lesions in TVX/TNFor-treated mice appear similar to those seen after galactosamine/1' NFa coexposure, suggesting commonalities in mechanisms (251). Recently, cytochrome P450 2E1 (CYP2E1) induction by pyrazole was shown to sensitize mice to TNFa-induced liver injury (252). It is unlikely that TVX/TNFa-induced liver injury is related to an effect on CYP2E1 activity by TVX, as TVX treatment alone did not have any effect on CYP2E1 expression (unpublished results). However, to exclude this possibility CYP2E1 activity would need to be measured following TVX exposure. It is also possible that TVX treatment reduced TNFa clearance, and that the prolonged presence of TNFa resulted in cell death. This is consistent with the prolonged presence of TNFor in the plasma of TVXILPS-treated mice compared to LPS treatment alone (216). TNFa inactivation and clearance is mediated by 146 soluble forms of the two receptors (253). It is possible that TVX reduces the cleavage or expression of these receptors, in turn reducing TNFa clearance. Further studies are required to better understand the mechanism by which TVX and TNFa interact to cause hepatocellular damage. In summary, TVXILPS-induced liver injury depended on the presence of both TNF receptors, p55 and p75. The p75 receptor may play an even more important role than the p55 receptor in the progression of TVXILPS-induced hepatotoxicity. At the onset of liver injury, the TVX/LPS coexposure-related increase in several cytokines, active PAH and hepatic fibrin was TNFa- dependent. However, despite the observation that the induction of chemokines was TNFa-dependent, the hepatic PMN accumulation was independent of TNFa. The critical role of TNFa in TVXILPS-induced liver injury was likely through upregulation of cytokines and activation of the hemostatic system. The observation that the liver injury could be reproduced by substituting TNFa administration for LPS supports the critical importance of this cytokine in the pathogenesis. 147 CHAPTER 5 Shaw, P.J., Ditewig, A.C., Waring, J.F., Liguori, M.J., Blomme, E.A., Ganey, PE. and Roth, R.A. (2008). Coexposure of mice to trovafloxacin and lipopolysaccharide, a model of idiosyncratic hepatotoxicity, results in a unique gene expression profile and interferon gamma-dependent liver injury. Submitted to Toxicological Sciences. 148 5.1 Abstract The antibiotic trovafloxacin (TVX) has caused severe idiosyncratic hepatotoxicity in people, whereas levofloxacin (LVX) has not. Mice cotreated with TVX and lipopolysaccharide (LPS), but not with LVX and LPS, develop severe hepatocellular necrosis. Mice were treated with TVX and/or LPS, and hepatic gene expression changes were measured before liver injury using gene array. Hepatic gene expression profiles from mice treated with TVX/LPS clustered differently from those treated with LPS or TVX alone. Several of the probesets expressed differently in TVXILPS-treated mice were involved in interferon signaling and the JAK/STAT pathway. A timecourse of plasma concentrations of interferon gamma (IFNy) and interleukin-18 (IL-18), which directly induced IFNy production, revealed that both cytokines were selectively increased in TVX/LPS- treated mice. Both IL-18"' and lFNy"‘ mice were significantly protected from TVXILPS-induced liver injury. In addition, lFNy”' mice had decreased plasma concentrations of TNFa, lL-18 and lL-1B when compared to wild-type mice. In conclusion, the altered expression of genes involved in type II interferon signaling in TVXILPS-treated mice led to the finding that lL-18 and IFNy play a critical role in TVXILPS-induced liver injury. 149 5.2 Introduction Despite the rare occurrence of lADRs, they represent a serious hazard to public health and are an important issue for pharmaceutical companies and drug- regulatory agencies. Currentpreclinical testing protocols fail to identify drugs that cause lADRs because predictive animal or in vitro models are lacking. Despite the prevalence and threat of lADRs, the mechanisms underlying them are still unknown. As described in Section 1.1.2, we and others have hypothesized that an episode of inflammatory stress can render an individual susceptible to a normally nontoxic drug dose, thereby precipitating an lADR (55, 73, 254). In concordance with this hypothesis, an inflammatory stress renders mice sensitive to TVX-induced hepatotoxicity (56-59, 216). TVX is a fluoroquinolone antibiotic which has seen limited use due to its association with idiosyncratic hepatotoxicity in people. LVX is a fluroroquinolone antibiotic not associated with hepatotoxicity. In accordance with the lack of hepatotoxicity seen in people, coexposure to TVX/LPS, but not to LVX/LPS, was hepatotoxic to mice and rats (58, 216). Global gene expression analysis of livers at a time of maximal hepatotoxicity revealed distinct clustering of LPS/TVX-treated rats. We showed recently that mice cotreated with TVX and LPS also develop hepatocellular necrosis. Similar to the rat model, LVX did not interact with LPS to cause liver injury (216). Here we test the hypothesis that at a time before the onset of liver injury, TVX/LPS treatment of mice leads to a distinct pattern of gene expression. 150 In testing this hypothesis, we identified several genes involved in type II interferon signaling that were changed by TVX/LPS coexposure. IFNy is a proinflammatory cytokine that plays a key role in both the innate immune system and modulation of the adaptive immune system (255). It is a critical mediator of liver injury from several xenobiotic agents (132, 256, 257). Accordingly, we hypothesized that lFNy plays a critical role in TVXILPS-induced liver injury and investigated its influence on the induction of other proinflammatory cytokines. 151 5.3 Materials and Methods 5.3.1 Materials Please refer to Section 2.3.1 for information on this topic. 5.3.2 Animals Please refer to Section 2.3.2 for information on this topic. In addition, IL- 18"' mice and C57/Bl6 wild-type controls were purchased from Jackson Laboratory (Bay Harbor, ME). lFNy”' female Balb/C mice were a kind gift from Dr. Alison Bauer (MSU, East Lansing, MI), and corresponding female Balb/C control mice were purchased from Jackson Laboratory (Bay Harbor, ME). 5.3.3 Experimental protocol In a previous study, mice cotreated with nonhepatotoxic doses of TVX and LPS developed liver injury (216). The dose of LVX was chosen to approximate the ratio of TVX/LVX doses used clinically in humans. Mice were fasted for 12 h before each experiment. TVX (150 mglkg), LVX (375 mglkg) or their saline vehicle was administered to mice by oral gavage and then given LPS (2.0 X 106 EU/kg, i.p.) 3 h later. Food was returned immediately after LPS administration. Mice were anesthetized with sodium pentobarbital (50 mglkg, i.p.) at various times, and sacrificed by exsanguination. The left lateral liver lobe was fixed in 10% neutral buffered formalin and blocked in paraffin within 72 h. The right medial liver lobe was flash frozen in liquid nitrogen for total RNA isolation. 152 5.3.4 ALT activity and histopathology Plasma ALT activity was measured spectrophotometrically using Infinity ALT reagent purchased from Therrno Electron Corp. (Louisville, CO). Forrnalin- fixed liver Iobes were processed and embedded in paraffin. Paraffin sections were cut at 5 pm and stained with hematoxylin and eosin. 5.3.5 RNA isolation Frozen liver samples (50 mg of tissue per sample) were immediately added to 2 mL of TRlzol reagent (lnvitrogen Life Technologies, Carlsbad, California). One mL of the tissue homogenate was transferred to a microfuge tube, and total RNA was extracted with chloroform followed by nucleic acid precipitation with isopropanol. The pellet was washed with 80% ethanol and resuspended in molecular biology grade water. Nucleic acid concentration was determined spectrophotometrically at 260 nm (Smart-Spec, Bio-Rad Laboratories, Hercules, CA), and RNA integrity was evaluated using an Agilent bioanalyzer (Agilent Technologies, Model 2100, Foster City, CA). 5.3.6 Gene array analysis Microarray analysis was performed using the standard protocol provided by Affymetrix, Inc. (Santa Clara, CA). Briefly, approximately 5 ug of total RNA was reversed transcribed into cDNA using a Superscript II Double-Strand cDNA synthesis kit (lnvitrogen Life Technologies, Carlsbad, California). The primer used for the reverse transcription reaction was a modified T7 primer with 24 153 thymidines at the 5’ end (Affymetrix). The sequence was 5’GGCCAGTGAATTGTAATACGACTCACTATAGGGAGGCGG-(dT)24-3’. cDNA was purified via phenol/chloroform/isoamylalcohol (lnvitrogen Life Technologies, Carlsbad, California) extraction and ethanol precipitation. Biotin-labeled cRNA was synthesized according to the manufacturer’s instructions from the cDNA using the Enzo RNA Transcript Labeling Kit (Affymetrix). The labeled cRNA was then purified using RNeasy kits (Qiagen, Valencia, CA). cRNA concentration and integrity were evaluated. Approximately 20 pg of cRNA was then fragmented in a solution of 40 mM Tris-acetate, pH 8.1, 100 mM potassium acetate, and 30 mM magnesium acetate at 94°C for 35 minutes. Fragmented, labeled cRNA was hybridized to an Affymetrix mouse genome array, 430A 2.0 which contains sequences corresponding to roughly 22,600 transcripts, at 45°C overnight using an Affymetrix Hybridization Oven 640. The array was subsequently washed and stained twice with strepavidin-phycoerythrln (Molecular Probes, Eugen, OR) using a Gene-Chip Fluidics Workstation 400 (Affymetrix). The array was then scanned using the Affymetrix GeneChip® Scanner 3000. 5.3.7 Hepatic neutrophil accumulation Please refer to Section 3.3.6 for information on this topic. 5.3.8 Plasma cytokine measurements The plasma concentrations of IFNy, lL-18, IL-6, lL-10, MIP-1or, KC, MlP-2, MCP-1, VEGF, TNFor and IL-10 were measured using bead-plex kits purchased 154 from Bio-Rad Laboratories and measured using a Bio-Plex 200 system (Hercules, CA). 5.3.9 Statistical methods ALT activity and plasma protein concentration results are presented as mean 1 S.E.M. A 1-,2-, or 3-way analysis of variance (ANOVA) was used as appropriate after data normalization. All pairwise comparisons were made using Dunn’s method. The criterion for significance was p < 0.05. The results of the microarrays were analyzed using Rosetta Resolver error models, and ratios were built for each treatment array compared to the VehNeh control using the Resolver System. This analysis calculated a p-value for every gene’s fold change relative to VehNeh using the Rosetta Resolver error model (258). The gene expression change was considered significant if it had a p-value <0.001. Genes were considered regulated if the p-value was <0.001 for 2 of 3 VehNeh-, 2 of 3 TVXNeh-, 2 of 3 LVXNeh-, 4 of 5 Veh/LPS-, 3 of 4 LVX/LPS-, 5 of 7 TVXILPS-treated mice. Average link heuristic criteria and the Euclidean distance metric for similarity measure were used to perform the agglomerative cluster analysis of the treatment arrays using Rosetta Resolver software. Gene expression profile analysis was done using Ingenuity Pathway Analysis purchased from Ingenuity Systems (Redwood, CA). 155 5.4 Results 5.4.1 Development of hepatocellular injury after TVXILPS-, but not LVXILPS-coexposure The timecourse of hepatocellular injury was examined to determine the time of the onset of liver injury. Treatment with TVX, LVX, LPS, or LVX/LPS did not increase plasma ALT activity (Fig. 5.1). TVX/LPS coexposure, however, caused a significant increase in plasma ALT activity as early as 4.5 h which continued to increase through 15 h (Fig. 5.1). 5.4.2 Hepatic global gene expression changes before the onset of hepatotoxicity Hepatic gene expression was evaluated at 3 h after LPS, a time prior to the onset of liver injury. Analysis of Affymetrix Genechip 430 2.0 Array data identified probesets defined as regulated relative to VehNeh-treated mice, and these were subjected to hierarchical clustering analysis (Fig. 5.2). Within the cluster of LPS-treated mice, LVXILPS- and Veh/LPS-treated mice were distinguished from TVXILPS-treated mice (Fig. 5.2). Thus, hierarchical clustering applied to gene expression analysis was able to distinguish TVXNeh from LVXNeh-treated mice. In addition, at a time before liver injury, a separate cluster occurred for TVXILPS-treated mice, the only treatment that produced hepatotoxicity. 156 Fig.5.1. Development of TVXILPS-induced liver injury. Mice were treated with TVX (150 mglkg), LVX (375 mglkg) or Veh (saline) orally and then 3 h later with either LPS (2 x 106 EU/kg; i.p.) or Veh (saline). Mice were sacrificed at various times after LPS dosing, and plasma ALT activity was measured. n= 45 animals/group. * significantly different from 0 h respective control group. # significantly different from all other treatment groups at the same time. 157 *# 3 6000 —-e— VehNeh -. —-t-1— TVXNeh D --e— LVXNeh V —-e— Veh/LPS 3 —er— TVXILPS .— 4000‘ .......... LVX/LPS .2 H 0 (U I- 2000- :r‘ *t *# Om 0.0 1.5 3.0 4.5 0.0 150 Hours after LPS or Veh 158 Fig. 5.2. Hierarchical clustering of hepatic gene expression profiles. Mice were treated with TVX (150 mglkg), LVX (375 mglkg) or Veh (saline) orally and then 3 h later with either LPS (2 x 106 EU/kg; i.p.) or Veh (saline). Mice were sacrificed 3 h after LPS or Veh dosing, and total RNA was isolated from the liver. Gene expression was evaluated using Affymetrix Genechip 430 2.0 Arrays. RNA from each mouse was analyzed using a separate array. Gene expression profiles are analyzed relative to VehNeh-treated mice. Green represents probesets downregulated, whereas red represents probesets upregulated with respect to VehNeh-treated mice. 159 ps0.001; n= 0345 160 5.4.3 Gene expression changes within TVXILPS, TVXNeh and Veh/LPS treatment groups The gene expression changes in the TVXILPS-treated group are important because they can be anchored to hepatotoxicity. TVX/LPS treatment resulted in 2156 gene expression changes compared to VehNeh-treated mice. The gene expression changes induced by TVX/LPS treatment were compared to those induced by TVX or LPS alone revealing a large number of genes changed selectively in TVXILPS-treated mice (Fig. 5.3). The majority (580/773) of these TVX/LPS treatment-selective genes were downregulated. In addition, a large number of genes (983) were commonly regulated by TVX/LPS and Veh/LPS treatment. TVXILPS and TVXNeh treatments resulted in 619 similarly regulated genes. As expected, there were relatively few genes regulated by both TVX/Veh and Veh/LPS treatments which were not also affected by TVXILPS-treatment. 5.4.4 Several gene expression changes in TVXILPS-treated mice are involved in interferon signaling Gene expression changes in the TVXILPS-treated mice were of interest due to the development of hepatotoxicity and were examined further. To do this, a list of genes expressed differently (p<0.001) in the TVXILPS-treated mice compared to all other treatment groups (VehNeh, TVXNeh, LVXNeh, Veh/LPS, and LVX/LPS) was created using TVXILPS-treatment as the baseline for ratio building. 254 probesets were selectively altered by TVX/LPS treatment; of these probesets, 142 represented functional genes as determined by Ingenuity 161 Fig. 5.3. Venn diagram depiction of probeset regulation of TVXNeh-, Veh/LPS- and TVXILPS-treated mice relative to VehNeh-treated controls. Mice were treated with TVX and/or LPS as described in Methods. Three hours after LPS administration, total RNA was isolated from the liver, and gene expression was evaluated using Affymetrix Genechip 430 2.0 Arrays. The number of probesets increased or decreased relative to VehNeh-treated mice is shown. Probesets were defined as regulated if p <0.001. 162 1364 i173 VehILPS 163 Systems analysis. A full list of these 142 TVXILPS-selective genes can be found in Supplemental Table 5.3. The 142 genes selectively changed in expression by TVX/LPS were analyzed using Ingenuity Pathway Analysis to determine highly affected pathways. Table 5.1 Is a list of pathways impacted by the genes selectively altered by TVX/LPS treatment in order of increasing p—values. Pathways with p- values > 0.1 were excluded from the list. In addition to the p-values, the fraction (ratio) of genes affected within each pathway is reported. Of the pathways affected, two which had the highest ratios, low p-values and are involved in several models of liver injury were JAK/Stat signaling and Interferon signaling (Table 5.1). Type I and II interferons signal via specific receptors through JAK/Stat signaling within cells (259). To examine further the potential role of interferons in TVXILPS-induced gene expression changes, genes in the set of 142 TVXILPS-selective genes were examined for their relationship to interferons. Of the 142 genes, 26 (18.3%) are regulated by interferons (Table 5.2). Of these IFN-regulated genes, all but 3 were increased in expression. A number of the genes have potential importance in the development of hepatotoxicity by being pro-apoptotic, chemokines, pro-inflammatory or involved in immune responses (Table 5.2). The majority of these genes were regulated by IFNy. Accordingly, we examined the role of lFNy in TVX/LPS- induced liver injury. 164 Table 5.1. Pathways highly affected by the 142 functional genes selectively changed by TVXILPS-treatment compared to all other treatment groups. Mice were treated as described in methods. Three hours after LPS administration, RNA was isolated from the liver, and gene expression was evaluated using Affymetrix Genechip 430 2.0 Array. Probesets were defined as regulated if p<0.001. The 254 gene-associated probesets changed by TVXILPS-coexposure when compared to all other treatment groups were analyzed by Ingenuity Pathway Analysis. Of these 254 probesets, 142 were identified as functional genes. The pathways highly affected (p <0.1) are listed along with the ratio of the number of genes changed selectively by TVX/LPS to the total number of genes in the pathway as identified by the Ingenuity database. The specific genes selectively changed by TVX/LPS that are in each pathway are listed. Some of these genes are involved in more than one pathway and are, therefore, listed more than once. 165 5000 .55 5005 .255 .5592 2.50:2 50.5 8.585 0525.065 cacao: 8.22.2 .5225. 505.5 205.5 2.50:2 59o 8055.5 55.295 08382 0.2.2.2 .225 .500 2.me2 $55 8.5555 55.205 5-... ER .2555 .5005 .050 58.5 8.525 55.205 2552ch cozmzwom meow .5 5.2.2 .205 .2220. .2.._0m_> 58.0 8-52.5 :8 20.95 0:80:55ch cease... 50... .5595. 2.50:2 58.0 8-52.5 . 55.205 .28ch 93.2 5x22: .555 5.22.2 .2522 .5595. 2.50:2 ~30 3.58.. 55.855 50.0.2 252“. .59 .235 .5202 53.5 8.58.? 8558 5x552 mmw 2.50:2 Sod 8.553 5.859228 as s 55.20.... 06%: 2x02: .225 .50“. .55005 2.50:2 Ego 3-52.5 55.29... o; _ .555: 5.20.2 .2 .5: .5225d 020052 .5005 .255 .e0m_m .8052 Sod 8-523 855555: 59ch 52.2.5 20.2: .5005 .5322 .225 22.2.55 .805 .50“. .55005 .5822 2.50:2 59o 8-55.5 550:0... 8:8.ch cases $82 52.2.55 .55 .222 .305 .50... 2.50:2 5.322 220.00 ..mmm: .225 2.550 .220. .5252 53.5 8-52.5 55.205 5580.. 28588.0 50:00 030m 03.2.0 5355a. ...m 030... 166 w¥mm<2 .m._.<._.m .mfiwom .<.mv.u_z .Nmmo< mom—m .vOmzm . Zw._.n_ .mumum. .<_.Zv_n_0 E< 55.2.0.5 22.”. 00.00300. 02.09.0050 085% 220 2.8 0.26 ..00 52.9.5.5 ..-.02 5.52000 mmeo> 05.2.95 5.550558 0300030.. 55.9.5.5 .00.. 65.600002: 0.050020 05.2.0.5 0.20.5.0 2.0.5.5505 c09E< 05.2.90 .3523“... 05.2.95 .2080. 009000.02. _>.< 20.28. 9.5 032 167 Table 5.2. Genes selectively altered in expression by TVXILPS that are regulated by interferons Mice were treated as described in methods. Three hours after LPS administration, RNA was isolated from the liver, and gene expression was evaluated using Affymetrix Genechip 430 2.0 Array. Probesets were defined as regulated if p<0.001. Of the 142 genes changed selectively by TVXILPS-coexposure, the 26 listed have been reported to be regulated by interferons as determined by Ingenuity Pathway analysis. 168 5.500.025 .50... 0. 00290. 0.5 4 0.00.00 02.100.00.00 00.52020 .(0010 00000.2. 0500050. . 0000 0500050. I 00.00.8000. 02.0025 0.... 0 m 4 000.00 000200.050 0.2002 000.52 .0 N.N0.2. 020.00.002-20 . w 0500... 0500... I .553500 22.. .0 0.02.5. 50.0>..00 2 mm 4 05000. 0.0.0.0 00.02.00-509... 9.00.2.2 .0 080.2. . 0000 I I h .. 4 050200090 00.0.:0...5 00.0.0.0. 0N0w_ .0 0 0000.2. 2.2000000 20. 4 . .200. 00.0500. 00.0.0.0. Em. .0I0lom2022. 090550.222. 0... 4 2 .2080. 2 0.0.8.0.... 20:... .mmmoaomv. 0.0 4 05000.0 0002.0. 0Ew000 00.2.0.0. 00.0. .0 2.2.2. . 2000 I 02.02.00 000 0.0.0.500. 00.09.00. 0 .2 4 . 2:00.00. 00.50.00 22.00.00. ...—<0. .0 .002N2. 0500050. . < ._ 5520 - I 00:00.. .00..0.0050.0 000000 . 01.2 2 N. 4 0.0.0.000 0:5..0008005... .00.). < (..I ..0 2NmmN2. 0500050. . . - I I 0.555. 02.02050... $0.00 _ 0:2 2. 4 8.00. 0 N ......n_.358o.m.: 0.. u: .0 x .852. - . N 0200.0... 00000000 I 0.6.080 o... 5 2 4 50.”. was 5.8.8.0250» 5.0., ~55 .0 5505.2. 8.00.9.0 0.09.39. .00.0.2 8. 4 5 0:05.. 0.60. 0-x-0. 00059.0 30x0 02.83. 00.20.0000 2.00.500 .0N-n__s_ 2.00 4 N 0000.. 2.00. 0-x-0. 00.200000 N._oxo .0 00N0.2. 0:505 :00 0255.0 ..00 0.0 D 50.00.00 .0000 «.010 .0IxI0Nm2m2. .. . . . . 0.... 9.0 085.6. 5.; 2.8.09.0 . N .m 550.0 00.20.08 I . 0 4 0.00.00 x..00-002-x..00 0.50m NmI._Im .0 080.2. 0.2.80-0... 5.2 4 0.38.00.25.00 2mm 0:. .202. .0000. 0. 0000.308. 3.00.00 000000 02030.0”. 0.00.. 0000 30:50 .0000 0.0". 0000 00000000 «.0 0.00... 169 0000.30. 0.000230. 00.050.000.00 02.03080 00.20E000 .0 00.2008 00.000.000.00 55005 0>..00.x0 0. 0500050. 00..000.5 2.5.2.... 0. 0500050. 2000000. 02.0002 00..000.5 2.0.2.... 0. 0500050. 2000000. 02.0002 000500.. 00. .0.00. .9000 00.9.0... 2.5.22.2 0.0500050. 2000000. 02.0000 5000000 . 015:0 00.550020 0.0 0.. 0.2 0.. ..om 0.0. 0.. 0.. 0.0 N. 44444444>> < .200. 0.2.0.0 .0..0...0000 .0.3050> N 020.00.. 0 2.... ..0. .0 05500035 0 ....20 0 0000005000 .0 0.02.00 000 0030500.. .0020 .00000000..E .N 050.3E5.0 00.08.0030 0 00..000.5 00.2008 .0 35500030 N 00..000.5 00.2030 .0 55.50.0030 20000500.. 00. .0.00. .02020 00.038-00.20 0500.0.0.0..00 .0024. N 00... 8.0000. -000 .050.0..050..0 00.50.... 0.0.0.0 0 .000 000.0 ...03035 0E0500.0.0 .< <00w> NOSSw 0._.<._.w NDOw MwOOw NwOOw N<. .O._m 0_n_ IFN-I- 180 Fig. 5.8. Plasma concentrations of proinflammatory cytokines are reduced in lFNy"' mice. Female, Balb/C wild-type and lFNy"' mice were treated with TVX and LPS as described in Methods. Mice were sacrificed 6 h after LPS administration, and plasma concentrations of TNFa (A), lL-18 (B) and lL-1B (C) were measured by Bio-Rad bead-plex. Wild-type controls and lFNy"' mice had equivalent baseline concentrations of TNFa (210 1 41 pg/mL), lL-18 (25 1 10 ngmL) and lL-1B (75 1 16 pg/mL). n = 5 animals/group. * significantly different from wild-type controls. 181 A Wild-ty pe IFN-I- ... ... * N * N * .ir .ir w w .III. ...-M .II. M .l]. w w mmmommmmmommmmmm. 3 .52: cuzp 3 see c w... 3 .553 n 3. B C 182 5.5 Discussion This study confirmed previous results that TVX-pretreatment, but not LVX- pretreatment, interacted with an inflammatory stress induced by LPS to cause liver injury in mice. We extended this to define the early timecourse of hepatocellular injury as measured by plasma ALT activity. TVXILPS-treated mice had a small but significant increase in plasma ALT activity as early as 4.5 h after LPS, whereas there was not a significant increase in plasma ALT activity in any other treatment group at any time measured (Fig. 5.1). Based on these results, hepatic gene expression changes were examined at 3 h, a time just before the onset of liver injury. Similar to results obtained in LPS/TVX-treated rats after the onset of liver injury (58), we observed that TVXILPS-coexposure resulted in unique gene expression changes in mice prior to the onset of liver injury (Fig. 5.2). The distinct gene expression profiles were observed despite differences in species, timing, and routes of LPS and TVX administrations in the two studies (58). These results in mice suggest that global gene expression change is an earlier marker of liver toxicity than plasma ALT activity in this model. Although, numerous groups of genes might be involved in injury, the set of genes selectively affected by TVX/LPS was chosen as a starting point for mechanistic analysis. There were several pathways affected by TVX/LPS- treatment that suggest that TVX magnifies the inflammatory response induced by LPS treatment (Table 5.1). This finding is consistent with the finding that TVX pretreatment enhances the plasma concentrations of several pro-inflammatory 183 cytokines induced by LPS (See Fig. 3.2). Of the pathways impacted by TVX/LPS- treatment, JAK/STAT and interferon signaling pathways had among the highest ratios and low p—values. The actions of lFNy are exerted through the activation of the JAK/STAT pathway (259). Of the 142 TVXILPS-selectively expressed genes, 26, or 18%, can be regulated by interferons (Table 5.2). Furthermore, a significant number of these genes have roles in functions related to hepatotoxicity, such as apoptosis, leukocyte migration, pro-inflammatory cytokine production and immune response. Most (23/26) of these IFN-regulated genes were enhanced in expression in TVXILPS-treated mice, suggesting an increase in interferon signaling. The majority of these are known to be regulated by lFNy, rather than by lFNoc or IFNB. lRF-1, one of the genes selectively upregulated by TVX/LPS, encodes for a transcription factor activated as a result of JAK/STAT activation by IFNy (259). This pathway is activated in several other models of liver injury, including concavalin A, acetaminophen, ischemialreperfusion and LPS/galactosamine (115, 116, 131, 132). However, the role of IFNy or IL-18 has not been examined in a model of liver injury from drug/inflammation interaction; therefore, the role of IL- 18 and IFNy was explored in TVXILPS-induced injury. The first step was to measure the plasma concentrations of lL-18 and IF Ny. IL-18 stimulates IF Ny production (260). The plasma concentrations of both IL-18 and IF Ny were selectively increased by TVX/LPS treatment (Fig. 5.4A, B). This enhancement and prolongation of the lFNy induction by LPS has not been shown previously in any drug/LPS-coexposure model of liver injury. The mechanism by 184 which this interaction occurs is unknown. However, we reported previously that TVX pretreatment prolongs the LPS-induced plasma TNFa peak (216). It is thus possible that this prolongation of the TNFa peak drives the prolongation of the plasma IFNy peak. Based on the selective increases in lL-18 and lFNy in TVXILPS-treated mice, we examined the role each cytokine plays in the development of TVXILPS- induced hepatotoxicity using transgenic mice. IL-18 and lFNy are components of the same signaling pathway. Both lL-18"' mice and IFNy”' mice had significantly reduced hepatocellular injury following TVXILPS-treatment compared to their respective wild-type controls (Fig. 5.5 and 5.6), implicating pathophysiological roles for lL-18 and IF Ny. Female Balb/C mice were used to generate the data for Fig. 56-8. The reason for the gender and strain change related to the availability of lFNy"' mice of this background. Despite the wild-type controls having lower plasma ALT activity in response to TVXILPS-treatment, the extent of histopathologically evident hepatocellular injury was similar to that seen in male C57Bll6 mice. It is unknown whether the difference seen in the magnitude of ALT activity increase in response to TVXILPS is due to a strain or gender difference. Regardless, the pathway by which lL-18 increases lFNy is common to both male CS7BI/6 and female Balb/C mice. Thus, the observation that both male 057BI/6 lL-18"' mice and female Balb/C |FNy"' mice were protected from TVX/LPS- induced liver injury suggests that the importance of this pathway in TVXILPS- induced liver injury is not gender— or strain-specific. 185 Several inflammatory cytokines and chemokines were measured in wild- type and |FNy"' mice treated with TVX/LPS at a time before the onset of liver injury. This time was selected to exclude the possibility that a difference seen was a result of injury. The plasma concentrations of several cytokines were unchanged, but TNFa, lL-1B, and IL-18 were significantly reduced in lFNy”' mice. Previously, TNFa inhibition attenuated TVXILPS-induced liver injury (216). IFNy enhances the production and release of TNFa, as shown in vitro (121). In vivo, lFNy induction of TNFa production is mediated by lRF-1 (261), a transcription factor selectively upregulated in TVXILPS-treated mice (Table 5.2). Interestingly, TNFa inhibition reduced the TVXILPS-induced increase in IFNy (see Fig. 4.2), suggesting that each of these cytokines regulates the expression of the other. Additionally, lFNy"' mice had reduced levels of lL-1B and lL-18. These cytokines exist as pro-forms of the proteins in cells and are cleaved by caspase 1 to their active forms. The mRNA levels of IL-16 and lL-18 were unchanged in gene expression analysis. Therefore, we hypothesize that IFNy positively feeds back to increase caspase 1 activity either directly or indirectly. The consequent increase in lL-18 could then cause more production of lFNy, potentially resulting in a proinflammatory vicious cycle. lFNy causes apoptosis in primary hepatocytes (262), and it is possible that TVX directly sensitizes hepatocytes to IFNy-induced cell death. Additionally, TVX enhanced the LPS-induced increases in both TNFa (216) and lFNy, which synergize to cause primary hepatotocyte cell death (263). It is also possible that 186 Fig. 5.9. Hypothesized role of IFNy in TVXILPS-induced liver injury. Dashed arrows represent untested hypotheses. LPS causes hepatic neutrophil accumulation and TNFa release. TVX enhances the LPS-induced increase in TNFa (216). TNFa can increase lL-18, which in turn induces lFNy production. In addition, lFNy can feedback to increase both lL-18 and TNFa (Fig. 8). This has the potential to create a vicious, proinflammatory cycle of lFNy and TNFa production. In addition, PMNs migrate into tissues and become activated, causing death of hepatic parenchymal cells. It is possible that lFNy enhances PMN activation. In addition, TVX might act directly on hepatocytes to sensitize them to cell death induced directly by lFNy, lFNy/TNFa coexposure or toxic factors released from PMNs. 187 TVX acts directly on hepatocytes to enhance TNFor/lFNy-induced cell death. Fig. 9 illustrates the proposed pathway to TVXILPS-induced liver injury and the interaction between TNFa and IFNy. In addition to effects on other proinflammatory cytokines, IFNy can play a direct role in the regulation and activation of many cell types that might be involved in liver injury. For example, PMN activation is critical for TVX/LPS- dependent liver injury (see Fig. 3.6) and IFNy can increase PMN activation in vitro as measured by oxidative burst and differential gene expression (118, 264, 265). Although PMN accumulation after TVXILPS-cotreatment was unchanged in IFNy”' mice, it is possible that activation of accumulated PMNs was decreased in these mice, and this could be a reason for the reduced hepatotoxicity in response to TVXILPS (as depicted in Fig. 5.9). In addition, IFNy plays a role in the activation of CD8+ T cells and macrophages (104, 266). Accordingly, it is possible that IFNy is involved in hepatotoxicity by activating one or more of these cell types. In summary, TVX interacts with LPS, but not LVX, to cause liver injury in mice. That hepatotoxicity only occurred for the drug with lADR-potential in humans raises the possibility that inflammatory stress might play a role in the pathogenesis of idiosyncratic liver injury caused by TVX in people. Gene expression analysis revealed distinct clustering by treatment at a time before the onset of liver injury. A small group of genes was identified which changed in expression only after TVXILPS-treatment compared to all other treatment groups. Of these genes, a large number were related to interferon signaling. This 189 observation led to experiments that demonstrated the involvement of lL-18 and IF Ny in the pathogenesis of TVXILPS-induced liver injury in mice. 190 Supplemental Table 5.3. Functional genes selectively changed by TVXILPS- treatment compared to all other treatment groups. Mice were treated as described in methods. Three hours after LPS administration, RNA was isolated from the liver, and gene expression was evaluated using Affymetrix Genechip 430 2.0 Array. Probesets were defined as regulated if p<0.001. The 254 gene- associated probesets changed by TVXILPS-coexposure when compared to all other treatment groups were analyzed by Ingenuity Pathway Analysis. Of these 254 probesets, 142 were identified as functional genes. The specific genes selectively changed by TVX/LPS are listed. 191 mEmEou coma: ac: £55; :8 .8536 :8 mm. > use 5855.. 55, 269.555 510 scrmrmmmvmi mm b 2% cmma 5me 5 ~82: ommEmc S .9525 <20 2 $839 3258995 m. 3 4 $2: 252855.565 32x8 5:83va cozmzwom =8 c. 265% .8582 955:: we b 2822: £8 38 6:883? 951 cemmoao. o.» 4 2:898 8&8 «moo eezmoo 6:858va we 4 o 8.59:8 555% __8-uc__co ooooo 5U 58: 3 b 925:8 22.8 55 598 5 82m: 3 > F 5255 85583 296 =8 FzEnZo eminence: v 9:59:00 5 b -582 as 6.323% coma. Humans: m 9.39:8 3 4 -589 as 3.3233 8mm 56881 N .m mmmfi .mEEmEoo to 4 5558 5.2.5825: 25cm 851m swimwear: 2558953 92 4 5.3259229 .28 51:2va omcoamo. 0.52:5 BoEB macaw 52625ch 59.5 _ 0:5. mm b 255 2 8555 5:55:52 5.2%. “mime EN: 3 4 mm 52.8 525995 5: E m8E< 5:258: mm b >.< 59989.85. 20% afloat: Z 4 38.8: xmc< can? a 35:: 9x82. 2 528.8. 3” 4 .5 557555852 vfieoz< “micrometer oomommo xn_<.>_ m9m>=om mdm 4 6382 2995.? Na «mm—9‘ “argue”: lounges". .025» £30 .235 E soc—wrong 33:39". Eon. 532.63. 2:2. 2.00 980 cocoa—cow Qm aim... 39.028396 192 m._.<._.w w9m>=om .:o=m>=om xmwimSZ oomommo ¥a<_2 8 xomnuooc o>=mmoc 8380 x532 so 8883 5268: £288. :8 8 :28.qu magma 8:20:88 .2858: .nNdzz 820.88 .2888: .5N-n:.2 c8888.: oEEEocoofio mé o.m QN m.N_. m.N_. 06 ms Wmm N.m N.N m.m cor wdw o.N fin o.N 9v 0N 44>>><<>>>> >44‘4>< o 88$ 888cm: 2889.3. o..=w < gcznam .m 889. 582 8:955: 25:93 <95 Eon N :csnam N 883 8825 88.25: 25283 $35 8889 Loss 53ch 55528 meow m $92888 3850on .25 woman S 322585 858% .98 2.88 mm 8652633128 $2.28 N 8980.: 283 52.8 3..-:8 Emma 2 $8885 x8. 98 2x8 :R 83 559528 258 9 0:8: @5ch .88 x oEomoEoEo m _. ”.moxo m .88: £55 0-x-ov 558558 2on N 959. 955 0-x-ov 55.5258 «.88 88:8; 08.808 3.5 we; «88me x8-> xmo owmaoemc«53506-0-.05880 H.200 F 88895 528.0 E.z_._o 3 Enzo-0 $2040 988 mmmsx 950:0 (x10 66:339. science: scrmmoevi 5:828: 5:153: scmcueovmeee cc 5 sconce, smhwoemmee .5 m mmsmmee en ooemmv. 51583.3 5:853: 5:88,: scmmvmmvi 55:85:? on «memmes .mrmvmomi Cc c.eo~omvs 8.2.8. a.» cane 5:55.285 193 0000.0 02.0.0.8 03.000900. 00.8000. 088:... 52.0.8008 009.00 . 01.2 008000. 00:08.. 50.0.8008 009.00 . 01.2 832.00 mumz .0 02.0.3000 0>...000 80.020.008.08 0.8800008 0.8800008 de V.N_. V... 50 N0 mé ©.N_. 0.0N NV ...N 0.0 m0 fin. .... MN ...m mN QN 44>>4>>>>>4>>><44 F 000000.08 080... < ._ 000.0 90.0800 9.300058%... 5.02 8.00. o .N 28.008020... 0 .0 2.0.0.030 0.90... 509800.00 .900 N .0 2:00.030 0.900 .c09.c00.:0 .900 F 000.080.053.000 .2 0.0500029008000030 .. 0.8.0 8.0.08 8.8.8.. 000 P 000200020 .2300 0.50.00 .0009. 050.080.080.20 8.0000. 80.0020 003:0 0.50.00 00088008000820 0 00.0.8: 00.3.0“. @0808... 020808 .0..> 088800000 00.85 am“. 00..> 0. 0.0.08 .80.”. 00059.0 0.00 3.0“. N 00.008; 0000008 on 0E.> 0.00.00.85.80 0.0-> P 8.3.2:. 0.000000. 8.0000. mmmm. SAC—2.... <.<._I 0..-NI $00400 N<0..OO _.._.8.0EE0..5-08 0.03.0000 0.2.. .0 0.052 00.02.00 >0>>:.00 00.0.2 .0 :0..0.:00. 0>...000 02“.. 0:0 02.... 00.0.0000 0..0.0000-0.0 8905800508 :0..0>..00 0:0 808.880. 0.89.00. 0.0 0.0 0.00 0.0 0.0. N... 0.0 oNN m. .... n. 2. N00 0.0 0.0 méu 0N 0.0 0.0.0. 0... >4><<444444>>4>44> 00 000000 000.000 5.0.00.2 0000.. 50.08 05050 000 .0802 0.205. 0 0005.. 000:0. 000:... 0.0.90 00.02.00-609... 00.0032 0 0005.. 000:0. 0.0.90 00.02.00-509... 00.0032 5.00. 02. 08:05-00. .5: 8.0000. 508.000.. >..0:00 2.0.. «in... 8.0000 05.0:90 000005-025 EN ..<<_v. 080008088 0 :2. mznw 50.08 . 05050 <52 02$ 00:02.5 0m<_.mz>. 0:00 0000.0:c0x0 00.0.0.5.0 888.5 000.0. .. 8.00. 0.8.0.000. 8.0.00.5 Em. 4.8.0000. n. 5.00.8.5 <05... 000.00 000005 05500 8.0.8.5 n.._.0_ . 50.08 05050 8.00. £2,000 00...-5305 .0000. . 8.0.000. .0.:0E00_0>00 00.08.090.008. Em... 0 00.00... <20 .0 5.5.00. 00_ .0000. . 0500.08 8.00200 8.2.00.0.5 .520. n. 508.000.0380.. 80.00: 000080088... .0002... .0H0H0000.0. .0 0 000000. .0:0000.0. .0:0|000.00. .0H0000.0. .m_000.00. .0 m 00.000. .0 0000.0. .0|0|.00000. .0H0H0000.0. .0m800. .0 000000. .0|.0.0.0. .010.00.0. .mlm000.0. .0 0 000000. .0Ihmovwv _. 000000000. .0008. 0.0 0.00. 300520000 195 0:00 .0..0:0..00..E .0. 0.508000. 0.00550 .00”. 5 00295 :0..0>..00 9.02 00000.05 .:0..000.000 0.240 82,500 00.0.2 .0 :0..0.000. 02.000: 00:0000. >8.0:.E0..5 05.080 «...-0 00:0000. 000..0 0>..00.x0 :0..0.0.E 00.09.00. 000000.000 x...0:. .0.0..000..x0 N0 NNN 0.0 ...m 0.0 FNV 0.0. 0.0 odh 0.0 0.00 0.0.. 0.0 0.0 0.0 Nov 1N.- 0.0 4 <>>>4 4 4 b 4} >4 44 444 000.0E>.00 .0..0:0..00...2 m 000:0. 0.5.0.00 000.080.0000 058030008020 0 . 00052-0 0.0.8098 -100005300000800 to. 50:80 02.70.00.980. 500.00... 050000 -0520 .00 000.000.0208... . 00000. 0.0.0.0 00.500 ... .00... 00.0.0002 00.0 8.0000. 80802 000.000.8008 00.000.002 0.00 .8..0.:5 0:00-0 5 80880 0:00 00.800200 E9. 0000.. .0 8.00. 80.00: 0.80 00.020. 0.00-0 5 808.80 0:00 00.800200 E9. 0000.. .0 8.00. .0080: 00000800 ..00 80.0008 .0802 00000.90 ..00 80.0008 .982 0:00 008000. 80.58 :0..0..:0.0...0 0.0.022 m: 5000.52.90... 0.. 0000..0000_.0.0:. x...0.2 0....-5088 05808.5 03.). him-.0... 00.00 00.0 <..v.mn:n. ..0..x0 0:0 800.80 00.09.00. >03:.00 00.02 .0 800.000. 02.000 05000.0 2.020... 0. 00:0000. 0.000000. 02.000: 000800008 0000000 . 01.2 .0 05000008 000E00 <20 0. 00:0000. .000.0000-0.0 80008.8 0... YN 0.0.. 0.. 0.0 0.0.. 0.0 N0 0... N... 00 0.0 b 4>4 4V4 4 >4 4>4>44 000508.008 .-0..0.< N 0000.000 00.0000 0:20 5.00.000. .. .< 0.0.0.0 00.00... 20.0.00 8.0 0 50.0.0 .005. 05m. 0 0000.0 0.80. 9.0 00.0.8: 0:0000:0 .0._> 0.00._0:.00:00.0000. .0.-> 5x.00. . 58050.00 .0 8.0.0000 0 50.0.0 0:08:88 0:80.880 0 00... 8.0000.-:0: .000.0:000...0 0500.... 50.0.0 . .<>. 00>. 000.0...0008 0500.... 50.0.0 00.9.8: 50:0. 0:0 000.0:000:0 0 .00... 0.8.0 ...:0000 080000.20 0002.0 80:88 ..8.0.000. 5.00.0.0 <0. ._:0000 0.8.0.000. .. 0008:0008 50.0.0 0.02.0me mmzmw w..mw ...<00 .0 002.". 002m. ..mm xom .2060 00.2.9... NZO...0 ..<.00._.n. Zm...n. 0<_>.wn. mum—n. <03. ..000 .0H00.0.0. .0 000000. .0:~...~0. .0H.00000. .0I000000. .0 .0.0.0. .mmm..o~0. .010:0..0.0. .0 0 .000.0. .0I0I. .800. 0:00.000. 0:02.000. .0|-.000. 0101000000. 0.0.1.8000. 0000000. ......000. 0.0 0.00. 300.00.00.00 197 mwozm m>=m2xo 2 $582 3.2% 252$. 2 $539 xomnumfi m>zmmmz 3.29m 252$ 2 $532 xomnnmmfi m>=mmmz toamcmz c2 .39: >50: 23055: B 8:525 .25 w._. fem adv EN ...m mé EN 9m mum NN n... m6 QN 0N QN m... Nwm <><<>44>4<4>>4444 .mtucocoozE .N $33.56 233.3% m 9:206 3336 E .ommmaqsw N 9.53% 95.23 “.0 83235 P mcEfiEoo EmEou c059. BEE _m>_>5w ND 55895 “.o 55:59 Emacmamu £8< .3322“ :23 2390 89.298 ammofi>£Eavom .mtoamcmz Bow oEEm 2:260 .mtoamcg EN 553%: 92393 Eauow 29.035: :0. .90.: E296 83:09:22; 9.385 3.75% m no: -mctmmEEEEm .863 mc_o__aw v so: -mctmmbcfifia .889. mc_o__am or no: -mctmmEcEEE 35$ mc_o__aw P so: -mEEwBEEQm .883 mc_o__aw v .2525 .2938 ammosEmmE NDOw mwoow NwOOw Fonz—2w Naomgzw mm wOOJw mm4 <4 4 4}} {D 44 0 0.0.000 00342... 00000... 00.N 00000090300 00.0.00x < .900. 0.30.0 .0..0....0000 .0.:000> . 000000000000 00.0.5 .Nm. 000.000 00..00:_000-0...:0.0: UN 0.00 0.30.... 0 0.0.0.0 000000.802... 0 0.0.0.0 00.0.00000 00.0000. 000000.. 0.03.... . 500000000800... N 0000.80....0000.... _. 0800050.. 3.00. 000005-000» 0000 0003.21... 00000000000550 00.00;... 00.003 0.E00.0.000>w N 00.00.00 0 03. ..E .0 0000000000 0....2w . 0.0500 00.050 6.0.0 0 0000000000. .0 5.02.00 000 0000000.. .0005 m 00.0.0.000 0.0.000 5.000.. 0.0.. 8.0.2-030 N 000000 .0885 N 00.0.0.000 0.0E00 ....om. moz Pan—D .NmmD ONmmD... mun—20.2... Mun—(KI... 5th N20... 0.0.0.. 6...... ...<._. n..moz>w NOEDw 2.me m._.<...w mom—(Pm N0<...w Nommaw .0wmn0000.0. .0 000.00. .0H000000. .0 000000. .0H0.0000. .0n.00000. .0 000000. .0ImN ..ng .mmm00000. .0 0 0000.0. .0 mw000000. .0:0000.0. .0u000.00. .0 000000. .0wmw.000.0. .0 000000. .0I0I000000. .0I0I000000. .0H000.00. .0 000000. .0008. 0.0 0.000 0.0052005 199 CHAPTER 6 Shaw, P.J., Fullerton, A.F., Ganey, PE. and Roth, R.A. (2008). The role of the hemostatic system in a murine model of idiosyncratic liver injury induced by trovafloxacin and lipopolysaccharide coexposure. 200 6.1 Abstract The use of the fluoroquinolone antibiotic TVX was severely restricted in 1999 due to its association with idiosyncratic hepatotoxicity. Previously, we reported that a nontoxic dose of TVX interacts with a nontoxic dose of LPS to cause robust hepatocellular injury in mice. This interaction with LPS was not seen in mice treated with LVX, a fluoroquinolone not associated with hepatotoxicity in people. TVXILPS-coexposure caused an increase in plasma ALT activity as early as 4.5 h after LPS administration and which progressed through 15 h. We examined the role of the hemostatic system in TVX/LPS- induced liver injury. At the onset of liver injury, coexposure to TVXILPS, but not exposure to TVX, LVX, LPS or LVX/LPS, caused increased plasma concentration of thrombin-antithrombin dimers and decreased plasma circulating fibrinogen. LPS treatment induced a small increase in plasma plasminogen activator inhibitor-1 (PAl-1) concentration, and TVX pretreatment enhanced this effect. TVX/LPS coexposure also resulted in hepatic fibrin deposition. Anticoagulant heparin administration reduced TVXILPS-induced hepatic fibrin deposition and liver injury. PAl-1"’ mice treated with TVX/LPS exhibited similar fibrin deposition to wild-type mice but had significantly reduced hepatocellular injury. PAl-1"' mice, but not heparin-treated mice, had reduced plasma concentrations of several cytokines compared to TVXILPS-treated controls. In summary, TVXILPS-coexposure caused an imbalance in the hemostatic system, resulting in increased thrombin activation, plasma concentrations of PAH and hepatic fibrin deposition. Both thrombin activation and PAl-1 play a critical role in 201 the progression of TVXILPS-induced liver injury, but through different modes of action. 202 6.2 Introduction The hemostatic system encompasses a number of factors involved in the complex interactions of platelets, von Willebrand factor, the coagulation system, anticoagulants and the fibrinolytic system. The activation of blood coagulation occurs predominantly through the tissue factor pathway, which leads to the formation of thrombin from prothrombin. Thrombin is a protease with a number of biological activities including the cleavage of circulating fibrinogen to fibrin, which polymerizes to form insoluble fibrin. Dissolution of fibrin is mediated by plasmin. The fibrinolytic system is controlled primarily by plasminogen activator inhibitor-1 (PAl-1), which inhibits the production of active plasmin. In addition to its role in fibrinolysis, PAl-1 has several other proinflammatory properties (146, 147). The coagulation and fibrinolytic systems exist in a delicate balance to prevent widespread blood loss while controlling fibrin deposition. If the balance of these two components is altered, a possible outcome is unregulated activation of the hemostatic system, which could lead to fibrin deposition and production of occlusive thrombi. These in turn have the potential to alter blood flow and result in local tissue hypoxia, thereby contributing to tissue injury (103, 152, 154). Whether an alteration in the hemostatic system occurs in the TVX/LPS- coexposure model of liver injury in mice has not been determined. The studies presented here were designed to test the hypothesis that TVXILPS-coexposure in mice results in hemostatic system dysregulation and that this plays a role in the development of TVXILPS—induced liver injury. To examine this hypothesis, biomarkers of thrombin activation, active PAl-1 concentrations and hepatic fibrin 203 deposition were evaluated at the time of onset of liver injury. To determine the importance of the hemostatic system in the development of TVXILPS-induced liver injury, mice were treated with anticoagulant heparin. The importance of PAI- 1 in TVXILPS-induced hepatotoxicity was determined using PAl-1"’ mice. Heparin and PAl-1 can have profound effects on inflammation. Heparin attenuates ischemialreperfusion-induced inflammatory responses (153), whereas PAl-1 induces proinflammatory cytokine expression (147). Therefore, we looked to determine what role thrombin activation and PAl-1 play in the TVX/LPS- induction of proinflammatory cytokines, some of which contribute to the pathogenesis in this model (216). 204 6.3 Materials and Methods 6.3.1 Materials Please refer for Section 2.3.1 for information on this topic. 6.3.2 Animals Please refer to Section 2.3.2 for information on this topic. In addition, PAI- 1"' and C57Bl/6J mice were purchased from Jackson Laboratory (Bay Harbor, ME). 6.3.3 Experimental protocols Mice fasted for 12 h were given various doses of TVX or Veh (saline) by oral gavage. They were then given LPS (2.0 x 106 EUIkg) or Veh (saline) by intraperitoneal injection 3 h later. Food was returned immediately after this dosing. Mice were anesthetized with sodium pentobarbital (50 mglkg; i.p.) and killed at designated times after the dose of LPS or Veh for various measurements. Blood was drawn from the vena cava into a syringe containing sodium citrate resulting in a final concentration of 0.76%. The left lateral lobe was fixed in 10% neutral buffered formalin and paraffin blocked. 6.3.4 Heparin treatment Heparin (3000 U/kg, so.) was administered at 2, 6 and 10 h after LPS administration. Mice were killed at 15 h for experiments used in the generation of 205 Figs. 6.5A, 6.7A, and 6.70. Mice sacrificed at 4.5 h (Figs. 6.58, 6.50, 6.8) were treated with heparin only at 2 h after LPS dosing. 6.3.5 Histopathology Please refer to section 2.3.4 for information on this topic. 6.3.6 Hemostatic system measurements Please refer to Section 4.3.7 for information on this topic. 6.3.7 Cytokine measurements Please refer to Section 3.3.5 for information on this topic. 6.3.8 Statistical analyses All bar graph results are presented as mean 1 S.E.M. A 1- or 2-way analysis of variance (ANOVA) was used as appropriate after data normalization. All multiple pairwise comparisons were done using Tukey’s Test. The criterion for significance was p < 0.05. 206 6.4 Results 6.4.1 Changes in the hemostatic system in TVXILPS-induced liver injury TVXILPS-coexposure causes hepatotoxicity in mice, whereas treatment with TVX, LVX, LPS, or LVX/LPS is nontoxic (216). In a preliminary study in TVXILPS-treated mice, the plasma concentration of TAT dimers, a biomarker of coagulation system activation, peaked at 4.5 h after LPS (data not shown). This corresponds to the onset of liver injury (see Fig. 3.1). In further studies at 4.5 h, treatment with TVX, LVX, LPS or LVX/LPS did not significantly affect the plasma concentration of TAT dimers, whereas TVXILPS-treated mice had a significant elevation (Fig. 6.1A). Circulating fibrinogen is consumed during coagulation system activation. Treatment with either TVX or LVX alone did not affect the plasma fibrinogen concentration (Fig. 6.1 B). Administration of LPS alone caused a small increase in plasma fibrinogen concentration (acute phase response) which was unaffected by pretreatment with LVX. In contrast, TVXILPS-coexposure caused a marked decrease in plasma fibrinogen concentration (Fig. 6.18), suggesting coagulation system activation. The plasma concentration of active PAl-1 was measured as a marker of fibrinolytic system downregulation. No effect on active PAl-1 concentration was seen when mice were treated with TVX or LVX alone (Fig. 6.2). LPS treatment caused a small increase in plasma active PAI-1 concentration, which was 207 Fig. 6.1. Effect of fluoroquinolones and LPS on coagulation system activation. Mice were treated as described in Materials and Methods with TVX, LVX or Veh and then 3 h later with LPS or Veh. They were sacrificed 4.5 h after LPS administration, and plasma concentrations of TAT (A) and fibrinogen (B) were measured. n = 4-6 animals/group.*significantly different from respective control group without LPS treatment. ” significantly different from Veh/LPS group. 208 # T LPS “{‘{“{“““““ 000.00.x...“.....03000 v.0.0>0»0.Q.0.0.0.0.0.Qv0.0.0. O O TTI * # LPS * ’ D ’ _b Veh Veh mTVX -LVX mTVX —va 1:: Veh 1:: Veh 100 0 0 m 0 0 0 30.0.... 00000.5... 3.0... :0. 209 Fig. 6.2. Effect of fluoroquinolones and LPS on plasma concentration of active PAl-1. Mice were treated as described in Materials and Methods with TVX, LVX or Veh and then 3 h later with LPS or Veh. They were sacrificed 4.5 h after LPS administration, and plasma concentration of PAH was measured. n = 4-6 animals/group.*significantly different from respective control group without LPS treatment. " significantly different from Veh/LPS group. 210 active PAl-1 (nglmL) § 55 O S 8 o =Veh mTVX —LVX 9: 9"... V A .9 A 6",- . 0 go 9 O A -.- o o 9’ :0 A O o ’o A. . A .0 A ‘9 Veh 211 markedly enhanced by TVX pretreatment (Fig. 6.2). In contrast, LVX pretreatment did not affect the LPS-induced increase in active PAI-1 (Fig. 6.2). When the rate of fibrin polymerization exceeds the rate of fibrinolysis, fibrin deposition occurs. Administration of TVX or LVX alone did not result in hepatic fibrin deposition (Fig. 6.3). Treatment with LPS alone caused a slight, but significant increase in sinusoidal fibrin when compared to VehNeh-treated mice (Fig. 6.3). LVX pretreatment did not affect the LPS-induced fibrin deposition, but TVXILPS-cotreated mice had significantly increased hepatic sinusoidal fibrin compared to both VehNeh- and LPSNeh-treated mice (Fig. 6.3). 6.4.2 Anticoagulant heparin protects from TVXILPS-induced liver injury To determine if the activation of the coagulation system seen in TVX/LPS- treated mice plays a critical role in the progression of liver injury, mice were treated with anticoagulant heparin. Heparin inhibits coagulation system activation by increasing the affinity of endogenous antithrombin for thrombin and other serine proteases of the coagulation pathway. Extensive fibrin deposition observed in the livers of TVXILPSNeh-treated mice at 4.5 h was significantly reduced by heparin treatment (Fig. 6.4A). Treatment with heparin also reduced the increase in plasma ALT activity in mice given TVX/LPS (Fig. 6.43). This protection was confirmed by histopathological examination. TVX/LPSNeh- treated mice developed large lesions of necrosis with evidence of apoptosis in some, whereas TVX/LPS/heparin-treated mice had less frequent and smaller necrotic lesions (Fig. 6.5, top). 212 Fig. 6.3. Effect of fluoroquinolones and LPS on hepatic sinusoidal fibrin deposition. Mice were treated as described in Materials and Methods with TVX, LVX or Veh and then 3 h later with LPS or Veh. They were sacrificed 4.5 h after LPS administration, and hepatic fibrin deposition was quantified immunohistochemically as described in Materials and Methods. n 4-6 animals/group.*significantly different from respective control group without LPS treatment. # significantly different from Veh/LPS group. 213 .° .5 O i=1Veh mrvx -|_vx 9.0.0 288 Sinusoidal Fibrin Deposition (Fraction Total Area) 9 S *# ':3°8:$’3’3’3‘. v v v v ‘ $333.. ’9 'Q A :0 .0 O O 6: 0. . A '0 Veh -,_¢,,; VehNeh .. o ', ' ‘ h - . 4 2?.» . - . ‘ ’, ° 51-. - .. . I . t . /~ ,fi, ‘ . .. 7 . .- M I ' . I. I y“ o - 3.' s I . ~ . . . . . . .... .’ ‘ b. ... , . .‘ p These data were generated by Aaron Fullerton. 214 Fig. 6.4. Effect of heparin treatment on TVXILPS-induced hepatic fibrin deposition and liver injury. Mice were treated as described in Materials and Methods with TVX/LPS and heparin or saline vehicle. (A) They were sacrificed at 4.5 h, and fibrin deposition was quantified as described in Materials and Methods. n - 4-6 animals/group.*significantly different from respective control group without TVX/LPS treatment. " significantly different from TVX/LPS group not treated with heparin. (B) Mice were sacrificed 15 h after LPS dosing, and plasma ALT activity was measured. n = 8-12 animals/group.*significantly different from TVXILPSNeh-treated mice. 215 Slnusoldal Flbrln Deposltlon (Fractlon Total Area) ALT (UIL) 0.25 l * — Control 020 _ L:I Heparin 0.15 - 0.10 - 0.05 - 0.00 - VehNeh TVXILPS 8000 _ — Control :2: Heparin 6000 — 4000 - * T 2000 « .L o _ TVX/LPS These data were generated by Aaron Fullerton. 216 Fig. 6.5. Histopathology of heparin-treated and PAl-1"' mice treated with TVXILPS. Mice were treated as described in Materials and Methods with TVXILPSNeh or TVXILPS/heparin (top). In addition, wild-type or PAl-1"' mice were treated with TVX/LPS as described in Materials and Methods (bottom). Photomicrographs were taken of representative livers from mice sacrificed at 15 h after LPS administration. 217 .../Hie... u» . ...? ... r . ...siy . ' . t .I czaozamuii ..,\.-.» 218 6.4.3 PAl-1"' mice are protected from TVXILPS-induced liver injury To determine if PAl-1 plays a role in hepatic fibrin deposition at the onset of liver injury, wild-type and PAl-1"' mice were treated with TVX/LPS and killed at 4.5 h. TVX/LPS-coexposurecaused significant fibrin deposition in wild-type mice as well as in PAl-1"' mice (Fig. 6.6A). To confirm that PAl-1"' mice do not have reduced fibrin deposition at a different time, mice were killed at 15 h and hepatic fibrin deposition was quantified. At 15 h, PAl-1"' mice had the same amount of fibrin deposition as wild-type mice (Fig. 6.68) but had significantly reduced hepatocellular injury compared to wild-type mice after TVXILPS-coexposure (Fig. 6.6C). Histopathologic evaluation of liver sections confirmed this protective effect. Midzonal lesions of hepatocellular necrosis and apoptosis observed in TVX/LPS- treated wild-type mice (Fig. 6.5, bottom) were less frequent and smaller in PAl-1"‘ mice (Table 6.1). TVX/LPS coexposure caused grade 4 or 5 (most severe) necrosis in 8/14 wild-type mice, whereas none of the 9 PAl-1"' mice were grade 4 or 5. PAl-1"' mice showed a trend toward decreased inflammation and hemorrhage that did not reach statistical significance. 6.4.4 Role of coagulation system activation and PAl-1 in TVXILPS-induction of cytokines Heparin-treated, wild-type and PAI-1"' mice were cotreated with TVXILPS and killed at 4.5 h to determine what roles heparin and PAl-1 have in the TVXILPS-induction of inflammatory cytokines. Heparin treatment did not change the TVXILPS-induced increases in lL-18, IL-6, KC, lL-10, MCP-1, VEGF, or 219 Fig. 6.6. Effect of PAH deficiency on TVXILPS-induced hepatic fibrin deposition and liver injury. Wild-type and PAl-1"' mice were treated as described in Materials and Methods with TVX and then 3 h later with LPS. They were sacrificed at 4.5 h (A) or 15 (B), and fibrin deposition was quantified as described in Materials and Methods. n = 4-6 animals/group.*significantly different from VehNeh. (C) Mice were sacrificed 15 h after LPS dosing, and plasma ALT activity was measured. n = 13-22 animals/group.*significantly different from wild- type. 220 A *Tll. TVXILPS VehNeh - Wild-type C: PAI-1 "' a o mwmmm nm 0. 0 0 0 .82 32 5.88": 528%.. $5... 328:...» - Wild-type =1 PAl-l '4' m m m. m. 0 o o o :25 .23 5:08“: 8:83.. :3: 3282.5 w. o TVXILPS VehNeh — Wild-type i=i PAl-1"' 4000 l C 3000 « 00 3:: S< TVXILPS 221 Table 6.1. Scoring of histopathology of livers from wild-type and PAI-1"' mice cotreated with TVXILPS. Mouse strain Necrosis Inflammation Hemorrhage , Wild-type 3.2 i 0.4 1.4 d: 0.1 0.6 :l: 0.2 PAl-1"' 2.0 :l: 0.2 * 1.0 1: o o 1 o Wild-type and PAI-1"' mice were treated with TVX and LPS as described in Materials and Methods. They were sacrificed 15 h after LPS. Liver sections were cut 5 mm thick and stained with hematoxylin and eosin, and resultant slides were scored by a pathologist for necrosis, inflammation and hemorrhage. The scoring scale was set from 0-5 with the following criteria: no observation (0), mild (1), mild to moderate (2), moderate (3), moderate to severe (4), and severe (5). Scores are reported as average :I: S.E.M. n = 9-15 animals/group.*significantly different from wild-type group. 222 TNFa. However, it enhanced the TVXILPS-induction of lL-1B and lFNy (Fig. 6.7). In contrast, PAl-1"' mice had significantly reduced levels of several cytokines after TVX/LPS treatment. These included lL-1B, lL-6, KC, lL-10, MCP-1, and IFNy (Fig. 6.7). There wasa trend toward decreased plasma concentration of TNFa in PAl-1"' mice, but this was not statistically significant. Of the cytokines measured, only lL-18 and VEGF were increased to the same degree in wild-type and PAl-14' mice by TVXILPS-coexposure (Fig. 6.7). 223 Fig. 6.7. Effect of heparin treatment or PAl-1 deficiency on TVXILPS- induced increases in plasma cytokines. Mice were treated with vehicles or with TVXILPS and sacrificed 4.5 h after LPS administration. Plasma concentrations of lL-1B, lL-18, lL-6, KC, lL-10, MCP-1, VEGF, lFNy, and TNFa were measured as described in Materials and Methods. n = 4-6 animals/group.*significantly different from VehNeh. # significantly different from TVX/LPS control group. 224 uW////////////% 1111111 .. Theme 3.... TVX/LPS Veh .W//////////%% *M/%///////% TVX/LPS Veh Eff“: PAi-1+ 225 ’////A Heparin - Control Fig. 6.7 (cont’d). lL-18 (pglm L) Veh TVX/LPS ‘0 . 3 3° ‘ E 2’ 2. ‘9 i 10 < o . TVX/LPS 4000 1 3 3000 . E U) 3. 20004 2 =i' 1000 o . TVX/LPS - Control ‘ ff PAM" m Heparin 226 Fig. 6.7 (6°"t'd)' TVX/LPS Veh * * .I o o o m m m m TVX/LPS eh # * an * * v mumnmuo 4| ... ... i .3 ... W . AJctmavdn—ZF _ TVX/LPS Veh - C°“"°' f PAI-l" W/IA Heparin 227 6.5 Discussion Previous studies showed that TVX, but not LVX, interacts with a nontoxic dose of LPS to cause hepatotoxicity in mice (216). The studies presented here examined whether TVXILPS-coexposure altered the hemostatic system and, if so, what role the alteration played in the pathogenesis. A detailed timecourse of TVXILPS-induced liver injury showed that liver injury begins at ~4.5 h and peaks at 15 h after LPS administration (see Fig. 3.1). To understand the importance of the hemostatic system in the progression of liver injury, hemostatic system biomarkers were measured at the onset of liver injury. The hemostatic system normally functions as a balance between prothrombotic and antithrombotic factors to maintain homeostasis. Based on an increase in thrombin activation and a decrease in plasma fibrinogen concentration, TVXILPS-coexposure activated the coagulation system, but TVX, LVX, LPS or LVX/LPS treatment did not (Fig. 6.1). LPS treatment alone induced a small, but statistically significant increase in plasma fibrinogen. This response has been seen previouSly and likely reflects an acute phase response to LPS (267). Coagulation system activation accompanying liver injury occurs after cotreatment with LPS and other agents (268-270). PAl-1 inhibits fibrinolysis via inhibition of plasminogen activators (271). Like its effect on coagulation system activation, TVX, but not LVX, enhanced the LPS-induced increase in plasma concentration of active PAl-1 (Fig. 6.2). Whether TVX acts directly to enhance the LPS induction of coagulation factors and PAl-1 is unknown. It is possible that TVX directly upregulates tissue factor or acts indirectly by enhancing the LPS 228 induction of cytokines such as TNFa that can induce tissue factor production (272). TVX enhances the LPS-induced production of TNFa, and it is likely that this plays a role in the TVXILPS-induced coagulation system activation (216). TNFa is required for TVXILPS-induced liver injury, and the increased TNFa in TVXILPS-treated mice affected coagulation system activation and PAI-1 production (Chapter 4). The TVXILPS-mediated coagulation system activation and impairment of the fibrinolytic system suggests an imbalance in the hemostatic system that favors fibrin formation. Indeed, TVXILPS-coexposure did result in increased in fibrin deposition compared to all other treatment groups (Fig. 6.3). Sinusoidal fibrin can impair hepatic blood flow, and thus oxygen delivery to hepatocytes, causing tissue hypoxia (103). Hypoxia could play a role in the pathogenesis of TVXILPS-induced liver injury in a number of ways. For example, it sensitizes hepatocytes to cytotoxicity by proteases and hypochlorous acid (HOCl) released by activated neutrophils (103). In addition, hypoxia can alter cellular homeostasis by inducing oxidative stress and can directly cause cell death (273-275). To address whether thrombin activation was involved in the pathogenesis of liver injury, heparin was used to inhibit coagulation. Heparin inhibited thrombin activation and decreased the TVXILPS-induced fibrin deposition which in turn protected mice from TVXILPS-induced liver injury (Fig. 6.4). This suggests that coagulation system activation plays a critical role in the fibrin deposition and the pathogenesis. However, whether hepatic fibrin deposition, thrombin activation of receptors or both are critical for TVXILPS-induced liver injury is unknown. 229 Heparin can attenuate inflammation (153); accordingly, to address if this might be involved in its protective effect, plasma cytokine concentrations were measured. Heparin treatment failed to reduce plasma concentrations of any of the cytokines induced by TVXILPS-treatment (Fig. 6.7), suggesting that heparin did not provide protection against liver injury by reducing inflammatory cytokines. This result also suggests that coagulation system activation acts downstream of any effects initiated by cytokines. Heparin treatment significantly increased TVXILPS-induction of IFNy. This enhancement might have been a result of the inhibition of thrombin, which can drive a Th2 response that downregulates lFNy expression (142). In addition, after heparin treatment there was a slight, but not statistically significant decrease in the plasma concentration of VEGF, the expression of which can be driven by hypoxia (276). In other models, there is evidence that PAl-1 can play a role in both fibrin deposition and the progression of liver injury (277-279). However, hepatic fibrin deposition at both 4.5 and 15 h was similar in wild-type and PAl-1"' mice treated with TVX/LPS, yet both plasma ALT activity and histopathologic evaluation indicated lesser hepatocellular injury in the PAl-1"' mice (Fig. 6.5 and 6.6). Therefore, the results suggest that protection from TVXILPS-induced liver injury in PAl-1"‘ mice is not related to fibrin deposition. In addition to its role in fibrinolysis, PAl-1 has proinflammatory properties including induction of lL-6 (147). Moreover, PAl-1 can enhance LPS-induced neutrophil activation (146). Relative to wild-type mice, PAI-1"' mice treated with TVX/LPS had reduced concentrations of lL-1B, IL-6, lL-10, lFNy, MCP-1 and KC 230 at 4.5 h (Fig. 6.7). This result suggests a proinflammatory role for PAI-1 in the regulation of several cytokines. Our result for IFNy is in contrast to a report that PAl-1"' mice have an enhanced plasma lFNy response to a toxic dose of LPS (280). It may be that the induction of IFNy occurs by a different mechanism after TVXILPS-coexposure compared to administration of a toxic dose of LPS by itself. It is also possible that the attenuated cytokine response in PAl-1"' mice is secondary to a reduction in injury; however plasma concentrations were measured at 4.5 h, when there is only a slight increase in plasma ALT activity which is not attenuated in PAI-1"' mice (data not shown). Therefore, it seems likely that the reduced cytokine response underlies the protection seen in PAl-1"’ mice. However, it has been reported that PAl-1"' endothelial cells are resistant to apoptosis (281). If hepatocytes are also resistant to apoptosis in these mice, this might play a role that attenuated liver injury in PAl-1"' mice. In summary, TVX interacted with LPS to enhance coagulation system activation, increase PAl-1 production and cause liver injury. In contrast, LVX did not alter the hemostatic system or cause liver injury when coadministered with LPS. The TVXILPS-induced imbalance in the hemostatic system resulted in hepatic fibrin deposition. Anticoagulant heparin inhibited thrombin activation which significantly reduced fibrin deposition and protected mice from TVX/LPS- induced hepatocellular necrosis without decreasing plasma concentrations of proinflammatory cytokines. This suggests that active thrombin, perhaps through enhanced fibrin deposition, contributes to the liver injury. PAl-1"‘ mice did not have attenuated hepatic fibrin deposition but did have smaller plasma 231 concentrations of several inflammatory cytokines and less TVXILPS-induced liver injury. Accordingly, coagulation system activation was critical for TVXILPS- induced fibrin deposition, whereas PAl-1 was not, and the important role of PAl-1 in promoting the liver injury'appears to be unrelated to its ability to downregulate fibrinolysis. It is likely that thrombin activation and proinflammatory cytokines comprise two independent pathways that contribute to TVXILPS-induced liver injury, inasmuch as the removal of either one attenuated, but did not eliminate liver injury; in order to achieve complete protection from TVXILPS-induced liver injury the inhibition of both was required. 232 CHAPTER 7 Vascular endothelial growth factor has a proinflammatory role which is critical to trovafloxacin and lipopolysaccharide coexposure-induced liver injury. 233 7.1 Abstract TVX is a fluoroquinolone antibiotic which has caused idiosyncratic hepatotoxicity in a small fraction of people. Animal models predictive of drugs that cause lADRs are lacking in preclinical safety testing. Previously, we showed that a modest inflammatory stress induced by LPS renders mice sensitive to nonhepatotoxic doses of TVX. In contrast, LVX, a fluoroquinolone antibiotic that has not been associated with lADRs in humans, does not cause hepatotoxicity in mice cotreated with LPS. TVXILPS-induced liver injury is dependent on both coagulation and TNFa pathways. VEGF is a cytokine with proinflammatory properties which has the potential to be a part of either of the pathways involved in the pathogenesis. The purpose of this study was to explore interactions between VEGF and other proinflammatory mediators of liver injury and, in turn, the role of VEGF in TVXILPS-induced liver injury. TVX prolonged the LPS- induced increase in plasma VEGF. VEGF neutralization attenuated the TVXILPS-induced increases in both hepatic fibrin deposition and plasma TNFa concentration. Additionally, VEGF neutralization protected mice from TVX/LPS- induced liver injury, as reflected by both plasma ALT activity and liver histopathology. In summary, VEGF has proinflammatory properties and plays a critical role in the progression of TVXILPS-induced liver injury. 234 7.2 Introduction VEGF has been studied extensively for its angiogenic properties. However, recent work has focused on its proinflammatory properties. Several cytokines can affect VEGF expression; conversely, VEGF can induce TNFa, neutrophil chemokines and tissue factor (92, 179, 180). Inasmuch as VEGF has the potential to interact with several pathways involved in TVXILPS-induced liver injury, its interactions with various mediators of liver injury were examined. In accordance with its proinflammatory effects, VEGF is involved in the development of liver injury in animal models of endotoxemia and ischemialreperfusion (179, 180). The purpose of this study was to test the hypothesis that VEGF is involved in the pathogenesis of TVXILPS-induced liver injury. Furthermore, the potential interaction of VEGF with neutrophil accumulation, TNFa, IFNy and hepatic fibrin deposition, all of which play a role in TVXILPS-induced liver injury, was explored in this study. These pathways involved in the pathogenesis were explored at a time near the onset of liver injury. Additionally, the study explored novel proinflammatory properties of VEGF that might be of importance for inflammatory tissue injury. 235 7.3 Materials and Methods 7.3.1 Materials Please refer for Section 2.3.1 for information on this topic. VEGF antiserum and control rabbit serum were a kind gift from Dr. David Briscoe (Harvard Medical School, Boston, MA). 7.3.2 Animals Please refer to Section 2.3.2 for information on this topic. 7.3.3 Experimental protocols Mice fasted for 12 h were given TVX (150 mglkg) or Veh (saline) by oral gavage. They were then given LPS (2.0 x 106 EU/kg) or Veh (saline) by intraperitoneal injection 3 h later. Food was returned immediately thereafter. Mice were anesthetized with sodium pentobarbital (50 mglkg; i.p.) and killed at designated times after the administration of LPS or Veh for various , measurements. Blood was drawn from the vena cava into a syringe containing sodium citrate resulting in a final concentration of 0.76%. The left lateral lobe was fixed in 10% neutral buffered formalin and paraffin blocked. For VEGF neutralization studies, VEGF antiserum or control serum was administered (0.5 mL, i.p.) 15 h before and (0.8 mL, i.p.) 2 h after LPS. 236 7.3.4 Histopathology Please refer to section 2.3.4 for information on this topic. 7.3.5 Cytokine measurements Please refer to Section 3.3.5 for information on this topic. 7.3.6 Neutrophil staining Please refer to section 3.3.6 for information on this topic. 7.3.7 Hemostatic system measurements Please refer to Section 4.3.7 for information on this topic. 7.3.8 Statistical analyses Results are presented as mean i S.E.M. A Student’s t-test or a 2-way analysis of variance (ANOVA) was used as appropriate after data normalization. All pairwise comparisons were made using Tukey’s test with the criterion for significance at p < 0.05. 237 7.4 Results 7.4.1 Timecourse of plasma concentration of VEGF TVX alone did not have any effect on plasma VEGF concentration (Fig. 7.1). LPS treatment caused a significant increase in plasma VEGF at all times measured (Fig. 7.1). TVX treatment prior to LPS enhanced the LPS-induced increase in plasma concentration of VEGF at 4.5 (Fig. 7.1). 7.4.2 Effect of VEGF neutralization on TVXILPS-induced hepatotoxicity To explore the role of VEGF in TVXILPS-induced liver injury, VEGF was neutralized by VEGF antiserum, which reduced the TVXILPS-induced increase in plasma VEGF at 4.5 h (Fig. 7.2A). TVXILPS-treated mice were treated with control serum or VEGF antiserum and then killed at 15 h. TVXILPS-induced liver injury was attenuated by VEGF neutralization (Fig. 7.23). Histopathologic evaluation confirmed that mice were protected from TVXILPS-induced hepatotoxicity by VEGF neutralization. TVX/LPS coexposure caused hepatocellular necrosis and apoptosis primarily localized to the midzonal regions (Fig. 7.3). VEGF neutralization reduced the size and frequency of lesions induced by TVX/LPS coexposure (Fig. 7.3). 238 Fig. 7.1 Timecourse of VEGF plasma concentration. Mice were treated with TVX or Veh 3h before LPS or Veh as described in Materials and Methods. They were killed at various times and plasma concentration of VEGF was measured. n = 5—7 animals/group. *signiflcantly different from 0 h within same treatment group. 1"significantly different from Veh/LPS treatment group at same time. 239 VEGF (pglmL) 1200 ' - -0 - VehNeh - «3 - TVXNeh Hours after LPS or Veh 240 Fig. 7.2. Effect of VEGF neutralization on TVXILPS-induced plasma VEGF induction and liver injury. Mice were treated with TVXILPS or VehNeh in addition to VEGF antiserum or control serum as described in Materials and Methods. (A) They were killed 4.5 h after LPS administration. Plasma concentration of VEGF was measured as described in Materials and Methods. n = 4-6 animals/group. (B) Mice were killed 15 h after LPS administration and plasma ALT activity was measured. n = 6-10 animals/group. *significantly different from respective VehNeh group. 1”significantly different from TVXILPS/control serum-treated group. 241 1000 1 800 4 VEGF (ngmL) 8 § _ Control 12:: VEGF antiserum 8 8 ALT activity (UIL) O ”5 s 3 .L O 8 VehNeh TVXILPS - CODUOI * D VEGF antiserum VehNeh TVXILPS 242 Fig. 7.3. Protection from TVXILPS-induced liver pathology by VEGF neutralization. Mice were treated with TVX/LPS or VehNeh in addition to VEGF antiserum or control serum as described in Materials and Methods. They were killed 15 h after LPS administration, and photomicrographs were taken of representative livers. 243 \ U 5027'5.‘ ..m‘z; - 1.;- ”.49. 4 . 5" .- ' “a?“ 7*? . ...- ' bkiyfvfffizxfi 3’37" 4! ~ . 1.9. . . '\ ‘ '. 5 Q l _ ‘ Pf); ‘5 ‘33?“ ‘ “C‘ :35... :93 _mn_.__/ /xH\ activation did not increase plasma concentrations of any of the cytokines examined (Fig. 6.7). Thrombin activation was critical to the progression of TVXILPS-induced liver injury (Fig. 6.3), likely at least in part, via hepatic fibrin deposition. TVX enhanced the LPS-induced increase in the plasma concentration of TNFa, which appears to be a central player in TVXILPS-induced liver injury. TVXILPS-induced increases in the plasma concentrations of VEGF (Fig. 4.2), chemokines (Fig. 4.3), PAl—1 (Fig. 4.5), lL-18 (data not shown) and lFNy (Fig. 4.2) were TNFa-dependent. Due to their TNFa-dependence and because TNFa was enhanced before these factors, they are likely downstream of TNFa. Furthermore, TNFa and lFNy could have directly caused hepatocellular death which might be enhanced by TVX. Likely proinflammatory cycles were identified, inasmuch as several factors were found which upregulate one another. The removal or inhibition of VEGF (Fig. 7.4), PAI-1 (Fig. 6.7) or IFNy (Fig. 5.8) attenuated the TVXILPS-induced increase in plasma concentration of TNFa. Conversely, TNFor neutralization reduced the appearance of each of these. Similarly, lFNy"' mice had a reduced plasma concentration of lL-18 (Fig. 5.8) following TVXILPS coexposure, and IL- 18 is a known inducer of IFNy expression. PAl-1 also had other proinflammatory properties beyond upregulating TNFa, such as increasing plasma concentrations of chemokines and lFNy (Fig. 6.7) after TVX/LPS administration. These vicious proinflammatory cycles are summarized in Fig. 8.2. Several of these properties 269 found were novel and the inflammatory cycles of upregulation have the potential to be involved in the pathogenesis. 270 Fig. 8.2. Possible proinflammatory cycles induced by TVXILPS coexposure. Based on the results obtained, there exist several possible loops of uncontrolled upregulation. These vicious cycles of inflammation could be involved in TVXILPS-induced liver injury. See Section 8.2 for additional information. 271 \ > lL-18 'IFNy TNFa 11 r PAl-1 '— J VEGF 272 Section 8.3 Major findings and implications 1. TVX, but not LVX interacts with an inflammatory stress to cause liver injury in mice. This observation had been reported in rats (58). However, the development of hepatotoxic TVX-inflammation interaction in mice demonstrates that the phenomenon is not species-specific and might be extrapolated to TVX lADRs in humans. Furthermore, the degree of TVXILPS-induced liver injury was much greater in mice (Fig. 2.1) compared to rats (58). Both moderate and severe hepatotoxic responses have been reported in people who took TVX (206). The robustness of the murine model of liver injury resembles the severe hepatotoxicity caused by TVX in humans more so than the rat model. Additionally, that TVX, but not LVX interacts with an inflammatory stress to cause hepatotoxicity suggests that this model could distinguish between fluroquinolones based on their propensity to cause lADRs. This suggests that the drug/inflammation animal model could potentially be used as a preclinical tool to help select a drug candidate based on its potential lADR liability. 2. TVX interacted with an inflammatory stress induced by either gram- negative or gram-positive stimuli. That TVX interacted with either TLR2- or TLR4-activating ligands to cause liver injury proves that TVX/inflammation-induced liver injury is not specific to TLR4 activation. Indeed, it suggests that TVX interacts with an inflammatory stress, 273 regardless of its source, to precipitate liver injury. The result suggests that inflammatory stress induced by either gram-positive or gram-negative bacteria might play a role in TVX hepatotoxicity. This is of particular importance inasmuch as TVX was approved for treating both gram— positive and gram-negative bacterial infections by the FDA. . TVXILPS-coexposure resulted in unique gene expression changes in mice prior to the onset of liver injury. These results suggest that global gene expression change is an earlier marker of liver toxicity than plasma ALT activity in this model. It is possible that gene expression analysis could be used to identify drugs with lADR liability when coadministered with LPS, even if the coexposure does not result in hepatotoxicity. It is possible that in the drugflnflammation model gene expression analysis offers a more sensitive endpoint to filter out than liver injury to identify drug candidates with lADR liability. . TVXILPS-induced liver injury was dependent on PMN activation, TNFa, IFNy, thrombin activation, PAH and VEGF. It is possible that these mediators of inflammatory liver injury are involved in the pathogenesis of TVX-induced hepatotoxicity in people. 274 5. TVXILPS-induced liver injury is dependent on both TNF receptors. The p55 receptor has been well studied, but the role of the p75 receptor in inflammatory liver injury is unclear. The p75 receptor is not involved in some models of liver injury that are dependent on TNFa (96, 240) but is involved in others (234-236). Indeed, p754' mice were protected to a greater degree than p55”' mice from TVXILPS-induced liver injury. Therefore, TVX/LPS coexposure would be an ideal model to use for further studies examining the role of the p75 receptor in drug-induced liver injury. 6. The comprehensive studies exploring the roles of TNFa, IFNy, PAH and VEGF following TVX/LPS coexposure led to the finding that several proinflammatory cycles appear to be involved in the pathogenesis. It is possible that they result in an unregulated inflammatory response resulting in host tissue injury. These proinflammatory cycles could be involved in numerous models of inflammatory tissue injury and might not be specific to the liver or to TVX/LPS coexposure. 275 Section 8.4 Knowledge gaps and future studies Several important findings were identified by this research including the development of an animal model of TVX-induced hepatotoxicity. It is possible that the drug/inflammation model could be used preclinically to identify drugs with the propensity to cause lADRs. Further studies with additional fluoroquinolones would provide more positive and negative comparators and would add merit to the model. Additionally, extensive studies with other drugs linked with lADRs in people would be needed to validate the model. It is essential to determine the rate of false negatives and false positives identified by the model before any widescale use in drug development. The findings presented elucidated some of the mechanisms involved in TVXILPS-induced liver injury. However, the mechanism by which TVX interacts with TLR2 and TLR4 activation to cause liver injury is still unknown. Future studies will examine signaling pathways initiated by TLR activation to determine if TVX pretreatment alters the initial signaling pathways. The identification of specific pathways might lead to the finding of a specific enzyme or adapter protein which TVX interacts with. Whether TVX affects hepatocytes directly was not examined. It is possible that TVX directly sensitizes to insults such as PMN proteases, hypoxia, TNFa or IFNy. A direct sensitization of hepatocytes by TVX might account for the extensive liver injury caused by TVX/LPS coexposure. 276 TVX treatment prior to a nonhepatotoxic dose of recombinant murine TNFa resulted in significant liver injury. Other studies have shown that TNFa by itself does not cause liver injury in mice but can when administered with galactosamine or a DNA synthesis inhibitor (249, 250). Indeed, the hepatocellular lesions in TVXfI'NFa-treated mice appear similar to those seen after galactosamine/TNFa coexposure, suggesting commonalities in mechanisms (251). This raises the possibility that TVX affects DNA synthesis of eukaryotic cells, rendering them sensitive to inflammatory stress. Additional studies need to be done to determine if TVX interacts with eukaryotic cells to affect DNA synthesis to a greater degree than other fluoroquinolones. This could provide understanding of the mechanism by which TVX causes liver injury in people, whereas other fluoroquinolones do not. Another focus which requires further research is examination of specific cell types. Neutrophil activation was not measured. Furthermore, it is unknown whether any of the cytokines involved in the pathogenesis affect PMN activation. It is possible that they cause liver injury, at least in part, by enhancing PMN activation. Additionally, it is unknown whether cell types other than PMNs are involved in TVXILPS-induced hepatotoxicity. It is possible that T cells, NK cells, platelets and Kupffer cells are involved in the pathogenesis, and requires further examination. 277 10. 11. 12. 13. Reference List . Pirrnohamed M, James S, Meakin S, Green C, Scott AK, Walley TJ, et al. Adverse drug reactions as cause of admission to hospital: prospective analysis of 18 820 patients. BMJ 2004 Jul 3;329(7456):15-19. Lasser KE, Allen PD, Woolhandler SJ, Himmelstein DU, Wolfe SM, Bor DH. Timing of new black box warnings and withdrawals for prescription medications. JAMA 2002 May 1;287(17):2215-2220. Michelson S, Joho K. Drug discovery, drug development and the emerging world of pharmacogenomics: prospecting for information in a data-rich landscape. Curr Opin Mol Ther 2000 Dec;2(6):651-654. Sibbald B. Cisapride, before and after: still waiting for ADE-reporting reform. CMAJ 2001 Nov 13;165(10):1370. Kaitin Kl. Drug Safety Withdrawals in the US. not linked to speed of FDA approval. Tufts Center for the Study of Drug Development Impact Report 2005 2005 Sep;7(5). Ostapowicz G, Fontana RJ, Schiodt FV, Larson A, Davern TJ, Han SH, et al. Results of a prospective study of acute liver failure at 17 tertiary care centers in the United States. Ann Intern Med 2002 Dec 17;137(12):947- 954. Hunter EB, Johnston PE, Tanner G, Pinson CW, Awad JA. Bromfenac (Duract)-associated hepatic failure requiring liver transplantation. Am J Gastroenterol 1999 Aug;94(8):2299-2301. Graham DJ, Green L, Senior JR, Nourjah P. Troglitazone-induced liver failure: a case study. Am J Med 2003 Mar;114(4):299-306. Murphy EJ, Davern TJ, Shakil AO, Shick L, Masharani U, Chow H, et al. Troglitazone-induced fulminant hepatic failure. Acute Liver Failure Study Group. Dig Dis Sci 2000 Mar;45(3):549-553. Bemuau J, Mallet L, Benhamou JP. [Hepatotoxicity due to tienilic acid]. Gastroenterol Clin Biol 1981 Jun;5(6-7):692-693. Lazarczyk DA, Goldstein NS, Gordon SC. Trovafloxacin hepatotoxicity. Dig Dis Sci 2001 Apr;46(4):925-926. Nicholson SC, Webb CD, Moellering RC, Jr. Antimicrobial-associated acute hepatitis. Pharrnacotherapy 2002 Jun;22(6):794-796. Choi S. Nefazodone (Serzone) withdrawn because of hepatotoxicity. CMAJ 2003 Nov 25;169(11):1187. 278 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. Maniar JK, Shah SR, Verma R, Kamath R, Gupte P, Maniar A. Nevirapine—induced fulminant hepatitis. J Assoc Physicians India 2006 Dec;54:957-958. Sanne I, Mommeja-Marin H, Hinkle J, Bartlett JA, Lederman MM, Maartens G, et al. Severe hepatotoxicity associated with nevirapine use in HIV-infected subjects. Jlnfect Dis 2005 Mar 15;191(6):825-829. Temple RJ, Himmel MH. Safety of newly approved drugs: implications for prescribing. JAMA 2002 May 1;287(17):2273-2275. Hussaini SH, Farrington EA. Idiosyncratic drug-induced liver injury: an overview. Expert Opin Drug Saf 2007 Nov;6(6):673-684. Lee WM. Drug-induced hepatotoxicity. N Engl J Med 2003 Jul 31 ;349(5):474-485. Kaplowitz N, Aw TY, Simon FR, Stolz A. Drug-induced hepatotoxicity. Ann Intern Med 1986 Jun;104(6):826-839. Walgren JL, Mitchell MD, Thompson DC. Role of metabolism in drug- induced idiosyncratic hepatotoxicity. Crit Rev Toxicol 2005 Apr;35(4):325- 361. Kassahun K, Pearson PG, Tang W, McIntosh I, Leung K, Elmore C, et al. Studies on the metabolism of troglitazone to reactive intermediates in vitro and in vivo. Evidence for novel biotransforrnation pathways involving quinone methide formation and thiazolidinedione ring scission. Chem Res Toxicol 2001 Jan;14(1):62-70. He K, Talaat RE, Pool WF, Reily MD, Reed JE, Bridges AJ, et al. Metabolic activation of troglitazone: identification of a reactive metabolite and mechanisms involved. Drug Metab Dispos 2004 Jun;32(6):639—646. Uetrecht J. Evaluation of which reactive metabolite, if any, is responsible for a specific idiosyncratic reaction. Drug Metab Rev 2006;38(4):745-753. Williams DP, Park BK. Idiosyncratic toxicity: the role of toxicophores and bioactivation. Drug Discov Today 2003 Nov 15;8(22):1044-1050. Mitchell JR, Zimmerman HJ, lshak KG, Thorgeirsson UP, Timbrell JA, Snodgrass WR, et al. Isoniazid liver injury: clinical spectrum, pathology, and probable pathogenesis. Ann Intern Med 1976 Feb;84(2):181-192. Huang YS. Genetic polymorphisms of drug-metabolizing enzymes and the susceptibility to antituberculosis drug-induced liver injury. Expert Opin Drug Metab Toxicol 2007 Feb;3(1):1-8. 279 27. 28. 29. 30. 31. 32. 33. 35. 36. 37. 38. Gurumurthy P, Krishnamurthy MS, Nazareth O, Parthasarathy R, Sarrna GR, Somasundaram PR, et al. Lack of relationship between hepatic toxicity and acetylator phenotype in three thousand South Indian patients during treatment with isoniazid for tuberculosis. Am Rev Respir Dis 1984 Jan;129(1):58-61. Aithal GP, Ramsay L, Daly AK, Sonchit N, Leathart JB, Alexander G, et al. Hepatic adducts, circulating antibodies, and cytokine polymorphisms in patients with diclofenac hepatotoxicity. Hepatology 2004 May;39(5):1430- 1440. Uetrecht J. Screening for the potential of a drug candidate to cause idiosyncratic drug reactions. Drug Discov Today 2003 Sep 15;8(18):832- 837. Park BK, Coleman JW, Kitteringham NR. Drug disposition and drug hypersensitivity. Biochem Pharrnacol 1987 Mar 1;36(5):581-590. Robin MA, Le RM, Descatoire V, Pessayre D. Plasma membrane cytochromes P450 as neoantigens and autoimmune targets in drug- induced hepatitis. J Hepatol 1997;26 Suppl 1:23-30. Obermayer-Straub P, Strassburg CP, Manns MP. Autoimmune hepatitis. J Hepatol 2000;32(1 Suppl):181-197. Kitteringham NR, Kenna JG, Park BK. Detection of autoantibodies directed against human hepatic endoplasmic reticulum in sera from patients with halothane-associated hepatitis. Br J Clin Phamnacol 1995 Oct;40(4):379-386. Park BK, Pirmohamed M, Kitteringham NR. Role of drug disposition in drug hypersensitivity: a chemical, molecular, and clinical perspective. Chem Res Toxicol 1998 Sep;11(9):969—988. Liu ZX, Kaplowitz N. Immune-mediated drug-induced liver disease. Clin Liver Dis 2002 Aug;6(3):755-774. Kaplowitz N. Idiosyncratic drug hepatotoxicity. Nat Rev Drug Discov 2005 Jun;4(6):489-499. Uetrecht JP. New concepts in immunology relevant to idiosyncratic drug reactions: the "danger hypothesis" and innate immune system. Chem Res Toxicol 1999 May; 1 2(5):387-395. Seguin B, Uetrecht J. The danger hypothesis applied to idiosyncratic drug reactions. Curr Opin Allergy Clin Immunol 2003 Aug;3(4):235-242. 280 39. 40. 4 A 42. 43. 44. 45. 46. 47. 48. 49. 50. Pichler WJ. Pharmacological interaction of drugs with antigen-specific immune receptors: the p-i concept. Curr Opin Allergy Clin Immunol 2002 Aug;2(4):301-305. Engler OB, Strasser l, Naisbitt DJ, Cerny A, Pichler WJ. A chemically inert drug can stimulate T cells in vitro by their T cell receptor in non-sensitised individuals. Toxicology 2004 Apr 1;197(1):47-56. . Chinnery PF, Johnson MA, Wardell TM, Singh-Kler R, Hayes C, Brown DT, et al. The epidemiology of pathogenic mitochondrial DNA mutations. Ann Neurol 2000 Aug;48(2):188—193. Waring JF, Anderson MG. Idiosyncratic toxicity: mechanistic insights gained from analysis of prior compounds. Curr Opin Drug Discov Devel 2005 Jan;8(1):59-65. Boelsterli UA, Lim PL. Mitochondrial abnormalities-a link to idiosyncratic drug hepatotoxicity? Toxicol Appl Pharmacol 2007 Apr 1;220(1):92-107. Haasio K, Koponen A, Penttila KE, Nissinen E. Effects of entacapone and tolcapone on mitochondrial membrane potential. Eur J Pharmacol 2002 Oct 18;453(1):21-26. Keller BJ, Yamanaka H, Thurman RG. Inhibition of mitochondrial respiration and oxygen-dependent hepatotoxicity by six structurally dissimilar peroxisomal proliferating agents. Toxicology 1992;71(1-2):49—61. Petrescu l, Tarba C. Uncoupling effects of diclofenac and aspirin in the perfused liver and isolated hepatic mitochondria of rat. Biochim Biophys Acta 1997 Feb 15;1318(3):385-394. Schwab CE, Tuschl H. In vitro studies on the toxicity of isoniazid in different cell lines. Hum Exp Toxicol 2003 Nov;22(11):607-615. Shishido S, Koga H, Harada M, Kumemura H, Hanada S, Taniguchi E, et al. Hydrogen peroxide overproduction in megamitochondria of troglitazone-treated human hepatocytes. Hepatology 2003 Jan;37(1):136- 147. Boelsterli UA. Diclofenac-induced liver injury: a paradigm of idiosyncratic drug toxicity. Toxicol Appl Pharmacol 2003 Nov 1;192(3):307-322. Tirmenstein MA, Hu CX, Gales TL, Maleeff BE, Narayanan PK, Kurali E, et al. Effects of troglitazone on HepGZ viability and mitochondrial function. Toxicol Sci 2002 Sep;69(1):131-138. 281 51. 52. 53. 54. 55. 56. 57 58. 59. 60. 61. Ong MM, Latchoumycandane C, Boelsterli UA. Troglitazone-induced hepatic necrosis in an animal model of silent genetic mitochondrial abnormalities. Toxicol Sci 2007 May;97(1):205-213. Watkins PB. Idiosyncratic liver injury: challenges and approaches. Toxicol Pathol 2005;33(1):1-5. Black M, Mitchell JR, Zimmerman HJ, lshak KG, Epler GR. Isoniazid- associated hepatitis in 114 patients. Gastroenterology 1975 Aug;69(2):289-302. Li AP. 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 2002 Nov 10;142(1-2):7-23. Roth RA, Luyendyk JP, Maddox JF, Ganey PE. Inflammation and drug idiosyncrasyuis there a connection? J Pharmacol Exp Ther 2003 Oct;307(1):1-8. Buchweitz JP, Ganey PE, Bursian SJ, Roth RA. Underlying endotoxemia augments toxic responses to chlorpromazine: is there a relationship to drug idiosyncrasy? J Pharmacol Exp Ther 2002 Feb;300(2):460-467. . Deng X, Stachlewitz RF, Liguori MJ, Blomme EA, Waring JF, Luyendyk JP, et al. Modest inflammation enhances diclofenac hepatotoxicity in rats: role of neutrophils and bacterial translocation. J Pharmacol Exp Ther 2006 Dec;319(3):1191-1199. Waring JF, Liguori MJ, Luyendyk JP, Maddox JF, Ganey PE, Stachlewitz RF, et al. Microarray analysis of lipopolysaccharide potentiation of trovafloxacin-induced liver injury in rats suggests a role for proinflammatory chemokines and neutrophils. J Pharmacol Exp Ther 2006 Mar;316(3):1080-1087. Luyendyk JP, Maddox JF, Cosma GN, Ganey PE, Cockerell GL, Roth RA. Ranitidine treatment during a modest inflammatory response precipitates idiosyncrasy-like liver injury in rats. J Pharmacol Exp Ther 2003 Oct;307(1):9-16. Zou W, Devi SS, Younis HS, Roth RA, Ganey PE. Inflammation-mediated Liver Injury in Rats Cotreated with Sulindac and Lipopolysaccharide [Abstract]. The Toxicologist 2008;102. Hellman J, Roberts JD, Jr., Tehan MM, Allaire JE, Warren HS. Bacterial peptidoglycan-associated lipoprotein is released into the bloodstream in gram-negative sepsis and causes inflammation and death in mice. J Biol Chem 2002 Apr 19;277(16):14274-14280. 282 62. 63 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. Zahringer U, Lindner B, Rietschel ET. Molecular structure of lipid A, the endotoxic center of bacterial lipopolysaccharides. Adv Carbohydr Chem Biochem 1994;50:21 1-276. . Janssens S, Beyaert R. Role of Toll-like receptors in pathogen recognition. Clin Microbiol Rev 2003 Oct;16(4):637-646. Jerala R. Structural biology of the LPS recognition. Int J Med Microbiol 2007 Sep;297(5):353-363. Kitchens RL, Thompson PA. Modulatory effects of sCD14 and LBP on LPS-host cell interactions. J Endotoxin Res 2005;11(4):225-229. Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol 2004 Ju|;4(7):499-511. Cao Z, Henzel WJ, Gao X. IRAK: a kinase associated with the interleukin- 1 receptor. Science 1996 Feb 23;271(5252):1 128-1131. Cao Z, Xiong J, Takeuchi M, Kurama T, Goeddel DV. TRAF6 is a signal transducer for interleukin-1. Nature 1996 Oct 3;383(6599):443-446. Ninomiya-Tsuji J, Kishimoto K, Hiyama A, lnoue J, Cao Z, Matsumoto K. The kinase TAK1 can activate the NIK—I kappaB as well as the MAP kinase cascade in the lL-1 signalling pathway. Nature 1999 Mar 1 8;398(6724):252-256. Heumann D, Roger T. Initial responses to endotoxins and Gram-negative bacteria. Clin Chim Acta 2002 Sep;323(1-2):59-72. Yamamoto M, Sato S, Hemmi H, Hoshino K, Kaisho T, Sanjo H, et al. Role of adaptor TRIF in the MyD88-independent toll-like receptor signaling pathway. Science 2003 Aug 1;301(5633):640-643. Schafer SL, Lin R, Moore PA, Hiscott J, Pitha PM. Regulation of type | interferon gene expression by interferon regulatory factor-3. J Biol Chem 1998 Jan 30;273(5):2714-2720. Ganey PE, Luyendyk JP, Maddox JF, Roth RA. Adverse hepatic drug reactions: inflammatory episodes as consequence and contributor. Chem Biol Interact 2004 Nov 1;150(1):35-51. Kaufrnann SH. Bacterial and protozoal infections in genetically disrupted mice. Curr Opin Immunol 1994 Aug;6(4):518-525. Tracey KJ, Cerami A. Tumor necrosis factor, other cytokines and disease. Annu Rev Cell Biol 1993;9:317-343. 283 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. Colletti LM, Kunkel SL, Walz A, Burdick MD, Kunkel RG, Wilke CA, et al. The role of cytokine networks in the local liver injury following hepatic ischemialreperfusion in the rat. Hepatology 1996 Mar;23(3):506-514. Ishida Y, Kondo T, Tsuneyama K, Lu P, Takayasu T, Mukaida N. The pathogenic roles of tumor necrosis factor receptor p55 in acetaminophen- induced liver injury in mice. J Leukoc Biol 2004 Jan;75(1):59-67. Raftery G, Griffiths B, Kay L, Kane D. Chronic viral hepatitis and TNF- alpha blockade. Rheumatology (Oxford) 2007 Aug;46(8):1381-1382. Shimizu S, Yamada Y, Okuno M, Ohnishi H, Osawa Y, Seishima M, et al. Liver injury induced by lipopolysaccharide is mediated by TNFR-1 but not by TNFR-2 or Fas in mice. Hepatol Res 2005 Mar;31(3):136-142. Wajant H, Pfizenmaier K, Scheurich P. Tumor necrosis factor signaling. Cell Death Differ 2003 Jan;10(1):45-65. Thirunavukkarasu C, Watkins SC, Gandhi CR. Mechanisms of endotoxin- induced NO, lL-6, and TNF-alpha production in activated rat hepatic stellate cells: role of p38 MAPK. Hepatology 2006 Aug;44(2):389—398. Tracey KJ, Cerami A. Tumor necrosis factor: an updated review of its biology. Crit Care Med 1993 Oct;21(10 Suppl):S415-S422. Locksley RM, Killeen N, Lenardo MJ. The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 2001 Feb 23; 1 04(4):487-501. Tartaglia LA, Ayres TM, Wong GH, Goeddel DV. A novel domain within the 55 kd TNF receptor signals cell death. Cell 1993 Sep 10;74(5):845- 853. Murray J, Barbara JA, Dunkley SA, Lopez AF, Van O, X, Condliffe AM, et al. Regulation of neutrophil apoptosis by tumor necrosis factor-alpha: requirement for TNF R55 and TNFR75 for induction of apoptosis in vitro. Blood 1997 Oct 1;90(7):2772-2783. Dri P, Haas E, Cramer R, Menegazzi R, Gasparini C, Martinelli R, et al. Role of the 75-kDa TNF receptor in TNF-induced activation of neutrophil respiratory burst. J Immunol 1999 Jan 1;162(1):460-466. Ohira H, Abe K, Yokokawa J, Takiguchi J, Rai T, Shishido S, et al. Adhesion molecules and CXC chemokines in endotoxin-induced liver injury. Fukushima J Med Sci 2003 Jun;49(1):1-13. Hanlon AM, Jang S, Salgame P. Signaling from cytokine receptors that affect Th1 responses. Front Biosci 2002 May 1;7:d1247-d1254. 284 89. 90. 91. 92. 93. 94. 95. 96. 97. 98. Friedl J, Puhlmann M, Bartlett DL, Libutti SK, Turner EN, Gnant MF, et al. Induction of permeability across endothelial cell monolayers by tumor necrosis factor (TN F) occurs via a tissue factor-dependent mechanism: relationship between the procoagulant and permeability effects of TNF. Blood 2002 Aug 15;100(4):1334-1339. Esmon CT. Possible involvement of cytokines in diffuse intravascular coagulation and thrombosis. Baillieres Best Pract Res Clin Haematol 1999 Sep;12(3):343-359. Perez-Ruiz M, Ros J, Morales-Ruiz M, Navasa M, Colmenero J, Ruiz-del- Arbol L, et al. Vascular endothelial growth factor production in peritoneal macrophages of cirrhotic patients: regulation by cytokines and bacterial lipopolysaccharide. Hepatology 1999 Apr;29(4):1057-1063. Mechtcheriakova D, Schabbauer G, Lucerna M, Clauss M, De MR, Binder BR, et al. Specificity, diversity, and convergence in VEGF and TNF-alpha signaling events leading to tissue factor up-regulation via EGR-1 in endothelial cells. FASEB J 2001 Jan;15(1):230-242. Grell M, Douni E, Wajant H, Lohden M, Clauss M, Maxeiner B, et al. The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell 1995 Dec 1;83(5):793- 802. Grell M, Wajant H, Zimmermann G, Scheurich P. The type 1 receptor (CD120a) is the high-affinity receptor for soluble tumor necrosis factor. Proc Natl Acad Sci U S A 1998 Jan 20;95(2):570-575. Morio LA, Chiu H, Sprowles KA, Zhou P, Heck DE, Gordon MK, et al. Distinct roles of tumor necrosis factor-alpha and nitric oxide in acute liver injury induced by carbon tetrachloride in mice. Toxicol Appl Pharmacol 2001 Apr 1;172(1):44-51. Peschon JJ, Torrance DS, Stocking KL, Glaccum MB, Otten C, Willis CR, et al. TNF receptor-deficient mice reveal divergent roles for p55 and p75 in several models of inflammation. J Immunol 1998 Jan 15;160(2):943-952. Kusters S, Tiegs G, Alexopoulou L, Pasparakis M, Douni E, Kunstle G, et al. In vivo evidence for a functional role of both tumor necrosis factor (TNF) receptors and transmembrane TNF in experimental hepatitis. Eur J Immunol 1997 Nov;27(11):2870-2875. Schumann J, Bluethmann H, Tiegs G. Synergism of Pseudomonas aeruginosa exotoxin A with endotoxin, superantigen, or TNF results in T. Immunol Lett 2000 Oct 3;74(2):165-172. 285 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. Hayder H, Blanden RV, Korner H, Riminton DS, Sedgwick JD, Mullbacher A. Adenovirus-induced liver pathology is mediated through TNF receptors 1 and II but is independent of TNF or Iymphotoxin. J Immunol 1999 Aug 1;163(3):1516-1520. Jaeschke H, Smith CW. Mechanisms of neutrophil-induced parenchymal cell injury. J Leukoc Biol 1997 Jun;61(6):647-653. Hewett JA, Schultze AE, VanCise S, Roth RA. Neutrophil depletion protects against liver injury from bacterial endotoxin. Lab Invest 1992 Mar;66(3):347-361 . Jaeschke H, Farhood A, Bautista AP, Spolarics Z, Spitzer JJ, Smith CW. Functional inactivation of neutrophils with a Mac-1 (CD11b/CD18) monoclonal antibody protects against ischemia-reperfusion injury in rat liver. Hepatology 1993 May;17(5):915-923. Luyendyk JP, Shaw PJ, Green CD, Maddox JF, Ganey PE, Roth RA. Coagulation-mediated hypoxia and neutrophil-dependent hepatic injury in rats given lipopolysaccharide and ranitidine. J Pharmacol Exp Ther 2005 Sep;314(3):1023-1031. Boehm U, Klamp T, Groot M, Howard JC. Cellular responses to interferon- gamma. Annu Rev Immunol 1997;15:749-795. Hasbold J, Hong JS, Kehry MR, Hodgkin PD. Integrating signals from IF N- gamma and lL-4 by B cells: positive and negative effects on CD40 ligand- induced proliferation, survival, and division-linked isotype switching to lgG1, lgE, and IgG2a. J Immunol 1999 Oct 15;163(8):4175-4181. Shtrichman R, Samuel CE. The role of gamma interferon in antimicrobial immunity. Curr Opin Microbiol 2001 Jun;4(3):251-259. Nakamura S, Otani T, ljiri Y, Motoda R, Kurimoto M, Orita K. IFN-gamma- dependent and -independent mechanisms in adverse effects caused by concomitant administration of lL-18 and lL-12. J Immunol 2000 Mar 15; 1 64(6):3330-3336. Salkowski CA, Thomas KE, Cody MJ, Vogel SN. Impaired IFN-gamma production in IFN regulatory factor-1 knockout mice during endotoxemia is secondary to a loss of both lL-12 and lL-12 receptor expression. J Immunol 2000 Oct 1;165(7):3970-3977. Walker W, ste-Amezaga M, Kastelein RA, Trinchieri G, Hunter CA. lL-18 and C028 use distinct molecular mechanisms to enhance NK cell production of lL-12-induced IFN-gamma. J Immunol 1999 May 15;162(10):5894-5901. 286 110. 111. 112. 113. 114. 115. 116. 117. 118. 119. 120. 121. Bach EA, Aguet M, Schreiber RD. The IFN gamma receptor: a paradigm for cytokine receptor signaling. Annu Rev Immunol 1997;15:563-591. Farrar MA, Schreiber RD. The molecular cell biology of interferon-gamma and its receptor. Annu Rev Immunol 1993;11:571-611. Soh J, Donnelly RJ, Kotenko S, Mariano TM, Cook JR, Wang N, et al. Identification and sequence of an accessory factor required for activation of the human interferon gamma receptor. Cell 1994 Mar 11;76(5):793-802. Bach EA, Tanner JW, Marsters S, Ashkenazi A, Aguet M, Shaw AS, et al. Ligand-induced assembly and activation of the gamma interferon receptor in intact cells. Mol Cell Biol 1996 Jun;16(6):3214-3221. Briscoe J, Guschin D, Rogers NC, Watling D, Muller M, Horn F, et al. JAKs, STATS and signal transduction in response to the interferons and other cytokines. Philos Trans R Soc Lond B Biol Sci 1996 Feb 29;351(1336):167-171. Jaruga B, Hong F, Kim WH, Gao B. lFN-gammalSTAT1 acts as a proinflammatory signal in T cell-mediated hepatitis via induction of multiple chemokines and adhesion molecules: a critical role of lRF-1. Am J Physiol Gastrointest Liver Physiol 2004 Nov;287(5):G1044-G1052. Tsung A, Stang MT, lkeda A, Critchlow ND, lzuishi K, Nakao A, et al. The transcription factor interferon regulatory factor-1 mediates liver damage during ischemia-reperfusion injury. Am J Physiol Gastrointest Liver Physiol 2006 Jun;290(6):G1261-G1268. Cassatella MA, Cappelli R, Della B, V, Grzeskowiak M, Dusi S, Berton G. Interferon-gamma activates human neutrophil oxygen metabolism and exocytosis. Immunology 1988 Mar;63(3):499-506. Ellis TN, Beaman BL. Interferon-gamma activation of polymorphonuclear neutrophil function. Immunology 2004 May;112(1):2-12. Tennenberg SD, Fey DE, Lieser MJ. Oxidative priming of neutrophils by interferon-gamma. J Leukoc Biol 1993 Mar;53(3):301-308. Meda L, Gasperini S, Ceska M, Cassatella MA. Modulation of proinflammatory cytokine release from human polymorphonuclear leukocytes by gamma interferon. Cell Immunol 1994 Sep;157(2):448-461. Kawada N, Mizoguchi Y, Kobayashi K, Morisawa S, Monna T, Yamamoto S. Interferon gamma modulates production of interleukin 1 and tumor necrosis factor by murine Kupffer cells. Liver 1991 Feb;11(1):42-47. 287 122. 123. 124. 125. 126. 127. 128. 129. 130. 131. 132. Collins PW, Noble KE, Reittie JR, Hoffbrand AV, Pasi KJ, Yong KL. Induction of tissue factor expression in human monocyte/endothelium cocultures. Br J Haematol 1995 Dec;91(4):963-970. Amman V, Stemme S, Rymo L, Risberg B. Interferon-gamma modulates the fibrinolytic response in cultured human endothelial cells. Thromb Res 1995 Mar 1;77(5):431-440. Malaguarnera L, lmbesi R, Di RM, Scuto A, Castrogiovanni P, Messina A, et al. Action of prolactin, IFN-gamma, TNF-alpha and LPS on heme oxygenase-1 expression and VEGF release in human monocytes/macrophages. Int lmmunopharrnacol 2005 Aug;5(9):1458— 1469. Ray PS, Fox PL. A post-transcriptional pathway represses monocyte VEGF-A expression and angiogenic activity. EMBO J 2007 Jul 25;26(14):3360-3372. Greenlund AC, Farrar MA, Viviano BL, Schreiber RD. Ligand-induced IFN gamma receptor tyrosine phosphorylation couples the receptor to its signal transduction system (p91). EMBO J 1994 Apr 1;13(7):1591-1600. Darnell JE, Jr., Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 1994 Jun 3;264(5164):1415-1421. Hashimoto W, Takeda K, Anzai R, Ogasawara K, Sakihara H, Sugiura K, et al. Cytotoxic NK1.1 Ag+ alpha beta T cells with intermediate TCR induced in the liver of mice by lL-12. J Immunol 1995 May 1;154(9):4333- 4340. Liu ZX, Govindarajan S, Okamoto S, Dennert G. NK cells cause liver injury and facilitate the induction of T cell-mediated immunity to a viral liver infection. J Immunol 2000 Jun 15;164(12):6480-6486. Wiltrout RH. Regulation and antimetastatic functions of liver-associated natural killer cells. Immunol Rev 2000 Apr;174:63-76. Lee HJ, Oh YK, Rhee M, Lim JY, Hwang JY, Park YS, et al. The role of STAT1/lRF-1 on synergistic ROS production and loss of mitochondrial transmembrane potential during hepatic cell death induced by LPS/d-GalN. J Mol Biol 2007 Jun 15;369(4):967-984. Liu ZX, Govindarajan S, Kaplowitz N. Innate immune system plays a critical role in determining the progression and severity of acetaminophen hepatotoxicity. Gastroenterology 2004 Dec; 127(6):1 760-1774. 288 133. 134. 135. 136. 137. 138. 139. 140. 141. 142. 143. 144. Gwag BJ, Lee YA, Ko SY, Lee MJ, Im DS, Yun BS, et al. Marked prevention of ischemic brain injury by Neu2000, an NMDA antagonist and antioxidant derived from aspirin and sulfasalazine. J Cereb Blood Flow Metab 2007 Jun;27(6):1142-1151. Terada Y, Inoshita S, Kuwana H, Kobayashi T, Okado T, Ichijo H, et al. Important role of apoptosis signal-regulating kinase 1 in ischemic acute kidney injury. Biochem Biophys Res Commun 2007 Dec 28;364(4):1043- 1049. Sidelmann JJ, Gram J, Jespersen J, Kluft C. Fibrin clot formation and lysis: basic mechanisms. Semin Thromb Hemost 2000;26(6):605-618. Baglia FA, Walsh PN. Thrombin-mediated feedback activation of factor XI on the activated platelet surface is preferred over contact activation by factor Xlla or factor Xla. J Biol Chem 2000 Jul 7;275(27):20514-20519. Nesheim ME, Mann KG. Thrombin-catalyzed activation of single chain bovine factor V. J Biol Chem 1979 Feb 25;254(4):1326-1334. Pieters J, Lindhout T, Hemker HC. In situ-generated thrombin is the only enzyme that effectively activates factor VIII and factor V in thromboplastin- activated plasma. Blood 1989 Aug 15;74(3):1021-1024. Coughlin SR. Sol Sherry lecture in thrombosis: how thrombin 'talks' to cells: molecular mechanisms and roles in vivo. Arterioscler Thromb Vasc Biol 1998 Apr;18(4):514-518. Kahn ML, Nakanishi-Matsui M, Shapiro MJ, lshihara H, Coughlin SR. Protease-activated receptors 1 and 4 mediate activation of human platelets by thrombin. J Clin Invest 1999 Mar;103(6):879-887. Kinlough-Rathbone RL, Perry DW, Guccione MA, Rand ML, Packham MA. Degranulation of human platelets by the thrombin receptor peptide SFLLRN: comparison with degranulation by thrombin. Thromb Haemost 1993 Dec 20;70(6):1019-1023. Naldini A, Sower L, Bocci V, Meyers B, Carney DH. Thrombin receptor expression and responsiveness of human monocytic cells to thrombin is linked to interferon-induced cellular differentiation. J Cell Physiol 1998 Oct;177(1):76-84. Umarova BA, Dugina TN, Shestakova EV, Gluza E, Strukova SM. Activation of rat mast cells upon stimulation of protease-activated receptor (PAR-1). Bull Exp Biol Med 2000 Apr;129(4):314-317. Hayashi T. [Tissue-type plasminogen activator (t-PA) and plasminogen activator inhibitor (PAI)]. Rinsho Byori 1994 Apr;42(4):346-351. 289 145. 146. 147. 148. 149. 150. 151. 152. 153. 154. 155. Hjortland GO, Lillehammer T, Somme S, Wang J, Halvorsen T, Juell S, et al. Plasminogen activator inhibitor-1 increases the expression of VEGF in human glioma cells. Exp Cell Res 2004 Mar 10;294(1):130-139. Kwak SH, Wang XQ, He Q, Fang WF, Mitra S, Bdeir K, et al. Plasminogen activator inhibitor-1 potentiates LPS-induced neutrophil activation through a JNK-mediated pathway. Thromb Haemost 2006 May;95(5):829-835. Renckens R, Roelofs JJ, de W, V, Florquin S, Lijnen HR, Carmeliet P, et al. The role of plasminogen activator inhibitor type 1 in the inflammatory response to local tissue injury. J Thromb Haemost 2005 May;3(5):1018- 1025. Laurens M, Defamie V, Scozzari G, Schmid-Alliana A, Gugenheim J, Crenesse D. Hypoxia-reoxygenation-induced chemokine transcription is not prevented by preconditioning or intermittent hypoxia, in mice hepatocytes. Transpl Int 2005 Apr;18(4):444-452. Ertel W, Morrison MH, Ayala A, Chaudry IH. Hypoxemia in the absence of blood loss or significant hypotension causes inflammatory cytokine release. Am J Physiol 1995 Jul;269(1 Pt 2):R160-R166. Murata Y, Ohteki T, Koyasu S, Hamuro J. IFN—gamma and pro- inflammatory cytokine production by antigen-presenting cells is dictated by intracellular thiol redox status regulated by oxygen tension. Eur J Immunol 2002 Oct;32(10):2866-2873. Forsythe JA, Jiang BH, lyer NV, Agani F, Leung SW, Koos RD, et al. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol 1996 Sep;16(9):4604-4613. Ganey PE, Luyendyk JP, Newport SW, Eagle TM, Maddox JF, Mackman N, et al. Role of the coagulation system in acetaminophen-induced hepatotoxicity in mice. Hepatology 2007 Oct;46(4):1177-1186. Harada N, Okajima K, Uchiba M. Dalteparin, a low molecular weight heparin, attenuates inflammatory responses and reduces ischemia- reperfusion-induced liver injury in rats. Crit Care Med 2006 Jul;34(7):1883- 1891. Hewett JA, Roth RA. The coagulation system, but not circulating fibrinogen, contributes to liver injury in rats exposed to lipopolysaccharide from gram-negative bacteria. J Pharmacol Exp Ther 1995 Jan;272(1):53- 62. Smith MK, Mooney DJ. Hypoxia leads to necrotic hepatocyte death. J Biomed Mater Res A 2007 Mar 1;80(3):520-529. 290 156. 157. 158. 159. 160. 161. 162. 163. 164. 165. 166. 167. Ferrara N, Carver-Moore K, Chen H, Dowd M, Lu L, O'Shea KS, et al. Heterozygous embryonic lethality induced by targeted inactivation of the VEGF gene. Nature 1996 Apr 4;380(6573):439-442. Ferrara N. Vascular endothelial growth factor and the regulation of angiogenesis. Recent Prog Horrn Res 2000;55:15-35. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003 Jun;9(6):669-676. Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 1989 Dec 8;246(4935):1 306-1309. Carmeliet P. VEGF gene therapy: stimulating angiogenesis or angioma- genesis? Nat Med 2000 Oct;6(10):1102-1 103. Zachary I. Signaling mechanisms mediating vascular protective actions of vascular endothelial growth factor. Am J Physiol Cell Physiol 2001 Jun;280(6):C1375-C1386. Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L, Gertsenstein M, et al. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature 1996 Apr 4;380(6573):435-439. Vink A, Schoneveld AH, Lamers D, Houben AJ, van der GP, van Diest PJ, et al. HIF-1 alpha expression is associated with an atheromatous inflammatory plaque phenotype and upregulated in activated macrophages. Atherosclerosis 2007 Dec;195(2):e69-e75. Charalambous C, Pen LB, Su YS, Milan J, Chen TC, Hofman FM. Interleukin-8 differentially regulates migration of tumor-associated and normal human brain endothelial cells. Cancer Res 2005 Nov 15;65(22):10347-10354. Chu SC, Tsai CH, Yang SF, Huang FM, Su YF, Hsieh YS, et al. Induction of vascular endothelial growth factor gene expression by proinflammatory cytokines in human pulp and gingival fibroblasts. J Endod 2004 Oct; 30(1 0):704-707. Kawaguchi M, Akagi M, Gray MJ, Liu W, Fan F, Ellis LM. Regulation of vascular endothelial growth factor expression in human gastric cancer cells by interleukin-1beta. Surgery 2004 Sep;136(3):686-692. Konishi N, Miki C, Yoshida T, Tanaka K, Toiyama Y, Kusunoki M. Interleukin-1 receptor antagonist inhibits the expression of vascular endothelial growth factor in colorectal carcinoma. Oncology 2005;68(2- 3):138-145. 291 168. 169. 170. 171. 172. 173. 174. 175. 176. 177. 178. Repovic P, Fears CY, Gladson CL, Benveniste EN. Oncostatin-M induction of vascular endothelial growth factor expression in astroglioma cells. Oncogene 2003 Nov 6;22(50):8117-8124. Yeh HH, Lai W, Chen HH, Liu HS, Su WC. Autocrine IL-6-induced Stat3 activation contributes to the pathogenesis of lung adenocarcinoma and malignant pleural effusion. Oncogene 2006 Jul 20;25(31):4300-4309. Cross MJ, Dixelius J, Matsumoto T, CIaesson-Welsh L. VEGF-receptor signal transduction. Trends Biochem Sci 2003 Sep;28(9):488-494. Takahashi T, Yamaguchi S, Chida K, Shibuya M. A single autophosphorylation site on KDR/Flk-1 is essential for VEGF-A-dependent activation of PLC-gamma and DNA synthesis in vascular endothelial cells. EMBO J 2001 Jun 1;20(11):2768—2778. Warner AJ, Lopez-Dee J, Knight EL, Feramisco JR, Prigent SA. The Shc- related adaptor protein, Sck, forms a complex with the vascular- endotheliaI-grth-factor receptor KDR in transfected cells. Biochem J 2000 Apr 15;347(Pt 2):501-509. Wu LW, Mayo LD, Dunbar JD, Kessler KM, Ozes ON, Warren RS, et al. VRAP is an adaptor protein that binds KDR, a receptor for vascular endothelial cell growth factor. J Biol Chem 2000 Mar 3;275(9):6059-6062. Qi JH, Claesson-Welsh L. VEGF-induced activation of phosphoinositide 3- kinase is dependent on focal adhesion kinase. Exp Cell Res 2001 Feb 1;263(1):173—182. Rousseau S, Houle F, Landry J, Huot J. p38 MAP kinase activation by vascular endothelial growth factor mediates actin reorganization and cell migration in human endothelial cells. Oncogene 1997 Oct;15(18):2169- 2177. Bates DO, Hillman NJ, Williams B, Neal CR, Pocock TM. Regulation of microvascular permeability by vascular endothelial growth factors. J Anat 2002 Jun;200(6):581-597. Reinders ME, Sho M, lzawa A, Wang P, Mukhopadhyay D, Koss KE, et al. Proinflammatory functions of vascular endothelial growth factor in alloimmunlty. J Clin Invest 2003 Dec;112(11):1655-1665. Kayisli UA, Demir R, Erguler G, Arici A. Vasodilator-stimulated phosphoprotein expression and its cytokine-mediated regulation in vasculogenesis during human placental development. Mol Hum Reprod 2002 Nov;8(11):1023-1030. 292 179. 180. 181. 182. 183. 184. 185. 186. 187. 188. Tsao PN, Chan FT, Wei SC, Hsieh WS, Chou HC, Su YN, et al. Soluble vascular endothelial growth factor receptor-1 protects mice in sepsis. Crit Care Med 2007 Aug;35(8):1955-1960. Tsuchihashi S, Ke B, Kaldas F, Flynn E, Busuttil RW, Briscoe DM, et al. Vascular endothelial growth factor antagonist modulates leukocyte trafficking and protects mouse livers against ischemialreperfusion injury. Am J Pathol 2006 Feb;168(2):695—705. Luyendyk JP, Lehman-McKeeman LD, Nelson DM, Bhaskaran VM, Reilly TP, Car BD, et al. Unique gene expression and hepatocellular injury in the lipopolysaccharide-ranitidine drug idiosyncrasy rat model: comparison with famotidine. Toxicol Sci 2006 Apr;90(2):569-585. Boulday G, Haskova Z, Reinders ME, Pal S, Briscoe DM. Vascular endothelial growth factor-induced signaling pathways in endothelial cells that mediate overexpression of the chemokine IFN-gamma-inducible protein of 10 kDa in vitro and in vivo. J Immunol 2006 Mar 1;176(5):3098- 3107. Schabbauer G, Schweighofer B, Mechtcheriakova D, Lucerna M, Binder BR, Hofer E. Nuclear factor of activated T cells and early growth response-1 cooperate to mediate tissue factor gene induction by vascular endothelial growth factor in endothelial cells. Thromb Haemost 2007 Jun;97(6):988-997. Pepper MS, Ferrara N, Orci L, Montesano R. Vascular endothelial growth factor (VEGF) induces plasminogen activators and plasminogen activator inhibitor-1 in microvascular endothelial cells. Biochem Biophys Res Commun 1991 Dec 16;181(2):902-906. Fischman AJ, Babich JW, Alpert NM, Vincent J, Wilkinson RA, Callahan RJ, et al. Pharmacokinetics of 18F-Iabeled trovafloxacin in normal and Escherichia coli-infected rats and rabbits studied with positron emission tomography. Clin Microbiol Infect 1997 Jun;3(3):379. Dalvie DK, Khosla N, Vincent J. Excretion and metabolism of trovafloxacin in humans. Drug Metab Dispos 1997 Apr;25(4):423-427. Outman WR, Nightingale CH. Metabolism and the fluoroquinolones. Am J Med 1989 Dec 29;87(6C):37S-42$. Melnik G, Schwesinger WH, Teng R, Dogolo LC, Vincent J. Hepatobiliary elimination of trovafloxacin and metabolites following single oral doses in healthy volunteers. Eur J Clin Microbiol Infect Dis 1998 Jun;17(6):424-426. 293 189. 190. 191. 192. 193. 194. 195. 196. 197. 198. 199. 200. Sun Q, Zhu R, Foss FW, Jr., Macdonald TL. Mechanisms of trovafloxacin hepatotoxicity: studies of a model cyclopropylamine-containing system. Bioorg Med Chem Lett 2007 Dec 15;17(24):6682-6686. Stahlmann R. Clinical toxicological aspects of fluoroquinolones. Toxicol Lett 2002 Feb 28;127(1-3):269-277. De SA, De SG. Adverse reactions to fluoroquinolones. an overview on mechanistic aspects. Curr Med Chem 2001 Mar;8(4):371-384. Chen HJ, Bloch KJ, Maclean JA. Acute eosinophilic hepatitis from trovafloxacin. N Engl J Med 2000 Feb 3;342(5):359-360. Lucena MI, Andrade RJ, Rodrigo L, Salmeron J, Alvarez A, Lopez-Garrido MJ, et al. Trovafloxacin-induced acute hepatitis. Clin Infect Dis 2000 Feb;30(2):400—401 . Blum MD, Graham DJ, McCloskey CA. Temafloxacin syndrome: review of 95 cases. Clin Infect Dis 1994 Jun;18(6):946-950. Zimpfer A, Propst A, Mikuz G, Vogel W, Terracciano L, Stadlmann S. Ciprofloxacin-induced acute liver injury: case report and review of literature. Virchows Arch 2004 Jan;444(1):87-89. Girard AE, Girard D, Gootz TD, Faiella JA, Cimochowski CR. In vivo efficacy of trovafloxacin (CP-99,219), a new quinolone with extended activities against gram-positive pathogens, Streptococcus pneumoniae, and Bacteroides fragilis. Antimicrob Agents Chemother 1995 Oct;39(10):2210-2216. Sokol WN, Jr., Sullivan JG, Acampora MD, Busman TA, Notario GF. A prospective, double-blind, multicenter study comparing clarithromycin extended-release with trovafloxacin in patients with community-acquired pneumonia. Clin Ther 2002 Apr;24(4):605-615. Croom KF, Goa KL. Levofloxacin: a review of its use in the treatment of bacterial infections in the United States. Drugs 2003;63(24):2769-2802. Onyeji CO, Bui KQ, Owens RC, Jr., Nicolau DP, Quintiliani R, Nightingale CH. Comparative efficacies of levofloxacin and ciprofloxacin against Streptococcus pneumoniae in a mouse model of experimental septicaemia. Int J Antimicrob Agents 1999 Jul;12(2):107-114. Lubasch A, Keller I, Borner K, Koeppe P, Lode H. Comparative pharmacokinetics of ciprofloxacin, gatifloxacin, grepafloxacin, levofloxacin, trovafloxacin, and moxifloxacin after single oral administration in healthy volunteers. Antimicrob Agents Chemother 2000 Oct;44(10):2600-2603. 294 201. 202. 203. 204. 205. 206. 207. 208. 209 210. 211. Witkamp R, Monshouwer M. Signal transduction in inflammatory processes, current and future therapeutic targets: a mini review. Vet Q 2000 Jan;22(1):11-16. Uetrecht J. Screening for the potential of a drug candidate to cause idiosyncratic drug reactions. Drug Discov Today 2003 Sep 15;8(18):832- 837. . Liguori MJ, Anderson MG, Bukofzer S, McKim J, Pregenzer JF, Retief J, et al. Microarray analysis in human hepatocytes suggests a mechanism for hepatotoxicity induced by trovafloxacin. Hepatology 2005 Jan;41(1):177-186. Ng W, Lutsar I, Wubbel L, Ghaffar F, Jafri H, McCracken GH, et al. Pharmacodynamics of trovafloxacin in a mouse model of cephalosporin- resistant Streptococcus pneumoniae pneumonia. J Antimicrob Chemother 1999 Jun;43(6):811-816. Teng R, Girard D, Gootz TD, Foulds G, Liston TE. Pharmacokinetics of trovafloxacin (OP-99,219), a new quinolone, in rats, dogs, and monkeys. Antimicrob Agents Chemother 1996 Mar;40(3):561-566. Nightingale SL. From the Food and Drug Administration. JAMA 1999 Jul 7;282(1):19. Teng R, Harris SC, Nix DE, Schentag JJ, Foulds G, Liston TE. Pharmacokinetics and safety of trovafloxacin (OP-99,219), a new quinolone antibiotic, following administration of single oral doses to healthy male volunteers. J Antimicrob Chemother 1995 Aug;36(2):385-394. Ernst ME, Ernst EJ, Klepser ME. Levofloxacin and trovafloxacin: the next generation of fluoroquinolones? Am J Health Syst Pharm 1997 Nov 15;54(22):2569-2584. . Colletti LM, Remick DG, Burtch GD, Kunkel SL, Strieter RM, Campbell DA, Jr. Role of tumor necrosis factor-alpha in the pathophysiologic alterations after hepatic ischemialreperfusion injury in the rat. J Clin Invest 1990 Jun;85(6):1936-1943. Tiegs G, Walter M, Wendel A. Tumor necrosis factor is a terminal mediator in galactosamine/endotoxin-induced hepatitis in mice. Biochem Pharmacol 1989 Feb 15;38(4):627-631. Windmeier C, Gressner AM. Pharmacological aspects of pentoxifylline with emphasis on its inhibitory actions on hepatic fibrogenesis. Gen Pharmacol 1997 Aug;29(2):181-196. 295 212. 213. 214. 215. 216. 217. 218. 219. 220. 221. Kiemer AK, Muller C, Vollmar AM. Inhibition of LPS-induced nitric oxide and TNF-alpha production by alpha-lipoic acid in rat Kupffer cells and in RAW 264.7 murine macrophages. Immunol Cell Biol 2002 Dec;80(6):550- 557. Tsukada S, Enomoto N, Takei Y, Hirose M, lkejima K, Kitamura T, et al. Dalteparin sodium prevents liver injury due to lipopolysaccharide in rat through suppression of tumor necrosis factor-alpha production by Kupffer cells. Alcohol Clin Exp Res 2003 Aug;27(8 Suppl):7S-11S. Khan AA, Slifer TR, Araujo FG, Suzuki Y, Remington JS. Protection against lipopolysaccharide-induced death by fluoroquinolones. Antimicrob Agents Chemother 2000 Nov;44(11):3169-3173. Purswani M, Eckert S, Arora H, Johann-Liang R, Noel GJ. The effect of three broad-spectrum antimicrobials on mononuclear cell responses to encapsulated bacteria: evidence for down-regulation of cytokine mRNA transcription by trovafloxacin. J Antimicrob Chemother 2000 Dec;46(6):921-929. Shaw PJ, Hopfensperger MJ, Ganey PE, Roth RA. Lipopolysaccharide and trovafloxacin coexposure in mice causes idiosyncrasy-like liver injury dependent on tumor necrosis factor-alpha. Toxicol Sci 2007 Nov;100(1):259-266. Schwandner R, Dziarski R, Wesche H, Rothe M, Kirschning CJ. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by toll-like receptor 2. J Biol Chem 1999 Jun 18;274(25):17406-17409. Yoshimura A, Lien E, lngalls RR, Tuomanen E, Dziarski R, Golenbock D. Cutting edge: recognition of Gram-positive bacterial cell wall components by the innate immune system occurs via Toll-like receptor 2. J Immunol 1999 Jul 1;163(1):1-5. Yee SB, Hanumegowda UM, Hotchkiss JA, Ganey PE, Roth RA. Role of neutrophils in the synergistic liver injury from monocrotaline and bacterial lipopolysaccharide exposure. Toxicol Sci 2003 Mar;72(1):43-56. Majcherczyk PA, Rubli E, Heumann D, Glauser MP, Moreillon P. Teichoic acids are not required for Streptococcus pneumoniae and Staphylococcus aureus cell walls to trigger the release of tumor necrosis factor by peripheral blood monocytes. Infect Immun 2003 Jul;71(7):3707-3713. Moreillon P, Majcherczyk PA. Proinflammatory activity of cell-wall constituents from gram-positive bacteria. Scand J Infect Dis 2003;35(9):632-641. 296 222. 223. 224. 225. 226. 227. 228. 229. 230. 231. 232. Weber JR, Moreillon P, Tuomanen El. Innate sensors for Gram-positive bacteria. Curr Opin Immunol 2003 Aug; 1 5(4):408-41 5. Gerosa F, Baldani-Guerra B, Lyakh LA, Batoni G, Esin S, Winkler-Pickett RT, et al. Differential regulation of interleukin 12 and interleukin 23 production in human dendritic cells. J Exp Med 2008 Jun 9;205(6):1447- 1461. - Perrin-Cocon L, Agaugue S, Diaz 0, Vanbervliet B, Dollet S, Guironnet- Paquet A, et al. Th1 disabled function in response to TLR4 stimulation of monocyte-derived DC from patients chronically-infected by hepatitis C virus. PLoS ONE 2008;3(5):e2260. Del VM, Bajetta E, Canova S, Lotze MT, Wesa A, Parmiani G, et al. Interleukin-12: biological properties and clinical application. Clin Cancer Res 2007 Aug 15;13(16):4677-4685. Ouaaz F, Li M, Beg AA. A critical role for the RelA subunit of nuclear factor kappaB in regulation of multiple immune-response genes and in Fas-induced cell death. J Exp Med 1999 Mar 15;189(6):999-1004. Li X, Klintman D, Liu Q, Sato T, Jeppsson B, Thorlacius H. Critical role of CXC chemokines in endotoxemic liver injury in mice. J Leukoc Biol 2004 Mar;75(3):443-452. Deng X, Luyendyk JP, Zou W, Lu J, Malle E, Ganey PE, et al. Neutrophil interaction with the hemostatic system contributes to liver injury in rats cotreated with lipopolysaccharide and ranitidine. J Pharmacol Exp Ther 2007 Aug;322(2):852-861. Jaeschke H, Farhood A, Fisher MA, Smith CW. Sequestration of neutrophils in the hepatic vasculature during endotoxemia is independent of beta 2 integrins and intercellular adhesion molecule-1. Shock 1996 Nov;6(5):351-356. Yoshimura T, Takahashi M. IFN-gamma-mediated survival enables human neutrophils to produce MCP-1/CCL2 in response to activation by TLR ligands. J Immunol 2007 Aug 1;179(3):1942-1949. Shiratsuchi Y, lyoda T, Tanimoto N, Kegai D, Nagata K, Kobayashi Y. Infiltrating neutrophils induce allospecific CTL in response to immunization with apoptotic cells via MCP-1 production. J Leukoc Biol 2007 Feb;81(2):412-420. Walcheck B, Herrera AH, St HC, Mattila PE, Whitney AR, Deleo FR. ADAM17 activity during human neutrophil activation and apoptosis. Eur J Immunol 2006 Apr;36(4):968-976. 297 233. 234. 235. 236. 237. 238. 239. 240. 241. 242. Bertino J, Jr., Fish D. The safety profile of the fluoroquinolones. Clin Ther 2000 Jul;22(7):798-817. Hayder H, Blanden RV, Korner H, Riminton DS, Sedgwick JD, Mullbacher A. Adenovirus-induced liver pathology is mediated through TNF receptors l and II but is independent of TNF or Iymphotoxin. J Immunol 1999 Aug 1;163(3):1516—1520. Kusters S, Tiegs G, Alexopoulou L, Pasparakis M, Douni E, Kunstle G, et al. In vivo evidence for a functional role of both tumor necrosis factor (T NF) receptors and transmembrane TNF in experimental hepatitis. Eur J Immunol 1997 Nov;27(11):2870-2875. Schumann J, Bluethmann H, Tiegs G. Synergism of Pseudomonas aeruginosa exotoxin A with endotoxin, superantigen, or TNF results in T. Immunol Lett 2000 Oct 3;74(2):165-172. Copple BL, Banes A, Ganey PE, Roth RA. Endothelial cell injury and fibrin deposition in rat liver after monocrotaline exposure. Toxicol Sci 2002 Feb;65(2):309-318. Natoli G, Costanzo A, Moretti F, Fulco M, Balsano C, Levrero M. Tumor necrosis factor (T NF) receptor 1 signaling downstream of TNF receptor- associated factor 2. Nuclear factor kappaB (NFkappaB)-inducing kinase requirement for activation of activating protein 1 and NFkappaB but not of c-Jun N-terminal kinase/stress-activated protein kinase. J Biol Chem 1997 Oct 17;272(42):26079-26082. Varghese J, Chattopadhaya S, Sarin A. Inhibition of p38 kinase reveals a TNF-alpha-mediated, caspase-dependent, apoptotic death pathway in a human myelomonocyte cell line. J Immunol 2001 Jun 1;166(11):6570- 6577. Nowak M, Gaines GC, Rosenberg J, Minter R, Bahjat FR, Rectenwald J, et al. LPS-induced liver injury in D-galactosamine—sensitized mice requires secreted TNF-alpha and the TNF-p55 receptor. Am J Physiol Regul Integr Comp Physiol 2000 May;278(5):R1202-R1209. Dopp JM, Sarafian TA, Spinella FM, Kahn MA, Shau H, de VJ. Expression of the p75 TNF receptor is linked to TNF-induced NFkappaB translocation and oxyradical neutralization in glial cells. Neurochem Res 2002 Nov;27(11):1535-1542. Tartaglia LA, Pennica D, Goeddel DV. Ligand passing: the 75-kDa tumor necrosis factor (T NF) receptor recruits TNF for signaling by the 55-kDa TNF receptor. J Biol Chem 1993 Sep 5;268(25):18542-18548. 298 243. 244. 245 246. 247. 248. 249. 250. 251. 252. 253. 254. Pelagi M, Cumis F, Colombo B, Rovere P, Sacchi A, Manfredi AA, et al. Caspase inhibition reveals functional cooperation between p55— and p75— TNF receptors in cell necrosis. Eur Cytokine Netw 2000 Dec;11(4):580- 588. Tukov FF, Luyendyk JP, Ganey PE, Roth RA. The role of tumor necrosis factor alpha in Iipopolysaccharide/ranitidine-induced inflammatory liver injury. Toxicol Sci 2007 Nov;100(1):267-280. . Kamochi M, Kamochi F, Kim YB, Sawh S, Sanders JM, Sarembock I, et al. P-selectin and lCAM-1 mediate endotoxin-induced neutrophil recruitment and injury to the lung and liver. Am J Physiol 1999 Aug;277(2 Pt 1):L310- L319. Dri P, Haas E, Cramer R, Menegazzi R, Gasparini C, Martinelli R, et al. Role of the 75-kDa TNF receptor in TNF-induced activation of neutrophil respiratory burst. J Immunol 1999 Jan 1;162(1):460-466. Nawroth P, Handley D, Matsueda G, De WR, Gerlach H, Blohm D, et al. Tumor necrosis factor/cachectin-induced intravascular fibrin formation in meth A fibrosarcomas. J Exp Med 1988 Aug 1;168(2):637-647. Takeshita Y, Takamura T, Hamaguchi E, Shimizu A, Ota T, Sakurai M, et al. Tumor necrosis factor-alpha-induced production of plasminogen activator inhibitor 1 and its regulation by pioglitazone and cerivastatin in a nonmalignant human hepatocyte cell line. Metabolism 2006 Nov;55(11):1464-1472. Schwabe RF, Brenner DA. Mechanisms of Liver Injury. I. TNF-alpha- induced liver injury: role of IKK, JNK, and ROS pathways. Am J Physiol Gastrointest Liver Physiol 2006 Apr;290(4):G583-6589. Sheri HM, Pervaiz S. TNF receptor superfamin-induced cell death: redox- dependent execution. FASEB J 2006 Aug;20(10):1589-1598. Gezginci S, Bolkent S. The effect of Z-FA.FMK on D-galactosamine/TNF- alpha-induced liver injury in mice. Cell Biochem Funct 2007 May;25(3):277-286. Wu D, Cederbaum A. Cytochrome P4502E1 sensitizes to tumor necrosis factor alpha-induced liver injury through activation of mitogen-activated protein kinases in mice. Hepatology 2008 Mar;47(3):1005-1017. Bemelmans MH, van Tits LJ, Buun'nan WA. Tumor necrosis factor: function, release and clearance. Crit Rev Immunol 1996;16(1):1-11. Tafazoli S, Spehar DD, O'Brien PJ. Oxidative stress mediated idiosyncratic drug toxicity. Drug Metab Rev 2005;37(2):311-325. 299 255. 256. 257. 258. 259. 260. 261. 262. 263. 264. 265. 266. Varrna TK, Lin CY, Toliver-Kinsky TE, Shem/cod ER. Endotoxin-induced gamma interferon production: contributing cell types and key regulatory factors. Clin Diagn Lab Immunol 2002 May;9(3):530-543. Kim WH, Hong F, Radaeva S, Jaruga B, Fan S, Gao B. STAT1 plays an essential role in LPS/D-galactosamine-induced liver apoptosis and injury. Am J Physiol Gastrointest Liver Physiol 2003 Oct;285(4):G761-G768. Kusters S, Gantner F, Kunstle G, Tiegs G. Interferon gamma plays a critical role in T cell-dependent liver injury in mice initiated by concanavalin A. Gastroenterology 1996 Aug;1 11(2):462-471. Rajagopalan D. A comparison of statistical methods for analysis of high density oligonucleotide array data. Bioinformatics 2003 Aug 12;19(12):1469-1476. Stark GR, Kerr IM, Williams BR, Silverman RH, Schreiber RD. How cells respond to interferons. Annu Rev Biochem 1998;67:227-264. Tsutsui H, Matsui K, Okamura H, Nakanishi K. Pathophysiological roles of interleukin-18 in inflammatory liver diseases. Immunol Rev 2000 Apr; 1 74: 1 92-209. Senaldi G, Shaklee CL, Guo J, Martin L, Boone T, Mak TW, et al. Protection against the mortality associated with disease models mediated by TNF and IFN-gamma in mice lacking IFN regulatory factor-1. J Immunol 1999 Dec 15;163(12):6820-6826. Kano A, Watanabe Y, Takeda N, Aizawa S, Akaike T. Analysis of IFN- gamma-induced cell cycle arrest and cell death in hepatocytes. J Biochem 1 997 Apr; 1 21 (4):677-683. Adamson GM, Billings RE. Cytokine toxicity and induction of NO synthase activity in cultured mouse hepatocytes. Toxicol Appl Pharmacol 1993 Mar;119(1):100-107. Edwards SW, Say JE, Hughes V. Gamma interferon enhances the killing of Staphylococcus aureus by human neutrophils. J Gen Microbiol 1988 Jan;134(1):37-42. Suzuki K, F urui H, Kaneko M, Takagi K, Satake T. Priming effect of recombinant human interleukin-2 and recombinant human interferon- gamma on human neutrophil superoxide production. Arzneimittelforschung 1990 Oct;40(10):1176-1 179. Adams DO, Hamilton TA. The cell biology of macrophage activation. Annu Rev Immunol 1984;2:283-318. 300 267. 268. 269. 270. 271. 272. 273. 274. 275. 276. 277. Koj A, Gauldie J, Regoeczi E, Sauder DN, Sweeney GD. The acute-phase response of cultured rat hepatocytes. System characterization and the effect of human cytokines. Biochem J 1984 Dec 1;224(2):505-514. Luyendyk JP, Copple BL, Barton CC, Ganey PE, Roth RA. Augmentation of aflatoxin B1 hepatotoxicity by endotoxin: involvement of endothelium and the coagulation system. Toxicol Sci 2003 Mar;72(1):171-181. Luyendyk JP, Lehman-McKeeman LD, Nelson DM, Bhaskaran VM, Reilly TP, Car 80, et al. Coagulation-dependent gene expression and liver injury in rats given lipopolysaccharide with ranitidine but not with famotidine. J Pharmacol Exp Ther 2006 May;317(2):635-643. Yee SB, Hanumegowda UM, Copple BL, Shibuya M, Ganey PE, Roth RA. Endothelial cell injury and coagulation system activation during synergistic hepatotoxicity from monocrotaline and bacterial lipopolysaccharide coexposure. Toxicol Sci 2003 Jul;74(1):203-214. Ranby M, Brandstrom A. Biological control of tissue plasminogen activator-mediated fibrinolysis. Enzyme 1988;40(2-3):130-143. Gnant MF, Turner EM, Alexander HR, Jr. Effects of hyperthermia and tumour necrosis factor on inflammatory cytokine secretion and procoagulant activity in endothelial cells. Cytokine 2000 Apr;12(4):339-347. Chandel NS, Maltepe E, Goldwasser E, Mathieu CE, Simon MC, Schumacker PT. Mitochondrial reactive oxygen species trigger hypoxia- induced transcription. Proc Natl Acad Sci U S A 1998 Sep 29;95(20):11715-11720. Chandel NS, McClintock DS, Feliciano CE, Wood TM, Melendez JA, Rodriguez AM, et al. Reactive oxygen species generated at mitochondrial complex Ill stabilize hypoxia-inducible factor-1alpha during hypoxia: a mechanism of O2 sensing. J Biol Chem 2000 Aug 18;275(33):25130— 25138. Smith MK, Mooney DJ. Hypoxia leads to necrotic hepatocyte death. J Biomed Mater Res A 2007 Mar 1;80(3):520-529. Forsythe JA, Jiang BH, lyer NV, Agani F, Leung SW, Koos RD, et al. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol 1996 Sep;16(9):4604-4613. Arteel GE. New role of plasminogen activator inhibitor-1 in alcohol-induced liver injury. J Gastroenterol Hepatol 2008 Mar;23 Suppl 12854-859. 301 278. 279. 280. 281. 282. Bergheim I, Guo L, Davis MA, Duveau I, Arteel GE. Critical role of plasminogen activator inhibitor-1 in cholestatic liver injury and fibrosis. J Pharmacol Exp Ther 2006 Feb;316(2):592-600. Deng X, Lu J, Lehman-McKeeman LD, Malle E, Crandall D, Ganey PE, et al. p38 MAPK-dependent TNF-{alpha} converting enzyme is important for liver injury in hepatotoxic interaction between lipopolysaccharide and ranitidine. J Pharmacol Exp Ther 2008 Apr 4. Renckens R, Pater JM, van der PT. Plasminogen activator inhibitor type- 1-deficient mice have an enhanced IFN-gamma response to lipopolysaccharide and staphylococcal enterotoxin B. J Immunol 2006 Dec 1;177(11):8171-8176. Balsara RD, Castellino FJ, Ploplis VA. A novel function of plasminogen activator inhibitor-1 in modulation of the AKT pathway in wild-type and plasminogen activator inhibitor-1-deficient endothelial cells. J Biol Chem 2006 Aug 11;281(32):22527-22536. Wanidworanun C, Strober W. Predominant role of tumor necrosis factor- alpha in human monocyte lL-10 synthesis. J Immunol 1993 Dec 15;151(12):6853-6861. 302 Illllllllllljllrilli