. . r. gm”. (vi. .4 $1.”: 42% W.% t... fin.” ~ I r . . 2:... w 5.. Au. . 7 .35 . . . . 1i. i : Cm“, A r: . 53.43.... . n (mnrfiv‘tiaa . . .27.th . 9! A . i a , a ‘3 w? a 3 7’1"}. .. .61 ~ . .53.! firm“- . . ..u;,.{..fia¢ .3. 6. if!!! “.35. P . «502,405.24. It?“ Amwrfiwnfiiramu .2 . . ham... flusflmwflki . 1...»... 9...... i... .ta...w .3. . insnfl .idmflmnm. has: 3% 2- Mm. _ 6. a .1... . . 94.3.. .9}: a MA 2 $8.”? 2 . .. . 3- fiéfimfizxfim » E 54!!" It; . 19 is; 349.5,}..713 4. .1. A3. .5 12. t){ '3. {.3 2.1.”... .‘ x ( er- a 3.431%?!- ....5. . 2m. , 3.....-«9». . a: .1 . Ln: . .. LIBRARIES ’3 MICHIGAN STATE UNIVERSITY d 0 05. EAST LANSING, MICH 48824-1048 6‘ ' l Biff/$3 This is to certify that the dissertation entitled ROLES OF PULMONARY ANGIOTENSIN SYSTEM IN THE DEVELOPMENT OF PULMONARY FIBROSIS presented by XIAOPENG LI has been accepted towards fulfillment of the requirements for the Ph. D. degree in Physiologi Major Professor's Signature lz { I; '[o 1— Date MSU is an Affirmative Action/Equal Opponunity Institution PLACE IN RE‘lURN 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 3%? 31‘ 2.011 v _ 1 I, 2/05 cEifiC/DatoDueIndd-pJS ROL ROLES OF PULMONARY ANGIOTENSIN SYSTEM IN THE DEVELOPMENT OF PULMONARY FIBROSIS By Xiaopeng Li A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PI-HLOSOPHY Department of Physiology 2004 ROLES OF Treatment ft suggesting tl llortallt} ot‘l Nth the pres aheolar epit'r fibrosis can b The l0: amptosis “a liltllt‘allon lhd of anglOlL’nsl mO-dels. Our AEC S tthgl“ Ligm N- amitosis can 3F;"S‘Hl'tillerbq'nogL ll recepmr l:\' Sh» _ Ufieqtlem 1W1;- ABSTRACT ROLES OF PULMONARY ANGIOTENSIN SYSTEM IN THE DEVELOPMENT OF PULMONARY FIBROSIS By Xiaopeng Li Treatment for IPF targeting the suppression of inflammation has not been successful, suggesting that inflammation is not the sole mechanism underlying lung fibrogenesis. Mortality of IPF patients is not dependent on severity of inflammation, but correlates well with the presence of “fibroblastic foci” and adjacent failure of reepithelization. Loss of alveolar epithelial cells (AECS) and failure of reepithelization characterized in pulmonary fibrosis can be considered as profibrotic and are believed to initiate the fibrotic lesion. The loss of AECS could result from necrosis and/ or apoptosis. Increased level of apoptosis was found in AECS in experimental and human pulmonary fibrosis. One indication that the angiotensin system is involved in fibrogenesis is findings that blockade of angiotensin systems blocked Pulmonary Fibrosis (PF) at least in several animal models. Our lab showed that angiotensin II (ANG II) induces apoptosis of the primary AECS through ANG 11 type I (AT1) receptor in vitro. Apoptosis of AECS induced by F as Ligand, TNF-alpha, and amiodarone requires angiotensin synthesis de novo as AECS apoptosis can be blocked by the antisense oligonucleotide against the mRNA of the angiotensinogen (ANGEN), angiotensin converting enzyme (ACE) inhibitors, and ANG II receptor (AT receptor) antagonists in vitro. Taken together, those data suggest that ANG II, the processed product of ANGEN, is the key to regulate apoptosis of AECS and subsequent lung fibrosis. lite existent endocrine K45~ sistem compo“C and amiodamne and mouse pulmi Bmuch still unki in bleemuin-intl‘ l to determine the pulmonan‘ libmsi including its con essential role in tl through ANG 11-. that: llAEC apo tritibiled b)‘ ANG to blenmycin at It Gillie ATl-Selet'l BLED-induced .-\ epithelial cell: and liltilo; 5’ Admin beemcin-induwd The existence of “local angiotensin system” in the lung, which is independent of the endocrine RAS, is supported by studies demonstrating the expression of angiotensin system components in cultured primary rat AECS in response to Fas Ligand, TNF—alpha, and amiodarone and myofibroblasts from human fibrotic lungs. Bleomycin-induced rat and mouse pulmonary fibrosis model is a well-studied model for fibrogenesis. But there is much still unknown about the components and roles of pulmonary angiotensin system in bleomycin-induced pulmonary fibrosis. The overall objective of my thesis project was to determine the roles of pulmonary angiotensisn system in the development of pulmonary fibrosis. We tested the overall hypothesis: the pulmonary angiotensin system including its components angiotensinogen, cathepsin D and AT1 receptor, plays an essential role in the development of bleomycin-induced pulmonary fibrosis at least in part through ANG II-ATl receptor pathway mediated apoptosis of AECS. Our data showed that: 1) ABC apoptosis in response to bleomycin (BLEO) requires ANG synthesis and is inhibited by ANG system antagonists; 2) Cat D is required for ABC apoptosis in response to bleomycin at least in part by converting angiotensinogen to ANGI; 3) Administration of the ATl-selective receptor antagonist and deletion of the ATla receptor gene block BLED-induced AEC apoptosis and lung fibrosis; 4) The apoptotic type II alveolar epithelial cells and myofibroblasts are the major cellular sources of lung-derived ANGEN in vivo; 5) Administration of the antisense against ANGEN messenger RNA attenuates bleomycin-induced AEC apoptosis and lung fibrosis. 7’0”}: ‘To my level) mfifllfin Luo, you malig tfiis possilifla. To my parents, Han-Tu Li and'QBao-‘Yu Clien, for your uttermost lime and support iv This dissertatit assistance ofrr First. I “ant to Ural. I thank I: [lit past four )i as how to \triti cntics. hon to r 1 next tub-h u. C0mmittee: DB [hank .VOU Of a] Particular”. l \\ “hen l “as rotnt mm] for all t grammar and SPC I “Out Id “1‘6 10 ti ~ I Acknowledgments This dissertation project was the outcome of my hard work and the input, advice, and the assistance of many other people. First, I want to express my gratitude, respect and admiration for my mentor, Dr. Bruce D. Uhal. I thank Dr. Uhal for his support, understanding and his patient training of me over the past four years. He trained me from every aspect to be an independent scientist such as how to write a proposal, how to design an experiment, how to response to scientific critics, how to review other scientists’ work and so on. I next wish to thank and acknowledge the members of my dissertation guidance committee: Drs. Greg Fink, David Kreulen, Stephanie Watts and Douglas Luckie. I thank you of all for your precious advice and endorsement for my dissertation project. Particularly, I would like to thank Dr. Kreulen for giving me the initial scientific training when I was rotating in his lab where I started my scientific life. In addition I am deeply grateful for all of you for your critical reading this dissertation and correction of the grammar and spelling mistakes, especially Drs. Watts, Luckie, Kreulen. I would like to thank the Department of Physiology for recruiting me and providing me the excellent academic and extracurricular trainings. I also thank the American Heart Association Midwestern Affiliate for awarding me a pre-doctoral fellowship (2004). I would like to acknowledge Dr. Uhal as my sponsor and Drs. Fink, Kreulen, Spielman for writing recommendation letters for the fellowship application. Then. I thank for part of m} helped me alt Zhuang. Dr. l Conrad. I also would BoeSen. I am: living in this e: Finally l “on tmpossible for their uttermmt Then, I thank Dr. Jump and Dr. Olson’s lab for letting me use their space and equipments for part of my experiments. In addition, I would to thank all those individuals who have helped me along the way. They are my colleagues in Dr. Uhal lab including Dr. Jiaju Zhuang, Dr. Huiying Zhang, Ruijie Shu, Jong-kyong Kim, Heather Rayford, Valerie Conrad. I also would like to thank my American Friendship family: Hank Hanson and Chris Boesen. I appreciate their friendship, support and endeavors to enrich my experience of living in this country for the past years. Finally I would like to acknowledge my wife Min Luo: without you, it would be impossible for me to achieve this goal, and my parents Han-F u Li and Bao-Yu Chen for their uttermost love and support. vi LIST OF F LIST OF A CHAPT ER 1. ldior 1.1 E TABLE OF CONTENTS LIST OF FIGURES .............................................................................. viii LIST OF ABBREVIATIONS ................................................................. xxiii CHAPTER 1: GENERAL INTRODUCTION ................................................. l 1. Idiopathic pulmonary fibrosis ........................................................... l 1.1 Epidemiology of IPF .................................................................... 1 1.2 Predisposing factors for IPF ............................................................ 1 1.2.1 Geneticfactors...... 2 1.2.2 Environmentalfactors... ......3 1.3 Clinical features ........................................................................ 4 1.4 Histopathology .......................................................................... 6 1.5 Therapeutic options for pulmonary fibrosis ......................................... 7 2. Bleomycin-Induced Experimental Pulmonary Fibrosis Model ................... 8 3. Mechanisms of Fibrogenesis ............................................................ 10 3.1 The roles of alveolar epithelial cells (AECS) ....................................... 11 3.2 Consequence of alveolar epithelial cells damage/injury .......................... 13 3.2.1 Loss ofpermeability barrier... ...13 3.2.2 Loss of the barrier limiting fibroblast migration into the alveolar a1rspace13 3. 2.3 Loss of cell surface that prevents collapse and fusion of alveolar walls] 4 3.2.4SurfactantAlterations.................................................................14 3.2.5 Loss ofthe Alveolar Progenitor Cells... ....15 3.2.6 Loss ofActive Transport Properties... 16 vii 3. 2. 7Loss of the ability to clear intra-alveolar fibrin and regulate the plasminogenactivationsystem...................................................17 3.2.8 Abnormal AECs as a primary source of profibrotic cytokines... 19 4. Apoptosis of AECs is involved in pulmonary fibrogenesis ..................... 19 4.1 General information about apoptosis .............................................. 19 4.2 Epithelial cell apoptosis in fibrotic lungs .......................................... 20 4.3 Mechanism and signaling of epithelial apoptosis in fibrotic lung ............. 23 5. Renin-Angiotensin System ............................................................. 24 5.1 General information .................................................................. 24 5.2 Effects of Angiotensin II ............................................................ 25 5.3 Angiotensin 11 signal pathway ...................................................... 25 6. Pulmonary angiotensin system in idiopathic pulmonary fibrosis ............... 27 6.1 Local pulmonary angiotensin system ............................................... 27 6.2 Pulmonary angiotensin system components ....................................... 28 6.2.2 Renin and Renin- like enzymes... ......29 6.2.3 ACE andACE-like enzymes... ...29 6.2.4Angiotensin IIreceptors...... ........30 6.3 Potential roles of pulmonary ANG II in pulmonary fibrosis ..................... 31 6. 3. 1 Pulmonary angiotensin system is linked to lung epithelial apoptosis....31 6.3.2 Mytogenic for fibroblasts and Activate TGF beta expression... 31 6.3.3 Inhibition ofthefibrinolyticpathway... ...32 6. 3. 4 Downregulation of hepatocyte growth factor (HGF) expression ......... 32 viii 6.4 At 6.1 7- “iorkir 8. Signific CHAPTER 2 CHAPTER 3: EPITHEUAI Abstracz. IntrodUm Mclhfildg Results. .. 6.4 Angiotensin system is involved in pulmonary fibrosis ........................... 33 6.4.1. Increased levels of converting enzymes and A T1 receptors in IPF ...... 33 6. 4. 2. Blockade of experimental lung fibrosis by angiotensin system ............ 34 antagonists 6.4.3. Blockade of apoptosis by ACE inhibitors or caspases inhibitors blocks pulmonaryfibrosis... ...35 7. Working hypothesis ........................................................................ 36 8. Significance .................................................................................. 37 CHAPTER 2: HY POTHESES AND SPECIFIC AIMS .................................. 38 CHAPTER 3: BLEOMYCIN-INDUCED APOPTOSIS OF ALVEOLAR EPITHELIAL CELLS REQUIRES ANGIOTENSIN SYNTHESIS DE NOV0.....41 Abstract ....................................................................................... 42 Introduction .................................................................................... 43 Methods ........................................................................................ 45 Results ......................................................................................... 50 Discussion ..................................................................................... 61 CHAPTER 4: ESSENTIAL ROLE FOR CATHEPSIN D IN BLEOMYCIN- INDUCED APOPTOSIS OF ALVEOLAR EPITHELIAL CELLS .................... 65 Abstract. ..................................................................................... 66 Introduction .................................................................................. 68 ix MCIhO RCSUII: Discus CHAPTER 1 BLEOMYCI Abstract lntroduc Methodt Results. _ Disc U551} CHXPTER 6: FIBROSIS m Absmq . IHIIOdUCIlk Methods. .. Reguhs‘ . . . DiSCUSSlrm. CHAPTER 7: ( J Methods ....................................................................................... 7 1 Results ......................................................................................... 75 Discussion .................................................................................... 86 CHAPTER 5: ESSENTIAL ROLES FOR ANGIOTENSIN RECEPTOR ATla in BLEOMYCIN-INDUCED APOPTOSIS AND LUNG F IBROSIS IN MICE ........ 91 Abstract ........................................................................................ 92 Introduction .................................................................................... 93 Methods ........................................................................................ 95 Results ........................................................................................ 100 Discussion ................................................................................... 11 1 CHAPTER 6: ATTENUATION OF BLEOMYCIN-INDUCED PULMONARY FIBROSIS BY INTRATRACHEAL ADMINISTRATION OF ANTISENSE OLIGO-NUCLEOTIDES AGAINST ANGIOTENSINOGEN mRN A ............... 116 Abstract. ..................................................................................... 117 Introduction .................................................................................. 1 19 Methods ....................................................................................... 122 Results ........................................................................................ 129 Discussion .................................................................................... 147 CHAPTER 7: GENERAL DISCUSSION AND CONCLUSIONS ..................... 152 Lun At 1.4 A] 1.5 P0 1. Angiotensin Receptor ATl mediates Bleomycin-Induced Apoptosis and Lung Fibrosis ........................................................................... 152 1.1 An Essential Role by Angiotensin Receptor ATl in Bleomycin-Induced Apoptosis in Alveolar Epithelial Cells (AECs): ................................ 152 1.2 An Essential Role of Angiotensin Receptor AT] in Bleomycin—induced Apoptosis and Fibrosis in Lung Explants: ........................................ 153 1.3 In vivo Inhibition of Apoptosis and Collagen Deposition by an ATI Antagonist and AT] Gene Deletion: .............................................. 154 1.4 AT1 receptor blockade block fibrosis via suppression of apoptosis ........... 155 1.5 Potential intracellular mechanisms underlying ATI mediated apoptosis. 157 2. Essential role for cathepsin D in bleomycin-induced apoptosis of alveolar epithelial cells ............................................................................ 158 2.1 CatD activity is upregulated in AECs apoptosis response to bleomycin and in fibrotic lungs: ......................................................................... 159 2.2 Bleomycin-induced apoptosis of AECs is reduced by blockade of CatD activity or synthesis: ................................................................ 160 2.3 CatD is involved in conversion of angiotensinogen to ANG 1 during bleomycin —induced AECs apoptosis .............................................. 161 3. Essential Roles for Angiotensinogen in Bleomycin-Induced Apoptosis and Lung Fibrosis .......................................................................... 163 xi 4- Ihfles induced Puln 5- Rate-Ii n 5-1 Rer 6- Therapeu. 6l EXIS 6-3 AT] anti 1‘; 6.3 pm“ anllSc: 6.3. / 6.3g) ex 3.1 Essential Roles for Angiotensinogen in Bleomycin-Induced Apoptosis in Alveolar Epithelial Cells: ............................................................ 163 3.2 Essential Roles for Angiotensinogen in Bleomycin-Induced Apoptosis and Lung Fibrosis in explants: ......................................................... 165 3.3 Essential Roles for Angiotensinogen in Bleomycin-lnduced Apoptosis and Lung Fibrosis in vivo: .............................................................. 166 4. Roles of Pulmonary Angiotensin System in Development of Bleomycin- induced Pulmonary Fibrosis ................................................................... 167 5. Rate-limiting step of pulmonary angiotensin II generation in bleomycin— induced pulmonary fibrosis ............................................................. 168 5.1 Renin and/or Cat D .................................................................. 168 5.2 ACE .................................................................................... 169 5.3 AN GEN and angiotensin peptides ................................................. 170 6. Therapeutic Implications ................................................................. 173 6.1 Existing clinical trials ............................................................... 173 6.2 ATl antagonist may be more efficient than ACE inhibitor in term of antifibrotic effect in vivo ............................................................ 176 6.3 Potential therapeutic approaches for IPF: Local administration of ANGEN antisense or AT] antagonist for IPF ............................................... 177 6.3.1 Local administration does not disturb the systemic angiotensin system. 6. 3.2 Local administration is a more effective approach with fewer side effects... ....178 xii 7. General I REFERENCI 7. General Conclusions ........................................................................ 178 REFERENCE ..................................................................................... 179 xiii Figural] Figure 3.1: D: , | CU. “<73 MCI “it? and label C01 0" (Elfin LIST OF FIGURES Figure 1.1 .............................................................................................. 36 Figure 3.1: Detection of apoptosis in primary AECs and A549 cells. A, B and C: Primary cultures of AECs were exposed to vehicle (A) or BLEO (B and C) at 25mU/m1 for 20hr and were then fixed in 70% ethanol without washing (see Methods). Cells exhibiting chromatin condensation and nuclear fragmentation with propidium iodide (arrowheads, B) were scored as described earlier (27 and Methods). C: BLED-exposed cells were fixed and prepared for in situ end labeling (ISEL, left) or TUNEL (right) of fragmented DNA (25); note colocalization of label (blue or brown, respectively) in nuclear fragments (arrowheads) identified by propidium in B. D, E and F: A549 cells exposed to vehicle (D) or BLEO at 25mU/m1 (E) or IOOmU/ml (F) for 20hrs were fixed and prepared for immunolabeling for the active form of Caspase 3 (see Methods). Note labeling of active Caspase 3 (purple) in cells with either normal morphology (arrowheads, E) or in cells with condensed cytoplasm and nucleus (arrows, E and F). Note also reduced total cell number with increasing BLEO doses (E and F) ................................................................ 52 Figure 3.2: Dose-dependent induction of nuclear fragmentation by bleomycin (BLEO) in primary rat AECs and blockade by inhibitors of caspases, endonucleases, ANG converting enzyme (ACE) and ANG-receptor interaction. Rat AECs were isolated and challenged with the indicated concentrations of BLEO on Day 2 of primary culture (see Methods). Putative inhibitors were added 30 xiv “it llii lo.- .\1 Ct in 11: Se minutes prior to addition of BLEO; nuclear fragmentation was scored as described in Fig.1B and Methods. ZVAD = ZVAD-fmk (N-benzylcarboxy- Val-Ala-Asp-[O-Me]—CH2F, 60uM); ATA = aurintn'carboxylic acid IOuM); CAPTO = captopril (SOOng/ml); SARAL = saralasin (50ug/ml). Bars are the mean j; S.E.M. of at least 4 observations; * = p<0.05 versus control (0.0 BLEO) ................................................................................... 54 Figure 3.3: Blockade of bleomycin-induced apoptosis in primary AECs by selective caspase or ANG receptor blockers. Rat AECs were isolated and challenged with 25mU/ml BLEO alone or in the presence of the Caspase 3-selective inhibitor DEVD-fmk (60uM) or the ANG receptor ATl-selective antagonist losartan (LOS, 10'6M). Control cultures (CTL) received BLEO and blocker vehicles only. Nuclear fragmentation was scored as described in Fig.1 and Methods. Bars are the mean i S.E.M. of at least 4 observations; * = p<0.05 versus control ........................................................................... 55 Figure 3.4: Inhibition of bleomycin-induced apoptosis of A549 cells by inhibitors of caspases or ANGII-receptor interaction. A549 cells were cultured to 8% confluence as described in Methods and were challenged with 25mU/ml BLEO in the presence or absence of the indicated compounds. Anti-ANGII = neutralizing antibody to ANGII (lug/m1); N.S.IgG = isotype matched nonimmune immunoglobulin (lug/m1); L158809 = ANG receptor ATl- selective antagonist (10'6 M); PD123319 = ANG receptor AT2-selective XV . Ff . am. on ant W Elite 3.6: Sci SQ antagonist (1045M). Other abbreviations and concentrations are the same as in Figs. 2 and 3. Nuclear fragmentation was scored as described in Fig.1 and Methods. Bars are the mean : S.E.M. of at least 4 observations; * = p<0.05 versus control (0 dose) .................................................................. 56 Figure 3.5: Induction of caspase 3 activity by bleomycin and inhibition by an ANG receptor antagonist. A549 cells were challenged with BLEO (25mU/m1) for 20 hours in the presence and absence of the nonselective ANG receptor antagonist saralasin (SARAL, 50ug/ml). Assay of Caspase 3 was conducted on adherent cells as described in Methods. * = p<0.05 versus control (CTL) and ** = p<0.05 versus BLEO ....................................................... 57 Figure 3.6: Semiquantitative RTPCR of angiotensinogen (ANGEN) mRNA in AECs after bleomycin exposure. Primary cultures of rat AECs (A) and A549 cells (B) were exposed to BLEO (25mU/ml) for the indicated times and total RNA was isolated. RTPCR was performed as described before with primers specific for rat or human angiotensinogen (ANGEN), B-microglobulin (B-MG) or B-actin as control mRNAs ......................................................... 58 Figure 3.7: Blockade of bleomycin induced apoptosis and net cell loss in AECs and A549 cells by antisense oligonucleotides against angiotensinogen mRNA. Primary rat AECs (A) or A549 cells (B) were transfected for 4 hours with antisense or scrambled sequence (scram) oligonucleotides as described earlier (see xvi Figure 4.1: fl L10 Mei an}. E9“ 4.2. Cu (1! Figure 4.1: Methods). Cells were challenged with bleomycin (BLE, 25mU/ml) for 20 hours immediately thereafter, and apoptosis (upper panel) was scored as detailed in Figs.1-3; net cell loss (bottom panel of B) was measured as detailed in Methods. lipo = lipofectamine; see Methods for details. Bars are the mean 1: S.E.M. of at least 4 observations; * = p<0.05 and ** = p<0.01 versus corresponding control (CTL) ................................................ 59 Cleavage of a fluorogenic substrate for Cathepsin D (CatD) is dependent on time and protein concentration. Lysates of primary alveolar epithelial cells (AECs) were incubated with the fluorogenic substrate MOCAc-Gly-Lys-Pro- Ile-Leu-Phe-Phe-Arg—Leu-Lys (an)-D-Arg-NH2, and generation of fluorescent product was monitored continuously over 30 minutes (see Methods for details). Note linearity of product formation with time and amount of ABC lysate ................................................................. 78 Figure 4.2. Bleomycin upregulates CatD activity and release from AECs. Primary cultures of rat alveolar epithelial cells (AECs) were exposed to bleomycin (BLEO) for 20 hours at a concentration known to induce AEC apoptosis (25mU/m1). CatD activity was measured in cell lysates as described in Figure 1, in the presence or absence of the aspartyl protease inhibitor pepstatin A (pepA). Inset: CatD activity was measured in concentrated cell culture medium collected from BLEO-treated (BL) and untreated (C) cells studied in xvii C0 Figure 4.3. l« plt‘. Figure 4.4. Bl cult. (Bl Imt panel A. Bars are the mean :t S.E.M. of n = 6; * = p<0.01 versus untreated control (CTL) by AN OVA and Student-Newman-Keul’s test .................. 79 Figure 4.3. Bleomycin does not alter steady-state levels of CatD mRNA. A: PCR products from two different primer sets (1 and 2, see Methods) used to amplify CatD mRNA by RTPCR; starting material was total RNA isolated from primary rat AECs exposed to BLEO or vehicle for the indicated times. B: Realtime RTPCR of CatD mRNA (primer set 2) at the indicated times after exposure to BLEO (see Methods). B-MG = B-microglobulin; bars are the mean+S.E.M. of three separate AEC cultures ...................................... 80 Figure 4.4. Bleomycin increases the release of immunoreactive CatD protein from cultured AECs. Primary cultures of AECs were exposed to bleomycin (BLEO) as in Figure 2; detergent lysates were harvested from the cells (monolayer), and the cell culture medium was collected and concentrated. Equal amounts of lysate protein (10ug/lane) or volume of culture medium (equivalent to 105 cells) were subjected to western blotting with Cat-D- specific antibodies (see Methods). Note increases in immunoreactive proteins of apparent MW~ 52, 48 and 44kda1 (arrowheads) in medium from BLEO- treated AECs ............................................................................ 81 Figure 4.5. Pepstatin A inhibits bleomycin-induced apoptosis of AECs in vitro. Primary cultures of rat AECs were exposed to BLEO in the presence or absence of xviii m. 30. of b) Figure 4.6. blc. SUI: Flflur , i “'7' Pmdu. A: Ali: With”:- the re. L... pepstatin A (pepA) at 100uM. Apoptosis was quantitated by scoring of nuclear fragmentation with propidium iodide (panel A) or by the enzymatic activity of Caspase 3 (B). See Methods for details. Bars are the mean + S.E.M. of n = 6; * = p<0.01 versus untreated control and ** = p<0.05 versus BLEO by ANOVA and Student-Newman-Keul’s test ................... 82 Figure 4.6. Antisense oligonucleotides reduce CatD immunoreactivity and inhibit Figure 4.7. bleomycin-induced apoptosis of AECs in vitro. A: Antisense (AS) or scrambled-sequence oligonucleotides (SCR) were transfected into primary cultures of rat AECs in the presence of lipofectin (LIPO, see Methods), without challenge with bleomycin (CTL). Western blotting of concentrated cell culture media was performed with CatD—specific antibodies; note decrease in immunoreactive CatD by AS but not SCR oligonucleotides. B: After antisense oligonucleotide transfection as in panel A, AECs were challenged with BLEO (25mU/m1) and harvested for detection of fragmented nuclei as in Figure 5. Bars are the mean + S.E.M. of n = 3; * = p<0.01 versus untreated control (CTL) and ** = p<0.05 versus BLEO by ANOVA and Student-Newman-Keul’s test ......................................................... 83 Production of ANGII from angiotensinogen fragment 1-14 in vitro. A: Angiotensinogen fragment 1-14 (F 1-14, SuM) was incubated in vitro (without cells) with the indicated purified enzymes; ANGII was measured in the reaction buffer by specific ELISA (see Methods for details). Note xix Figure 4.8. Figure 5.1. production of ANGII by the combination of purified CatD + purified angiotensin converting enzyme (ACE), but not by either enzyme alone. B: Primary cultures of AECs were exposed to 5uM F1-14, and ANGII was measured in the serum-free cell culture medium; note production of ANGII by AECs challenged with F1-14, but not by untreated AECs ....................... 84 CatD-dependent induction of ABC apoptosis by angiotensinogen fragment 1- 14. Primary cultures of ABC were incubated with F 1-14 as in Figure 7, in the presence or absence of pepstatin A (pepA, luM); CatD-specific neutralizing antibodies (CatD AB, 1:100) and the ANG receptor antagonists saralasin (SARAL, 50ug/ml) or L158809 (10'6 M). See Methods for details. Bars are the mean + S.E.M. of n = 3; * = p<0.001 versus untreated control (CTL) and ** = p<0.001 versus F 1-14 by ANOVA and Student-Newman-Keul’s test...85 Detection of DNA fragmentation, activation of caspase 3, and alveolar type II pneumocytes in mouse lung. Deparaffinized lung sections were prepared from mice instilled intratracheally 14 days earlier with sterile saline (A, C, and E) or BLEO (B, D, F, and G). The sections were subjectedto ISEL of fragmented DNA (A and B) or IHC with antibodies against the active form of caspase 3 (E—H) or with the type II cell-specific antibody MNF116 (C and D). G: Higher magnification of active caspase 3 labeling. in F. H: Active caspase 3 labeling in mice treated with BLEO and LOS, an AT1 receptor antagonist. Note ISEL and active caspase 3 labeling in cells in the comers of alveolar XX \\ ,\1 all of Figure 5.3. l-li.~. Her. 14 if IEXI -. X20 Figure 5.2. Figure 5.3. walls in the lungs of BLEO-treated mice (B, F, and G, arrowheads) but not in saline-treated mice (A and E) or in mice treated with BLEO and LOS (H). Note also the co-localization of MNF 1 16 (C) with anti-caspase 3 IHC (G) or ISEL (B) in BLEO-treated lungs. D reveals labeling of the type 11 cell marker MNF116 in relatively normal regions of BLEOtreated lung (D, right) but not in more severely affected regions (D, left). See text for details. Original magnifications: X400 (A, B, C, G); X200 (1)); x100 (E, F, H) .................................................................. 103 Histology of mouse lungs at 14 days after instillation of BLEO. A—C: Hematoxylin and eosin preparations of mouse lung instilled intratracheally 14 days earlier with sterile saline (A), BLEO (B), or BLEO and LOS (C). See text and Materials and Methods section for details. Original magnifications, X200 .................................................................................. 105 The ATl-selective receptor antagonist LOS inhibits BLEO-induced activation of caspase 3. A: BLEO was instilled intratracheally into normal mice with and without LOS in the intratracheal instillate (see Materials and Methods). Six hours later, the lungs were perfused to remove blood, excised, and the enzymatic activity of caspase 3 was measured in lung homogenates. B: Lung explants were prepared from normal mouse lung tissue perfused before excision (see Materials and Methods). BLEO (25 mU/ml) was applied in serum-free culture medium for 24 hours in the presence or xxi \‘E Figure 5.4. A act mi abs intr MIN “'35 the 1 Very F1gills-5.5.AT1 days PTL‘sQ tOIal l hbikl m absence of LOS (10_6 mol/L). Bars are the means i SEM of n = 6; *, P < 0.05 versus control (CTL); ** , P < 0.05 versus BLEOiLOS by analysis of variance and Student-Newman-Keul’s test .................................... 106 Figure 5.4. AT1 receptor blockade inhibits DNA fragmentation and caspase 3 activation in lung epithelial cells 14 days after BLEO instillation. Normal mice were given a single intratracheal instillation of BLEO in the presence or absence of LOS in the instillate. LOS also was administered thereafter daily intraperitoneally. Fourteen days later, lung sections were prepared and labeled by ISEL (A) or by IHC for the active form of caspase 3 (B). Labeling was quantitated in cells within the alveolar surfaces (see Figure 1C). Bars are the means i SEM of n = 6; *, P < 0.01 versus control (CTL); **, P < 0.05 versus BLEO by analysis of variance and Student-Newman-Keul’s test. . . .. 107 Figure 5.5. AT1 receptor blockade inhibits lung collagen accmnulation at 14 days after BLEO instillation. Normal mice were administered BLEO in the presence or absence of LOS as described in Figure 3. Fourteen days later, total lung collagen was determined by assay of total hydroxyproline (HP) in hydrolyzed lung tissue (see Materials and Methods). Bars are the means :t SEM of n = 6; *, P < 0.05 versus control (CTL) by analysis of variance and Student-Newman-Keul’s test ....................................................... 108 Figure 5.6. Mice deficient in angiotensin receptor ATla exhibit reduced DNA fragmentation and caspase 3 activation in lung epithelial cells 14 days after xxii \ti. XL“ Figure 5.7. M: 8C0. heir intru h} (In descr 0f the 31110111 yer-ills Keulig BLEO instillation. Normal [wild type (w.t.)] or heterozygous ATla knockout mice (+/-) were administered BLEO intratracheally as in Figure 3. Fourteen days later, lung sections were prepared and labeled by ISEL (A) or by IHC for the active form of caspase 3 (B), which were quantitated as described in Figure 4 and Materials and Methods. Bars are the means :1: SEM of n = 5; *, P < 0.001 versus wild-type unchallenged (w.t. - BLEO); **, P < 0.01 versus wild-type challenged (w.t. + BLEO) by analysis of variance and Student- Newman-Keul’s test .................................................................. 109 Figure 5.7. Mice deficient in angiotensin receptor ATla exhibit reduced lung collagen accumulation in response to BLEO instillation. Normal [wild type (w.t.)] or heterozygous ATla knockout mice (+/-) were administered BLEO intratracheally as in Figure 3 . Fourteen days later lung tissue was fast-frozen, hydrolyzed, and total collagen was measured by hydroxyproline assay (HP) as described in Materials and Methods. A: HP data are expressed as a percentage of the corresponding control (- BLEO). B: Data are expressed as the absolute amount of HP per left lung. Bars are the means i SEM of n = 5; *, P < 0.01 versus untreated (- BLEO) by analysis of variance and Student-Newman- Keul’s test ........................................................................... 110 Figure 6.1: Semiquantitative RT-PCR of angiotensinogen (ANGEN) mRNA in lungs 3hours afier Bleo exposure. Normal rats were intraltracheeally instilled with Bleo (8U/Kg) or saline (CTL) for 3hours and total RNA was isolated. RT- xxiii Figure 6.3; T} and 0f fun at}. It... (I Coj PCR was performed as described before (see Meterial and Methods) with primers specific for rat ANGEN, -Microglobulin (B-MG) as control mRNAs ................................................................................ 134 Figure 6.2: In situ hybridization of ANGEN mRNA demonstrated that positive labeling (dark purple, shown by arrowhead) increased 6h (B x 200 magnification, C x 400 magnification) and 14 days (E, Fx200 magnification) after BLEO instillation compared to the corresponding controls (A, D x 200 magnification). 6h after BLEO instillation, the positive labeling is mainly localized at the comers of the alveolar, which typically are the positions for type II alveolar cells (BC). 14 days afier BLEO instillation, the positive labeling is mainly localized at fibrotic foci (E) where myofibroblast / fibroblast accumulate and the corners of the alveolar (F) near the fibrotic region .................................................................................. 13 5 Figure 6.3: The sections were subjected to IHC with antibodies against the ANGI, lectin and a-SMA 24 hours (B, C) and 7 days (D, E, F) after intratracheal instillation of bleomycin. 24h after BLEO instillation ANGEN /ANG I (purple) was found in alveolar walls cell at the corners that did not label with lectin (compare B &C x400, same filed with matched arrowhead). In severely affected areas of the parenchyma of bleomycin— induced fibrotic rat lungs, regions of ANGEN / ANG I labeling (Dx200, black box) were observed to coincide with a loss of lectin labeling (Ex200, white box: double label of same xxiv Figure 6.4 P lit 'Jv 'J Stu Figure 6.5 H indL lun‘; Figure 6.6 The ‘\ lUng L section as D). Enlargement of the white-boxed region (F x400) revealed a- SMA immunoreactivity in the middle of the box on the adjacent serial section, suggestive of myofibroblasts ........................................................ 136 Figure 6.4 Pulmonary angiotensin II (ANG II) increased in bleo-induced lung injury. 6 hours and 14 days after bleo instillation, lungs were perfused, homogenized, and analyzed by ELISA specific for ANG II. Values are means of at least 5 separate determinations. * Significantly different from CTL, P < 0.05 (by Student's t-test) ....................................................................... 137 Figure 6.5 The Antisense oligonucleotides against ANGEN mRNA inhibit BLEO- induced activation of caspase 3. Lung explants were prepared from normal rat lung tissue perfused before excision (see Materials and Methods). BLEO (25 mU/ml) was applied in serum-free culture medium for 24 hours in the presence or absence of antisense (40nM)). Bars are the means 3: SEM of n = 3; *, P < 0.05 versus control (CTL+lipo) by analysis of variance and Student-Newman-Keul’s test ........................................................ 138 Figure 6.6 The Antisense oligonucleotides against AN GEN mRNA and non-selective AT receptor antagonist saralasin inhibit BLEO-induced collagen accumulation in lung explants. Lung explants were prepared from normal mouse lung tissue perfused before excision (see Materials and Methods). BLEO (25 mU/ml) was applied in 10% FBS (A) or 1% ITS (B) for 14 days in the presence or absence XXV Figure 6.7 l: 01 di int nor Hum68hw ;\\t 0ng Pane fluor fiekl rereu ofau Panel finenx “llll l; of SAR (50ug/ml) or antisense (40nM). Bars are the means i SEM of n = 3; *, P < 0.05 versus control (CTL) in A; *, P < 0.01 versus CTL in B by analysis of variance and Student-Newman-Keul’s test .................................... 139 Figure 6.7 Distribution of fluorescence in lung, liver and kidney 2 hous after instillation of BODIPY labeled oligonucleotides. Normal rats were instilled with different dose of BODIPY labeled oligonucleotides. Measure fluorescence intensity in the homogenates of the frozen lung, liver and kidneys after normalization to total tissue protein ............................................... 140 Figure 6.8 Localization of the intratracheal instilled fluoscence—labelled antisense against ANGEN mRNA on the lung sections from rats treated with 75ug of oligonucleotides. Panels with red fluorescence show Pl staining and panels with green fluorescence show BODIPY staining. Panel A, B (10 x10) show the same field from the same section. And so do panel C, D (40 x 10). Panel A, B revealed that BODIPY fluorescence was localized primarily to the epithelium of airways (arrow) and in isolated cells of the alveolar walls (arrowhead). Panel C, D revealed that some alveolar wall cells were stained with high intensity of BODIPY-oligonucleotides (arrows) while others were stained with very little of the oligonucleotides (arrowheads) ........................... 141 xxvi figure 6'9 " 5U," ins: f0 1’ den igure 6.1 l. Dete blots indie. treatr‘. fragn; bleorr. caspm Figure 6.9 A: Macroscopic photographs of the lungs 14 days after different treatment. Note the almost normal appearance of the lung in the Bleo +AS treated rat compared with the 8160- and bleo+SCR treated rat lungs, which are smaller with many patchy bleeding spots. B: Quantitation of total lung collagen by hydroxyproline (HP) assay. At 14 days post-Bleo, collagen was quantitated by HP assay applied to hydrolyzed lung tissue. See METHODS for details. * P < 0.05 vs. CTL by ANOVA ..................................................... 142 Figure 6.10 Bleomycin increased angiotensinogen protein expression, which was Figure 6.11. suppressed by the treatment of antisense in perfused lung tissues. 14 days after instillation of bleomycin or vechicle, lungs were perfused and homogenized for the western blot analysis. (A) Representative immunoblots and (B) densitometn'c evaluation of blot data. Data are presented as mean isSE (n=5). *: P<0.05 versus CTL ............................................................... 143 Detection of active caspase-3 by immunohistochemistry (IHC) and western blots (WB) 14 days after described treatment. Male Wistar rats received indicated treatment intratracheally. Lung tissues were harvested 14 days after treatment and subjected to IHC and WB by using anti-active caspsase-3 (p17 fragment) antibody. Upper penal (WB): Active caspase-3 was induced by bleomycin and suppressed by antisense treatment. Lower penal (IHC): active caspase —3 was localized in alveolar epithelial cells and alveolar macrophages. ......................................................................................... 144 xxvii Figure 6.12 Pt 0b the pos— did Fig. 6.13 The flaw “ere 3min “CFC the u. C(lnlli '- ‘o n n o u 1.. '- u 'o ‘. Figure 6.12. Detection of apoptotic cells by in situ end labeling (ISEL) of fragmented Fig. 6.13 Fig- 7.1. Fig. 7.2. DNA 14 days after described treatment. Male Wistar rats received indicated treatment intratracheally; lung tissues were harvested 14 days after treatment and subjected to ISEL coupled to a fast blue detection system (see METHODS). Positive reaction is blue. A: in CTL group, ISEL-positive nuclei were not observed. B: in bleo group, ISEL-positive nuclei were observed in cells within the alveolar walls, many ISEL-positive nuclei were observed in septal wall cells at the alveolar corners. C: administration of antisense decreased ISEL positive cells in response to Bleo. D: administration of scramble nucleotides did not decreased ISEL positive cells in response to Bleo ...................... 145 The Antisense oligonucleotides against ANGEN mRNA inhibited DNA fragmentation in lung epithelial cells 14 days after BLEO instillation. Rats were given a single intratracheal instillation of BLEO in the presence antisense or scramble ONT in the instillate. Fourteen days later, lung sections were prepared and labeled by ISEL. Labeling was quantitated in cells within the alveolar surfaces. Bars are the means :4: SEM of n = 5; *, P < 0.01 versus control (CTL); **, P < 0.01 versus BLEO or BLEO +SCR by analysis of variance and Student-Newman-Keul’s test ....................................... 146 ............................................................................................ 167 ............................................................................................ 178 xxviii ACE AC Eis AECS ANGEN ANT} ANGI ANG II APR u-SMA AS ATA AT 1 cell AT ll cell All Reeept0r Arr BALF blo‘dl'Tp BLEO BPAEC CAMP CAPT ACE ACEis AECs ANGEN ANG ANG I ANG II a-SMA AS ATA AT I cell AT 11 cell ATl Receptor ATF BALF bio-dUTP BLEO BPAEC cAMP CAPT LIST OF ABBREVIATIONS Angiotensin Converting Enzyme Inhibitors of ANG converting enzyme Alveolar Epithelial Cells Angiotensinogen Angiotensin angiotensin I angiotensin II Acute Phase Response Element Alpha-Smooth Muscle Actin antisense oligonucleotides aurintricarboxylic acid Alveolar type I cell Alveolar type II cell ANG 11 type I receptor Activating transcription factor bronchoalveolar lavage fluid biotinylated deoxyuridine trisphosphate bleomycin Bovine pulmonary artery endothelial cells Cyclic AMP captopril xxix CmD CMV CID-PF DAG dgdCIP DLco EBV ECL ECNI HJSA REL FBS FL14 HGF HP HRCT CatD CMV CTD-PF DAG dig-dUTP DLco EBV ECL ECM ELISA Fas L FBS F l - l 4 HGF HP HRCT IHC IL-6 ILD 1P3 IPF ISEL ISH cathepsin D cytomegalovirus Connective tissue disease-pulmonary fibrosis diacylglycerol digoxigenin-labeled deoxyuridine trisphosphate Diffusion capacity of carbon monoxide Epstein-Barr virus Enhanced chemiluminescence extracellular matrix Enzyme-Linked Irnmunosorbent Assay Fas ligand Fetal bovine serum angiotensino gen fragment 1 -14 Hepatocyte growth factor hydroxyproline High-resolution CT Immunohistochemistry Interleukin-6 Interstitial lung disease Inositol- l , 4, 5-trisphosphate Idiopathic Pulmonary Fibrosis In situ end labeling In Situ Hybridization XXX 1.1. [TS KGF IIPO LOS 110 Km 9 MG PAls PBS PepA PDGF pr PGE; Pl P1P: PM PRC 1’sz PIC B PLD PFOSP-C Rag SARAL SCR I.T. ITS KGF LIPO LOS LTo. Km B -MG PAIs PBS PepA PDGF PF PGEz PI PIP2 PKA PKC PLA2 PLC B PLD proSP-C SARAL SCR intratracheal Insulin-transferrin-selenium Keratinocyte growth factor lipofectin losartan lymphotoxin-o. Michaelis Constant [3 -Microglobulin plasminogen activator inhibitors Phosphate-buffered saline pepstatin A Platelet-derived growth factor Pulmonary Fibrosis Prostaglandin E2 propidium iodide Phosphatidylinositol-4, 5-bisphosphate Protein kinase A Protein kinase C phospholipase A2 phospholipase C-beta phospholipase D pro-surfactant protein-C renin-angiotensin system saralasin Scrambled-sequence control oligonucleotides xxxi SNPS SP-A TBS 101.5 INF-a TNF-Rll tPA TLNEL L'PA wr. ZVADfmlr SNPs SP-A UPA w.t. ZVADfrnk Single nucleotide polymorphisms Surfactant protein A Tris-buffered saline Transforming growth factor-beta Tumor necrosis factor alpha TNF-a/LTo. receptor 2 tissue-type plasminogen activator Terminal deoxynucleotidyl transferase (TdT) mediated deoxyuridine End Labeling urokinase-type plasminogen activator wild type N—benzylcarboxy-Val-Ala-Asp-fluoromethylketone xxxii GENERAL 1. Idiop Idiopathic pt; disease result to thickening unknoun etio reepitheliulim matrix. and d; 1.1 Epidem IPF is a relatixr Chapter 1 GENERAL INTRODUCTION 1. Idiopathic Pulmonary Fibrosis Idiopathic pulmonary fibrosis (IPF) is a progressive, usually fatal, form of interstitial lung disease resulting from injury to the lung and an ensuing fibrotic response. Fibrosis leads to thickening of the alveolar walls and the obliteration of the alveolar space with unknown etiology (Fonseca et al., 2000). The characteristics of IPF are failure of alveolar reepithelialization, persistence of fibroblasts/myofibroblasts, deposition of extracellular matrix- and distortion of lung architecture, which ultimately results in respiratory failure. 1.1 Epidemiology of IPF IPF is a relatively rare disease and more common in males than in females. The estimated annual incidence is 7 cases per 100,000 women and 10 cases per 100,000 men (Coultas et al., 1994). The incidence, prevalence, and death rate increase with age (Coultas et al., 1994; Mannino et al., 1996; Schwartz et al., 1994). For the age group between 35 to 44 Years there are 2.7 cases of IPF in a population of 100,000. For the age group older than 75 years there are about 175 cases in the same population (Coultas et al., 1994). Two thirds of patients diagnosed with IPF are with an age greater than 60 years (Johnston et al., 1997). The mean age at diagnosis of IPF is 66 years (Johnston et al., 1997; Carrington et al., 1978). 1.2 Pre Some studi solvents. or Hubbard et 1.2.1 Genetic factr the IPF patiei genes in thos. gene. which 1 protein (Thou mum“! Protein is ablmfmal (“I Produces prot. cornpanmenI tc abnormal SUrfa Geflellc 1' associated with ~ ”- 3001) Ar . | ACE COnferS a h 1.2 Predisposing factors for IPF Some studies indicate that genetic factors and environmental exposure to dusts, organic solvents, or urban pollution increases the risk of developing IPF (Johnston et al., 1997; Hubbard et al., 1996; Iwai et al., 1994). 1.2.1 Genetic factors Genetic factors may play an important role in the pathogenesis of IPF and about 3% of the IPF patients aggregated in families (White et al., 2003). Studies seeking the abnormal genes in those patients demonstrated a mutation in the pro-surfactant protein-C (proSP-C) gene, which lead to the substitution of leucine by glutamine in the C-terminus of that protein (Thomas et al., 2000). Electron microscopic studies demonstrated that this mutant protein is aberrantly located in the cell and lamellar bodies containing surfactants is abnormal (Thomas et al., 2000). These results suggest that the mutation of the gene produces protein that could not be properly processed to the correct subcellular compartment to be secreted out of type II alveolar epithelial cells. The mutation results in abnormal surfactant production in alveolar type II cells. These findings suggest that in this familial IPF, improper cellular processing of proSP-C may contribute to pulmonary fibrosis. Genetic polymorphisms of several other enzymes and cytokines have been associated with either the incidence or progression of IPF (Whyte et al., 2000; Pantelidis et al., 2001). Angiotensin converting enzyme (ACE) gene polymorphisms were studied in pulmonary fibrosis. The D allele of the insertion] deletion (I/D allele) polymorphism of ACE confers a higher level of ACE gene expression compared to I allele. The incidence ofD allele 1‘ moderate tr incidence o approximate et al.. 2001 ). angiotensin fibrosis (Mod Single nuclec in 74 IPF pat 2001). Four c I.Vmphoto.\rin. ”W- No dit found bemee frequency of Increased in asSOCiation “a Capach." for ck, diseaSe ’ Progrct dimmed prod. 1.2_2 E” _ 1,1 of D allele and D/D genotype of ACE gene in 24 patients with interstitial pneumonia and moderate to severe pulmonary fibrosis was examined (Morrison et al., 2001). The incidence of the D allele was approximately 15% higher and the D/D genotype was approximately 11% higher in the patients group than in the general population (Morrison et al., 2001). That study suggests that gene polymorphisms that confer higher levels of angiotensin converting enzyme (ACE) gene expression predispose patients to lung fibrosis (Morrison et al., 2001). Single nucleotide polymorphisms (SNPs) of some pro-inflammatory genes was evaluated in 74 IPF patients with confirmed diagnosis by clinical or biopsy data (Pantelidis et al., 2001). Four candidate genes were proposed including tumour necrosis factor-a. (TNF-a), lymphotoxin-a. (LTa), high affinity TNF-a/LTa. receptor 2 (TNF-RII), and interleukin-6 (IL-6). No difference in genotype, allele, or haplotype frequencies of those genes was found between patients inflicted with IPF and control population. However, the frequency of carriage of the IL-6 allele (intron 4G) and the TNF-RII allele (1690C) increased in patients, which was not observed in controls. In addition, a strong association was found between progression of IPF and IL-6 genotype since diffusion capacity for carbon monoxide (DLCO) was decreased, suggesting that the rapidity of disease progression is higher in patients with IL-6 genotype, this may account for decreased production of IL-6 (Pantelidis et al., 2001). 1.2.2 Environmental factors: Environmental factors have also been considered to contribute to the development of pulmonary fibrosis. Several VII’L implicated i detected in demonstrate 1999;1(elly Investigators occupational I . >3 CllniCal 1 The - Clin' . 10211 m._ 1031': FilaIOI-y Crag; Several viruses including Epstein-Barr virus (EBV), cytomegalovirus (CMV) have been implicated in pathogenesis of IPF as those viruses and latent viral infections have been detected in patients with IPF. However, there exists no convincing evidence to demonstrate certain viruses directly cause IPF (Yonemaru et al, 1997; Stewart et al., 1999; Kelly et al., 2002). Investigators have also made some extent of associations between unidentified occupational and dust exposures and IPF (Baumgartner et al., 2000; Mullen et al., 1998). Cigarette smoking is the most extensively studied environmental risk factor for IPF. Howerve, instead of showing smoking as a risk factor for IPF, some clinical studies indicate that current smokers have improved mortality in established cases of IPF (King et al., 2001; Flaherty et al., 2002). In agreement with those studies, those IPF patients who currently smoke also had lower granulation/connective tissue (FF) scores, another independent predictor of disease survival, indicating better prognosis (Flaherty et al., 2003; King et al., 2001). These observations thus suggest that some factors produced in the process of cigarette smoking may be helpful for treating IPF (N obukuni et al., 2002). Genetic susceptibility and environmental factors may contribute to the development of IPF, yet evidence of predisposing or etiologic factors is not strong since most patients do not demonstrate any obvious risk factors (Selman et al., 2001). 1.3 Clinical features The clinical manifestations include dyspnea on exertion, nonproductive cough, and inspiratory crackles; at the late stage of this disease patients also present shortness of breath at rest (Crystal et al., 1976). Bi-basilar and end-expiratory rales are demonstrated through physical examination in more than 80% of patients with IPF (ATS, 2000). Digital clubbing is noted in up to half of all patients (Johnston et al., 1997). Cyanosis and signs of pulmonary hypertension may be seen in patients at the late stage of IPF (Panos et al, 1999). There is no laboratory test specific for the diagnosis of IPF. Laboratory evaluation of patients with suspected IPF is primarily to rule out alternative causes of interstitial lung disease such as sarcoidosis or connective tissue disease—pulmonary fibrosis (CTD-PF). Pulmonary function tests reveal restrictive impairment, reduced diffusing capacity for carbon monoxide, and arterial hypoxemia exaggerated or elicited by exercise (Selman et aL,2001) More than 90% of patients with IPF at the time of diagnosis had abnormal chest radiographs (Johnston et al., 1997). The characteristic pattern of abnormal chest radiography is diffuse bilateral interstitial or reticulonodular infiltrates, predominantly in the basilar and subpleural regions of the lower lung (Guerry-Force et al., 1987). Diagnosis for patients with suspected IPF can be improved by using the high-resolution CT (HRCT) scanning since HRCT can produce reconstructed images of thin scan sections (1-2 mm slices) in three-dimensions to visualize the enhanced spatial structure of the lung parenchyma (White et al., 2003). Patterns of HRCT images typically seen in IPF patients include coarse reticular or linear opacities indicating intralobular and interlobular septal thickening, a predilection for the periphery and lower lobes of the lungs, honeycomb cysts, and traction bronchiectasis (Kazerooni et al., 1997). Ground glass opacities and ill-defined hazy zones that represent active alveolitis or fibrosis of the intralobular and alveolar septae can also be present (Kazerooni et al., 1997; Wells et al., 1993). Extensive honeycombing, septal thickening, and a lack of ground glass opacities often predict a poor prognosis. The definite diagnosis of IPF must meet the following criteria: 1) a compatible clinical history; 2) the exclusion of other known causes of interstitial lung disease such as drug injuries, environmental exposures, or collagen vascular disease; 3) a surgical lung biopsy showing usual interstitial pneumonia histologic pattern (ATS, 2000). The histologic hallmark of usual interstitial pneumonia is a heterogeneous appearance with alternating areas of dense fibrosis, fibroblastic foci, interstitial inflammation, and honeycomb change and normal lung at low magnification. 1.4 Histopathology In IPF the normal architecture and functional integrity of the lung are destroyed. The main histological features of the fibrotic lung are persistent and unrepaired epithelial damage, proliferation and accumulation of fibroblast/myofibroblast cells, and increased collagen deposition (Selman et al., 2001). Temporal heterogeneity is a histopathologic hallmark of IPF. At low magnification, a typical heterogeneous appearance can be observed with normal areas alternating with areas with peripheral fibrosis, interstitial inflammation, and honeycomb changes (ATS, 2000). The inflammatory component is typically not intensive and consists primarily of lymphocytes and plasma cells. Other inflammatory cells such as neutrophils and eosinophils may be present with low abundance. Under high-power magnification, fibroblastic foci can be found at the border between fibrotic and normal lung where rationales”i collagen bunt for IPF as it tibroblastic fr 2003:1(ing et Another imp- Alveolar epitl active fibrosis airspaces linec 1.5 Thera p The traditiona proudes the b treat IPF (Flah ImmUDOSUPpr-e 300t . ”ll. a-Lalhlu L'“ffmunatelx- 0111} Offer a m pinenjdone int a; . Id Or I \'i\'( l H0"met. fibroblasts/myofibroblasts accumulate with collection of dense, relatively acellular collagen bundles. The presence of fibroblastic foci may be an important prognostic factor for IPF as it represents the active lesion of IPF (Selman et al., 2001). The number of fibroblastic foci is correlated with the morality of patients with IPF (Nicholson et al., 2002; King et al., 2001). Another important characteristic of IPF is unrepaired alveolar epithelial damage. Alveolar epithelial injury with abnormal type II pneumocytes is often seen at areas of active fibrosis (Kuhn et al., 1991). Honeycomb changes are formed by enlarged, cystic airspaces lined by abnormal type II pneumocytes. 1.5 Therapeutic options for pulmonary fibrosis The traditional view considers pulmonary fibrosis is an inflammatory disorder, which provides the basis for using potent anti-inflammatory agents such as corticosteroids to treat IPF (Flaherty et al., 2001). Anti-inflammatory agents were often supplemented with immunosuppressive and cytotoxic agents such as cyclophosphamide (Zisman et al., 2000), azathioprine (Raghu et al., 1991) and colchicines (Douglas et al., 1998). Unfortunately, clinical studies indicate that those traditional therapeutic approaches can only offer a marginal benefit at best (King et al., 2000). In contrast, new approaches employing anti-fibrotic agents such as angiotensin system antagonists (captopril), pirfenidone, interferon-Y and statin have been shown to have antifibrotic effects in vitro and/or in vivo in experimental pulmonary fibrosis models (Selman et al., 2001). However, prospective, randomized, placebo-controlled multi-center clinical trials are needed to evaluate their efficacy in the treatment of IPF. 2. Bleomycin-Induced Experimental Pulmonary Fibrosis Model: A number of animal models of pulmonary fibrosis have been developed to address the pathogenesis of pulmonary fibrosis. One of them is the bleomycin-induced experimental pulmonary fibrosis model. [see review (Thrall and Scalise, 1995)]. Bleomycin is an antineoplastic antibiotic, which was isolated from a strain of Streptomyces verticillus (Umezawa et al., 1966). Bleomycin is not a single peptide. Instead, it consists of a family of complex glycopeptides with different amine groups (Umezawa et al., 1966, 1967; Umezawa, 1973, 1974). It is used in the treatment of squamous cell carcinomas and various lymphomas (Ichikawa et al., 1967, 1969; Yagoda et al., 1972; Blum et al., 1973). The mechanisms of antineoplastic action of bleomycin are complex and cell cycle-dependent. Briefly, bleomycin intercalates between DNA base pairs, causing DNA to unwind and impairing protein synthesis. Bleomycin increases radical oxygen species by reducing molecular oxygen to superoxide and hydroxyl radicals. Those free radicals then attack DNA and cause strand cleavage (Sausville et al., 1978) Lack of side effects on bone marrow (Kimura et al., 1972; Boggs et al, 1974) and immunocompetence (Dlugi et al., 1974; Lahane et al., 1975) are the major advantages of using bleomycin over other chemotherapeutic agents. However, the lung and skin are the major organ systems affected by the toxic side effects of bleomycin (Thrall and Scalise, 1995). The fact that these organ systems are susceptible to bleomycin can be explained in part, but not entirely, by the pharmacokinetics of bleomycin. Bleomycin reaches the highest concentration in the lung and skin afier parenteral administration. This is likely due to the lack of a hydrolase, which inactivates bleomycin, in the lung and skin as bleomycin-induced cytotoxicity is higher with low cellular levels of hydrolase activity (Umezawa et al., 1972). Currently the bleomycin induced pulmonary fibrosis animal model is the most commonly used model for studying the pathogenesis of fibrotic lung disease. The bleomycin—induced pulmonary fibrosis is dose and administration route dependent. Two most common administration routes are parenteral and intratracheal. Cumulative doses of 100-200 mg (units)/kg were used in the parenteral injection model whereas the intratracheal single-injection model uses doses in the range of 2.5-7.5 mg/kg (Thrall and Scalise, 1995). The parenteral injection produces lesions with a more diffuse pattern in perivascular and subpleural locations whereas the intratracheal injection produces patchy focal lesions in peribronchial locations. Various animals, including mice and rats, have been used for models of bleomycin induced pulmonary fibrosis (Thrall and Scalise, 1995) Generally speaking, the development of lung injury in various bleomycin animal models is quite similar regardless of species and route of administration. The lung injury progresses through three stages (Thrall and Scalise, 1995): (l) The acute inflammatory stage (1-3 days post-intratracheal instillation of bleomycin and later in the parenteral models). This stage features the activation of inflammatory mediator systems and emerging of pulmonary edema. (2) The subacute stage (4-21 days post intratracheal instillation of bleomycin). This stage is characterized by the synthesis of collagen and elevation of the levels of net lung collagen as well as other connective tissue components. (3) The chrC metabolism OI 111 Summary: carcinomas an confers the ”7 however. pr 0V1 m'th direct clin 3. Mechanisms IPF is the me physicians (M; pathogenesis. tl approaches u: iUltrrunosuppres. Recent NHLBI PthOgenesis of‘ (3) The chronic stage (21 days to termination). This stage is dominated by the metabolism of connective tissue towards reepithelialization. In summary, as an effective antineoplastic agent used in the treatment of various carcinomas and lymphomas, bleomycin has toxic side effects on lung tissues, which confers the major limitation to its therapeutic use. The bleomycin animal models, however, provide a valuable tool to investigate the pathogenesis of fibrotic lung disease with direct clinical relevance. 3. Mechanisms of F ibrogenesis IPF is the most common interstitial lung disease (ILD) encountered by respiratory physicians (Mapel et al., 1998). Unfortunately, despite years of research on its pathogenesis, the treatment of PF has not been successful. The traditional therapeutic approaches using potent anti-inflammatory agents supplemented with an immunosuppressive agent could only offer a marginal benefit at best (King et al., 2000). Recent NHLBI Workshops have concluded that our current understanding of the pathogenesis of the IPF is incomplete (Mason et al., 1999; Crystal et al., 2002). Current opinions regarding the pathogenesis of PF are controversial. The traditional view considers pulmonary fibrosis as an inflammatory disorder, in which inflammation in the lower respiratory tract (alveolitis) is responsible for derangements of the alveoli and results in scaring of the lung parenchyma (Gallin et al., 1992). The consequence of the inflammation is the loss of fimctional alveolar—capillary unit, the accumulation of collagen, and the formation of honeycomb lung. The limited success of anti-inflammz alternative by Current evolv failure of reer h}pothesis me and efficient e 1990,). There . this hjpothesis facilitates sub Imeent fibroti 1998). 3-1 The rof anti-inflammatory/ immunosuppressive therapy for IPF has stimulated the generation of alternative hypothesis regarding the pathogenesis of IPF (Crystal et al., 2002). Current evolving hypothesis is that IPF results from the epithelial microfoci injury and a failure of reepithelization (Selman et al., 2001). Haschek and Witschi first proposed this hypothesis more than 20 years ago, speculating that epithelial damage drives fibrogenesis and efficient epithelialization would prevent fibrogenesis (Haschek et al., 1979; Witschi, 1990). There is some evidence from both animal model and human patients that supports this hypothesis. In the animal models, experimental delay of epithelial repair after injury facilitates subsequent fibrogenesis (Haschek et al., 1979). In human lung biopsies, nascent fibrotic foci were colocalized with unrepaired or abnormal epithelia (Uhal et al., 1998). 3.1 The roles of alveolar epithelial cells (AECs) The lung is the respiratory organ that functions to maintain the constant internal environment by inspiring oxygen into blood circulation and removing carbon dioxide from blood circulation. To fulfill this function, the lung has special architecture. The basic functional unit of the lung is alveoli where gas exchange occurs between the air and the circulating blood. Alveoli consists of several components: 1) the alveolar epithelium that covers the air space; 2) the endothelium lining the capillaries; 3) thin interstitium separating the epithelium and the endothelium, which includes the connective tissue and extracellular matrix components with a few resident fibroblasts (Fonseca et al., 2000). The intact alveolar epithelium is composed of two cell types, alveolar type 1 cells (AT I cell) and type 11 cells (AT 11 cell) (Uhal et al., 1997; Sutherland et al., 2001). They ll are morphologically and functionally different although the cell numbers of these two types are similar (Mason and Williams, 1991). AT 1 cells are large elongated cells and are terminally differentiated. They cover over 90% of the alveolar surface. The thin and attenuated cytoplasm of the AT 1 cell facilitates the gas exchange by minimizing the diffusion distance between alveolar gas and blood. These cells are metabolically active and harbor cell surface receptors for a variety of substances, including extracellular matrix (ECM) proteins, growth factors, and cytokines. AT 11 cells are cuboidal in shape with rounded nuclei. They are predominantly located in the comers of the alveoli and cover about 10% of the alveolar surface. Type 11 cells have distinctive features such as apical microvilli and cytoplasmic lamellar bodies that contain the alveolar lining material surfactant. Type II cells have abundant intracellular organelles including extensive endoplasmic reticula, Golgi complexes, peroxisomes, and mitochondria. AT II cells are not terminally differentiated and have a number of unique functions (Sutherland et al., 2001). For example, AT 11 cells can synthesize and secrete surfactant that is involved in regulating the surface tension in the alveolar epithelium. AT 11 cells are also involved in xenobiotic metabolism by regulating the activities of the cytochrome P-450 monooxygenase, transepithelial ion and H20 movement. In addition, AT 11 cells have immuno-modulatory functions and regulate the lung extracellular matrix (ECM) metabolism (Pardo et al., 1997). Another important function of the AT II cells is the maintenance of the alveolar epithelium. It can function as stem cells to proliferate to give rise to new AT 11 cells or differentiate into AT I cells. As such, AT 11 cells play important role in the repairing of the alveolar epithelium by replacing the lost cells and restoring normal tissue architecture and lung function (Uhal, 1997). 12 3.2 Cons' Loss of AEC PathoPhFSiOI‘ 1211 lite mallOr fur alveolar air at which facilitz endothelial CC Space. In 0rdc prevented frOfT this function. I alveolar epilhc benveen adj ace epithelial cells ltrnphatic s_vstc elllut of plastr. 3.2 Consequence of alveolar epithelial cells damage/ injury: Loss of AECs during IPF has a number of adverse effects, which likely contribute to the pathophysiology of IPF [see review (Simon, 1995)]. 3.2.1 Loss of permeability barrier The major function of type I alveolar cells is gas exchange. Diffusion distances between alveolar air and capillary blood are very short due to the thin shape of the type I cells, which facilitates gas exchange. Intercellular junctions between adjacent capillary endothelial cells are not tight. Therefore, plasma proteins can leak into the interstitial space. In order to maintain the functional integrity of the lung, the plasma has to be prevented from leaking into the airspaces. The alveolar wall provides a barrier that fulfills this function. This barrier of the alveolar wall consists of the capillary endothelium and alveolar epithelium (Gorin and Stewart, 1979). Compared to the intercellular junctions between adjacent capillary endothelial cells, the intercellular junctions between alveolar epithelial cells are much tighter which prevent air space from flooding. The pulmonary lymphatic system also helps to prevent air space fiom flooding by draining the normal efflux of plasma components back to the circulation. However, in patients with IPF, plasma proteins, e. g., fibrinogen, leak into the alveolar space (Basset et al., 1986; Kuhn et al., 1989; Crouch, 1990). 3.2.2 Loss of the barrier limiting fibroblast migration into the alveolar airspace Migration of fibroblasts into the airspaces would lead to collagen deposition and obliteration of alveoli. In addition to providing a barrier for plasma proteins leakage, the 13 aheohri Rat trachr epithelial m'thout e becomes 1 dssue. Vl' fibrosis d preventing fibrosis. Ft ls a meat 198:; Lani Image ("8.1 (Borok et a Sl'mhcslze ] alveolar epithelium also prevents the migration of fibroblasts into the alveolar airspaces. Rat tracheal grafts with or without denuded epithelial cells were used to demonstrate that epithelial cells suppress migration of fibroblasts (Terzaghi et al., 1978). When the grafts without epithelial cells were transplanted into another rat, the tracheal lumen rapidly becomes obliterated by the inward migration of fibroblasts and formation of connective tissue. When the tracheal grafts were repopulated with epithelial cells, intralumenal fibrosis did not occur. Therefore, an intact epithelia layer appears to be crucial for preventing fibrosis. This is likely due to the fact that AECs produce inhibitory factors for fibrosis. For example, prostaglandin E2 (PGE2) that is secreted by alveolar epithelial cells is a potent inhibitor of fibroblast collagen synthesis and proliferation (Goldstein et al., 1982; Lama et al., 2002). Furthermore, the PGE2 levels are lower in broncho-alveolar lavage (BAL) fluid from patients with IPF compared to those from control patients (Borok et al., 1991). Therefore, loss of AECs or damaged AECs with deceased ability to synthesize PGE2 may contribute to pathogenesis of IPF. 3.2.3 Loss of cell surface that prevents collapse and fusion of alveolar walls Alveolar collapse and fusion of adjacent walls can occur in severely damaged areas of lung parenchyma in IPF patients due to loss of the epithelial cell surface. Fibrotic Connective tissue replaces the collapsed alveolar space and nuked basement membrane. Those processes lead to the decrease of alveolar surface area and contribute to the restrictive lung volumes (Simon, 1995). 3.2.4 Surfactant Alterations l4 Surfactant llS functit geometry Bronchoal surfactant ('Lovv, 198 abnormal. surfactant. 1991). It 1: PTOgnosis s The delicic abnonnal s Collapse of 0fHal-red b3 the hl'drosr (Gullon an companmer airspace am Wactam tL nothly gem 3.25 Surfactant is a lipid-rich material that is synthesized exclusively by type II alveolar cells. Its function is to lower the surface tension of the air-fluid interface so that alveolar geometry can be maintained to protect smaller alveoli from collapsing. Bronchoalveolar lavage fluid obtained from patients with IPF has abnormalities in its surfactant components. Particularly, the composition of phospholipids is out of ratio (Low, 1989; McCormack et al., 1991). The protein component of surfactant in IPF is also abnormal. For example, surfactant protein A (SP-A), the major protein species within surfactant, was shown to decrease in bronchoalveolar lavage fluid (McCormack et al., 1991). It was also revealed that patients with better-preserved levels of SP-A had better prognosis showed by lung functions test (McCormack et al., 1991). The deficiencies in surfactant have several impacts on the pathophysiology of IPF. First, abnormal surfactant causes higher surface tension on the alveoli that can lead to the collapse of smaller alveoli. Permanent collapse will occur where those alveoli have areas of naked basement membrane. Second, the abnormally high surface tension will decrease the hydrostatic pressure within the alveolar lining fluid below the air-fluid interface (Guyton and Moffatt, 1981). Pressure gradients between the vascular and interstitial compartments and the alveolar space will increase causing water movement into alveolar airspace and leading to alveolar flooding. Third, the deficiency of SP-A can affect Surfactant turnover as SP-A serves as a ligand for the uptake and reprocessing of pre- viously secreted surfactant by alveolar epithelial cells (Wright et al., 1987). 3. 2.5 Loss of the Alveolar Progenitor Cells 15 Type 11 cells whereas Type damaged area and differenti: ope ll cells It The regenerat normal healing FEE-W ding the 1 failure of reepi 3.2.6 L. Tll‘e ll cells, COmpaflmem b the Sodium-pm Strrface 0f the Process “as fr re Type 11 cells are known to serve as the progenitor cells for the alveolar epithelium whereas Type I cells are terminally differentiated and do not proliferate (Uhal, 1997). In damaged areas of the alveolar surface where type I cells are lost, type 11 cells proliferate and differentiate into type 1 cells. If the injury to the alveolar surface is too severe for type II cells to survive, the potential for reconstituting a normal alveolar surface is lost. The regeneration and reepithelialization of the normal alveolar wall are critical for normal healing without the consequence of fibrosis. Hence, the current presiding view regarding the mechanism of fibrogenesis holds that IPF results from epithelial injury and failure of reepithelization (Selman et al., 2001). 3.2.6 Loss of Active Transport Properties Type 11 cells, and probably type 1 cells, help to maintain a relatively dry alveolar compartment by clearing fluid within the compartment via active transporter coupled to the sodium-potassium ATPase. The sodium-potassium ATPase located on the basolateral surface of the cells pumps sodium ions out of the cell into the interstitial space. This process was first noted through tissue culture studies which demonstrasted domes formed by monolayers of type 11 cells (Mason et al., 1982; Goodman and Crandall, 1982). These domes are caused by accumulation of the fluid from the media under the monolayer as a result of import of sodium molecules and accompanying water. Consequently, the trapped fluid lifts up the monolayer and form domes (Goodman et al., 1983; Cott et al., 1986). Studies have also been performed on intact lungs and confirmed that solute molecules can be removed from the alveolar space against concentration gradients (Bassett et al., 1987; Matthay and Wiener-Kronish, 1990). Therefore, damage to or loss of the filled: ability of“ 1' 3.2.7 1 Plasmlnoge" a Alveolar epllh‘ ultra-alveolar l as nbanogen ls ll alveolar cell: 1985) come” The clearance 1 (Sitrin et al.. 1‘ fashion. it is pr “ill attach to interstitial colla, of pulmonary fit The fate of th of the alveolar epithelium would destroy the active transport system and impair the ability of the lung to clear fluid from the alveolar space. 3. 2. 7 Loss of the ability to clear intro-alveolar fibrin and regulate the plasminogen activation system: Alveolar epithelial cells are in part responsible for both the formation and clearance of intra-alveolar fibrin. When alveolar epitheliums are damaged, plasma components such as fibrinogen leak into the alveolar space. Tissue factors expressed on the surface of type II alveolar cells (Gross et al., 1991) and alveolar macrophages (McGee and Rothberger, 1985) convert fibrinogen to fibrin, which can act as scaffolds for fibroblast migration. The clearance of this intra-alveolar fibrin is critical to the outcome of the lung injury (Sitn'n et al., 1987; Brown et al., 1989). If extravascular fibrin is cleared in an orderly fashion, it is possible to reconstitute the normal alveolar space. Otherwise, fibroblasts will attach to and migrate along the remaining fibrin leading to the deposition of interstitial collagens and obliteration of the alveolar space, which are the characteristics of pulmonary fibrosis. The fate of the newly formed fibrin in alveolar space depends on whether the profibrinolytic and antifibrinolytic processes within the alveolar compartment is balanced. The fibrinolytic process is mediated by the plasminogen activation system, which tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) can activate (Martin et al., 2002). Both tPA and uPA can transform the plasminogen to plasmin, the primary fibrinolytic enzyme responsible for degrading fibrin. uPA is considered to be the major contributor for the fibrinolytic activity within 17 bronchoalveolar lavage fluid from normal lungs (Chapman et al, 1986; Hasday et al., 1988; Idell et al., 1989). In IPF the plasminogen activation system has been implicated to regulate fibrin turnover and ECM degradation (Simon et al., 1995). The antifibrinolytic process is mediated by plasminogen activator inhibitors (PAIs), which negatively regulate the activity of plasminogen and thus plasmin. Studies using animal models suggest that overexpression of PAIs (inhibiting plasmin activity) promotes fibrosis, whereas lack of PAIs (allowing greater plasmin activity) prevents the formation of significant fibrosis (Eitzman et al, 1996). Epithelial cells control the balance between fibrolytic and antifibrolytic process by synthesizing both urokinase (Gross et al., 1990) and plasminogen activator inhibitor-1 (Gross et al., 1990, 1991; Simon et al., 1992; Hasegawa 1997). Epithelial cells express urokinase that activates the plasminogen present within the plasma clot. Alveolar epithelial cells also express cell surface receptors for urokinase, which contain the fibrinolytic activity on the cell surface. Normal alveolar epithelium is efficient in clearing intra-alveolar fibrin and profibrinolytic. However, damaged alveolar epithelial cells often show impaired ability to remove intra-alveolar fibrin. For example, the fibrinolytic activity of bronchoalveolar lavage fluid from IPF patients was deficient (Chapman et al., 1986). The compromised fibrinolysis was due to a decreased level of urokinase protein and an increased level of the inhibitors for urokinase and plasmin, an imbalance between profibrinolytic and antifibrinolytic processes (Chapman et al., 1986). Kotani et al showed that BAL fluid from patients with IPF contains significantly greater amounts of tissue factor and PAIs than normals whereas uPA levels were similar between the two groups (Kotani et al., 1995). Combined together, those studies suggest that the alveolar microenvironment in 18 IPF favors a pro-coagulant, anti-fibrinolytic state promoting ECM accumulation and retarding alveolar re-epithelialization. 3. 2.8 Abnormal AE Cs as a primary source of profibrotic cytokines Dying or abnormal AECs in IPF synthesize numerous growth factors and cytokines that activate fibroblasts and mesenchymal cells. AECs are the primary source for transforming growth factor-beta (TGF-B) which transdifferentiate normal fibroblasts into the myofibroblast producing most of the ECM in the fibrotic lungs (Khalil et al., 1996). Additionally, AECs produce ANG II, platelet-derived growth factor (PDGF) (Antoniades et al., 1990), tumour necrosis factor alpha (TNF-u.) (Kapanci et al., 1995), and endothelin-l (Giaid et al., 1993). PDGF is a potent rrritogen and chemoattractant for fibroblasts. The mRNA and protein level of PDGF have been shown to be upregulated in epithelial cells of patients with IPF (Antoniades et al., 1990). TNF-a promotes DNA synthesis and proliferation of fibroblasts (Battegay et al., 1995) and was secreted by hyperplastic type II AECs in pulmonary fibrosis (Kapanci et al., 1995; Nash et al., 1993). Endothelin-l has also been shown to stimulate fibroblast DNA synthesis and proliferation as well as to induce transdifferentiation of fibroblasts to myofibroblasts (Shahar et al., 1999). The various fibrosis-promoting effects of those factors produced by damaged AECs support the view that death of the AECs within alveolar epithelium can be regarded as profibrotic and could initiate a fibrotic lesion. 4. Apoptosis of AECs is involved in pulmonary fibrogenesis 4.1 General information about apoptosis: 19 There death necros proces cellula death. enzym (I'hal .' c_\t0pla 311011101 from Its un“ante cell rem 1. SUll’lerI There are two kinds of cell deaths in alveolar epithelium: necrosis and programmed cell death also known as apoptosis. The morphological and biochemical characteristics of necrosis and apoptosis are different. Necrosis is considered as a passive and un-regulated process associated with inflammation which results in disintegration of membrane and cellular organelles (Saraste and Pulkki, 2000). In contrast to necrosis or "accidental" cell death, apoptosis is an active form of cell death that requires the activation of specific enzymes such as caspases, endonucleases and other components of signaling pathways (Uhal 2002). Apoptotic cells often are characterized with intact cell membrane, shrunk cytoplasm and fragmented DNA. These hallmarks are commonly used to identify apoptotic cells (Allen et al., 1997). Apoptosis results in single cell death and deletion from tissues with minimum inflammation. By doing so, apoptosis leads to the removal of unwanted or damaged cells. As apoptosis is a highly regulated physiological process of cell removal, it plays a fundamental role in the homeostatic control of cell population (Sutherland et al., 2001). 4.2 Epithelial cell apoptosis in fibrotic lungs A relatively small increase in the incidence of apoptosis within a given cell population can result in considerable cell loss over time. Therefore, a seemingly minor upregulation of apoptosis is theoretically capable of accounting for the excessive loss of AECs and the failure of the reepithelization characteristic of pulmonary fibrosis. Recent studies strongly suggested a role for epithelial apoptosis as a key profibrotic event in lung fibrogenesis. Evidence in support of this viewpoint is summarized below. 20 First. at models bronchir 1996) at et al.. 1‘ labeling biopsies epitheliu; positive 1' myofibro fccentl), Filho er al Consisten AECS vvit et al.. 199 SOlUble Fa 6131. 2C 103 Showed 91‘ induced Pu the [mind 1997). Second. ind In animal p First, apoptosis of AECs is found both in the lungs of patients with IPF and in animal models of the disease. Fragmented DNA, a hallmark of apoptosis, was found in bronchiolar cells and AECs within lung biopsies from patients with IPF (Kuwano et al., 1996) and in the lungs of mice and rats with bleomycin-induced lung fibrosis (Hagimoto et al., 1997; Wang et al., 2000). This finding was confirmed by simultaneous double labeling of fragmented DNA and a-smooth muscle actin, a marker for myofibroblasts, in biopsies from patients with IPF (Uhal et al., 1998). Fragmented DNA in the alveolar epithelium was found frequently and immediately adjacent to u-smooth muscle actin- positive interstitial cells and foci of collagen. Thus epithelial apoptosis colocalizes with myofibroblasts where collagen deposition is severe, at least in patients with IPF. Very recently, apoptosis within AECs of fibrotic human lungs was reconfirmed by Barbas- Filho et a1. (Barbas-Filho et al., 2001), also through the detection of fragmented DNA. Consistent with those findings, the "death receptor" Fas was found to be expressed in AECs within the lungs of IPF patients by several independent research groups (Kazufumi et al., 1997; Domagala—Kulawik et al., 2000). Thereafter, increased circulating levels of soluble FasL were shown to correlate with disease activity in patients with IPF (Kuwano et al, 2002). In animal models, similar observations were obtained; Hagimoto et al. (1997) showed epithelial apoptosis and upregulation of Fas on the epithelium in bleomycin- induced pulmonary fibrosis in mice. Another study demonstrated that Fas is expressed on the luminal surface of a subset of alveolar type 11 cells in the mouse model (Fine et al., 1997) Second, induction of apoptosis in the epithelium is sufficient to initiate a fibrotic response in animal models. Hagimoto et a1. (1997) showed that intratracheal instillation of an 21 antibody express 1 knockout effect of developrr instillatio Show in the devel pathuats Third. ph; al. (Wang Could be r d311," intr lZVADfm i“duction “mg the Strategy 1. Dummy} antibody that activates Fas-induced apoptosis of bronchial and AECs (both of which express Fas constitutively) initiated a fibrotic response detectable 1 wk later. Moreover, knockout mice deficient in the receptor Fas were found to be resistant to the profibrotic effect of bleomycin (Kuwano et al., 1999). However, another study found that the development of bronchiolar and alveolar epithelial apoptosis and fibrosis after bleomycin instillation in the lungs in Fas-null lpr mice and gld mice was similar to development shown in wild-type mice (Aoshiba et al., 2000). Thus the role of Fas-induced apoptosis in the development of the pulmonary fibrotic response is still controversial; in addition, pathways other than F as can initiate epithelial apoptosis and facilitate fibrogenesis. Third, pharmacological blockade of apoptosis can prevent the fibrotic response. Wang et al. (Wang et al, 2000) first found that bleomycin-induced accumulation of lung collagens could be blocked by the angiotensin-converting enzyme (ACE) inhibitor captopril or by daily intraperitoneal injections of N-benzylcarboxy-Val-Ala-Asp-fluoromethylketone (ZVADfmk), a broad-spectrum inhibitor of caspases (cysteine proteases) required for the induction of apoptosis. Soon thereafter, Kuwano et a1. (2001) confirmed the blockade by using the same caspase inhibitor (ZVADfmk) administered by aerosol to mice. Another strategy to interrupt AEC apoptosis proved effective in blocking bleomycin-induced pulmonary fibrosis; Inoshima et al (2004) showed that the forced expression of p21, predominantly in lung epithelial cells, exerted both antiapoptotic and antifibrotic effects. Thus the blockade of collagen deposition in vivo by inhibitors of apoptosis suggests that the fibrotic response is secondary to the apoptotic death of certain lung cell types. This premise in turn is consistent with the theories put forth by Witschi (1990) and Adamson and Bowden (1976) that alveolar reepithelialization is necessary to prevent subsequent 22 fibrogenesis after lung injury. Other data consistent with this theory include studies of keratinocyte growth factor (KGF), a potent proliferation and differentiation factor for alveolar type 11 cells known to promote alveolar epithelial repair (Stern et al., 2003). Both, KGF and the related hepatocyte grth factor (HGF) prevented bleomycin-induced lung fibrosis in rats and mice (Yi et al., 1996; Deterding et al., 1997; Sugahara et al, 1998; Yi et al., 1998; Dohi et al., 2000). 4.3 Mechanism and signaling of epithelial apoptosis in fibrotic lung Alterations in the expression of various antiapoptotic and proapoptotic factors likely regulate apoptosis of AECs. The proapoptotic factors p53 and p21 are upregulated in bronchiolar and AECs within lung biopsy specimens from patients with IPF and in bleomycin-induced pulmonary fibrosis animal models (Kuwano et al, 1996; Kuwano et al., 2000). Upregulation of the proapoptotic factor BAX in patients with diffuse alveolar damage and in bleomycin-induced pulmonary fibrosis may enhance the susceptibility of AECs to apoptosis (Guinee et al., 1997; Kuwano et al., 2000). In vitro studies of epithelial cell lines or primary cells showed that JNK, a member of the MAPK family is involved in stress-induced apoptosis. Activation of JNK (Adler et al., 1995: Janssen et al., 1999; Lander et al., 1996) leads to phosphorylation of its targets, c- Jun and activating transcription factor (ATF)-2 (Ip et al., 1998; Schaeffer et al., 1999), and activation of downstream gene expression when epithelial cells are exposed to oxidative stress, which is avery important contributor to pulmonary injury and fibrosis. An in vivo study of p38 MAPK in bleomycin-induced pulmonary fibrosis showed that p38 MAPK and its substrate, ATF-2, were phosphorylated in BAL fluid cells after 23 intratracheal instillation of bleomycin. The phosphorylation of ATF-2 was inhibited by subcutaneous administration of a specific inhibitor of p38 MAPK, FR-167653. FR- 167653 also prevented the apoptosis of lung cells and fibrosis induced by bleomycin administration (Matsuoka et al, 2002). Signaling pathways that can contribute to the apoptosis of AECs in IPF patients were investigated by Yoshida et al. (2002), who found that active ERK was decreased and active JNK was increased in epithelial cells and was accompanied by the progression of fibrosis. Activated p38 MAPK in epithelial cells was increased at the intermediate stage of fibrosis, in which the TUNEL-positive cells were predominantly detected (Yoshida et al., 2002). 5. Renin-Angiotensin System 5.1 General information The conventional renin-angiotensin system (RAS) is an endocrine system known to regulate fluid homeostasis, electrolyte metabolism and blood pressure etc. Renin, an aspartyl protease, is secreted by the granular cells of the juxtaglomerular apparatus of the kidney. Synthesis and release of renin into the circulation is considered as the rate- limiting step of the RAS and is activated by decreased renal perfusion and plasma sodium concentration. Angiotensinogen (ANGEN), the precursor for angiotensin II (Ang II), is secreted by the liver and cleaved by circulating renin to form the decapeptide: angiotensin I (Ang I). Angiotensin converting enzyme primarily located at the endothelial surface of pulmonary capillaries converts Ang I to an octapeptide Ang II. Ang II is considered the primary effector molecule of the RAS (Bader et al., 1994; Griendling and Alexander, 1994). 24 5.2 Effects of Angiotensin II ANG II exerts its effects through at least two subtypes of Ang 11 receptors, AT] and AT2. These receptors are G-protein coupled receptors that have seven transmembrane domains (Murphy et al., 1991; Sasaki et al., 1991). The majority of physiological and pathophysiologic functions of ANG II are mediated by ATI receptors. Those effects include, but are not limited to: l) Vasoconstriction in arterial smooth muscle and concomitant increase of blood pressure; 2) Secretion of aldosterone from the adrenal gland, which increases renal SOdlM‘ reabsorption; 3) Facilitated release of catecholamines in the sympathetic nerve, which increases cardiac output and total peripheral resistance and blood pressure; 4) Elevated sympathetic nerve activity and vasopressin secretion in the brain; 5) Enhanced cardiac contractility and ventricular hypertrophy; 6) Remodeling of the vascular wall resulting in increased total peripheral resistance (Geisterfer et al., 1988; Robertson and Nicholls, 1993); and 7) Direct action on the renal tubular cells to promote sodium and water reabsorption in the kidney (Deshmukh et al., 1998). ANG II is also proposed to play an important role in the pathogenesis of lung injury that will be discussed later in this chapter. 5.3 Angiotensin 11 signal pathway There exist several signaling pathways to transduct the effects of ANG II (Timmerrnans et al., 1993; Lee and Severson, 1994). AT] receptor-mediated signaling pathways have been more extensively studied than that AT2 mediated. ATl receptors are guanine 25 nucleo mediat 1) The bi phospl‘ (PIP2) binds t. the rele activate the cm Which calmed kinases also act Phosph; PhOSphr p hOSPh: leardjng phl'Siol, 2; A AT] rec Wovens PIOsragIa nucleotide-binding regulatory proteins (G proteins)-coupled. The signaling pathways mediated by ATI receptors are as follows: 1) Increases intracellular [Caz +1 : The binding of ANG II to AT] receptors activates the Gq protein and consequently phospholipase C-beta (PLCB). PLC B hydrolyzes phosphatidylinositol-4, 5-bisphosphate (PIP2) to generate inositol-l, 4, 5-trisphosphate (1P3) and diacylglycerol (DAG). 1P3 binds to receptors on IP3-sensitive organelles with intracellular Ca2+ stores and stimulates the release of intracellular Ca2+. In addition, binding of ANG II to AT1 receptor directly activates the cytoplasmic membrane-bound Ca2+ channels to allow the influx of Ca2+ into the cytoplasm. Both pathways lead to the elevation of intracellular Ca2+ concentration, which produce a variety of effects. Then Ca2+ binds to calmodulin, and the Ca2+/ calmodulin complex activates a number of intracellular enzymes, such as ATPase and kinases that contribute to the cellular response. The binding of ANG II to ATl receptors also activates phospholipase D (PLD), which hydrolyzes phosphatidylcholine to generate phosphatidic acid. Phosphatidic acid is then transformed into DAG by phosphatidate phosphohydrolase. DAG derived from phosphatidylcholine through PLD and phosphatidylinositol-4, 5-bisphosphate through PLC activates protein kinase C (PKC) leading to the phosphorylation of downstream proteins and cascade events to produce physiological response. 2) Increases Arachidonic Acid metabolites: AT1 receptor activation also stimulates activation of phospholipase A2 (PLA2), which converts phosphatidylcholine to arachidonic acid. Arachidonic acid is metabolized to prostaglandins and thromboxane A2 by cyclooxygenase and to hydroxyeicosatetraenoic 26 acids and IC activate a con 3) Decre. In some cells (onandsube of intracellula inhibited activ of substrates ft 4) Increw Activation of expression of t such as AP] ( STATts) active proteins such a In addition. int DNA associate PDGFrRe.200 acids and leukotrienes by lipoxygenase. Those metabolites of arachidonic acid then activate a complex of cellular signaling pathways. 3) Decrease intracellular [CAMP]: In some cells AT1 receptor activation leads to the activation of the inhibitory G-protein (Gi) and subsequently inhibition of adenylyl cyclase activity and thus the decreased level of intracellular cyclic AMP (CAMP). Decreased intracellular [CAMP] level leads to the inhibited activity of protein kinase A (PKA) and thereby supressed phosphorylation state of substrates for PKA causing corresponding biological effect. 4) Increases growth factors production: Activation of AT] receptors by ANG II stimulates cell growth by regulating gene expression of transcriptional factors of MAP kinase and JAK tyrosine kinase(s) pathway such as AP] (F05 and Jun) and STAT(s) (Bhat et al., 1994). AP] (Fos and Jun) and STAT(s) activate the transcription of a number of growth factors and extracellular matrix proteins such as TGF [3 and collagen. In addition, internalized ANG 11 together with intracellular ANG II can directly bind to DNA associated AT] like receptors and regulate gene transcriptions of renin, ANGEN. PDGF (Re, 2004). 6. Pulmonary angiotensin system in idiopathic pulmonary fibrosis 6.1 Local pulmonary angiotensin system: Recently a few studies suggested the existence of “local” angiotensin systems in various organs and tissues. For example, the ANG 11 concentrations in the interstitial compartment of heart and eye were found to be 5-100 fold higher (about 50 —500pM) 27 than that in plasma (~5-10pM) (van Kats et al., 1998; Danser et al., 1994). The higher interstitial levels of ANG 11 compared to the circulating level could not be explained by diffusion and/or receptor-mediated uptake of circulating angiotensin 11. These results thereby suggest that tissue angiotensin II is largely, if not completely, synthesized locally. Furthermore, cultured cells from various organs including heart (Lindpaintner et al., 1988), vascular endothelium (Li et al., 1999), brain (Campbell et al., 1986; Dzau et al., 1982; Ohkubo et al., 1986) and lung (F ilippatos et al., 2001) were shown to express the RAS components such as ANGEN, ANG II and their corresponding converting enzymes and angiotensin receptors. In contrast to the classical endocrine system of RAS in which angiotensin II is delivered to tissues via circulating blood, local angiotensin systems can be from either “intrinsic “(independent of the endocrine RAS) or “extrinsic” (relying on the endocrine RAS as its components sources) sources. 6.2 Pulmonary angiotensin system components: The local pulmonary angiotensin system appears to consist of ANGEN, ANG I, ANG II and their corresponding converting enzymes and angiotensin receptors. 6.2.1 ANGEN: It was shown that two types of cells in the lung could produce ANGEN in vitro under certain conditions: AECs and myofibroblasts. For example, primary AECs could synthesize and secrete ANGEN, which was converted to ANG II when undergoing apoptosis induced by Fas ligand (Wang et al., 1999), TNF-alpha (Wang et al., 2000), amiodarone (Bargout et al., 2000) and bleomycin (Li et al., 2003). Primary myofibroblasts isolated from fibrotic human lungs (IPF biopsies) expressed the ANG II 28 precursor ANGEN mRNA and protein (Wang et al., 1999), suggesting that human lung myofibroblast can synthesize ANGEN in culture. Furthermore, preincubation of the culture medium of myofibroblast with purified renin and ACE increased the Enzyme- Linked Immunosorbent Assay (ELISA)- detectable ANG 11 concentration eight folds, indicating that there are abundant ANGEN synthesized constitutively waiting to be converted to AN G II (Wang et al., 1999). Despite of evidence from in vitro studies, there is no in vivo study demonstrating the existence of lung-derived ANGEN. 6.2.2 Renin and Renin- like enzymes: Production of the local angiotensin systems can use elements that are “extrinsic” such as relying on the renin from the endocrine RAS. In many organs and tissues such as the heart and vessel wall, the synthesis of local ANG II was shown to depend on the uptake of circulating renin and/ or prorenin either via diffusion into the interstitial space or through binding to prorenin receptors (Re, 2004). No renin mRNA has been detected in the lungs and it is unclear whether there exists samilar mechanism in the lung to locally produce ANG 11. Nevertheless, there exist non-renin proteases capable of generating angiotensins in the lung such as cathepsin D. Cathepsin D might be responsible for the synthesis of generating angiotensin I from ANGEN and subsequent synthesis of ANG II in some cells like vascular interstitial cells (Weber et al., 1995). Other proteases, such as the enzyme tonin, can also generate angiotensin I from angiotensinogen (Re, 2004). 6.2.3 ACE and ACE-like enzymes: 29 There exist two forms of ACE. One is membrane-bound and the other is soluble. The membrane-bound ACE is attached to the plasma membrane of endothelial cells via a short hydrophobic sequence at the C-terminus and is found in most organs. The soluble ACE, detached form of the membrane-bound ACE, is found in most of the body fluid including lymph, plasma, cerebrospinal fluid and amniotic fluid (Erdos, 1990). The soluble form lacks the hydrophobic terminus and is derived from the membrane-bound form via post-translational cleavage by an ACE-secretase. Membrane-bound ACE is abundantly expressed by the vascular endothelium of the pulmonary circulation and is primarily responsible for the conversion of ANG I to ANG II in the circulation (Oparil et al., 1971). The extent of the conversion of ANGI to ANG II is a function of the vascular surface area and the transit time of blood circulating through the lung (Oparil et al., 1971). The contribution of soluble ACE to ANG II generation is negligible (N g and Vane 1968; Admiraal et al., 1993). Primary cultures of AECs were shown to express the ACE mRNA (Wang et al., 1999a, b). Other non-ACE enzymes that also convert angiotensin I to angiotensin 11 include chymase found in human cardiac tissue, cathepsin G and ACE2, a recently described enzyme (Re, 2004). 6.2.4 Angiotensin II receptors: Bullock et al. demonstrated that in the human lung AT1 receptor mRNA and protein were localized on vascular smooth muscle cells, macrophages and in the stroma underlying the airway epithelium, possibily relating to underlying fibroblasts (Bullock et al., 2001). However, the AT] receptor protein was not detected in the epithelium although there was 30 a lovv 16"51 O the within“ underlying ’1 study. h Owev (Bullock 3‘ a localized 0“ endothelial C increased ”13 generate lib“ pulmonarl' Iii” 6.3 Potent 6.3.1 P Purified ANG This ellect ml apoptosis of A 2000). amiodl angiotensin s_\ oligonucleotide (AI receptor) inducers of alt. induced bv- Fa~ a low level of mRNA. In contrast, AT2 receptor RNA and protein was strongly stained in the epithelium, particularly on the bronchial epithelial cell brush border and the underlying mucous glands as well as some endothelial cells (Bullock et al., 2001). This study, however, did not exam the AT] and AT2 receptors in the alveolar epithelial cells (Bullock et al., 2001). Consistently, another study on rat lung reported that AT] was localized on alveolar macrophages, alveolar type II cells, vascular smooth muscle cells, endothelial cells and fibroblasts (Otsuka et al., 2004). AT] expression in the lung increased markedly after intratracheal administration of BLM, which is known to generate fibrosis, suggesting that angiotensin II as a ligand of AT] is involved in pulmonary fibrosis (Otsuka et al., 2004). 6.3 Potential roles of pulmonary AN G 11 in pulmonary fibrosis: 6.3.1 Pulmonary angiotensin system is linked to lung epithelial apoptosis: Purified ANG II was shown to induce apoptosis of AECs in culture (Wang et al., 1999). This effect was mediated by AT] receptors (Papp et al., 2002). It was also shown that apoptosis of AECs induced by Fas Ligand (Wang et al., 1999), TNF-alpha (Wang et a1 2000), amiodarone (Bargout et al., 2000) and bleomycin (Li et al., 2003) required angiotensin synthesis de novo as AECs apoptosis could be blocked by the antisense oligonucleotides against mRNAs of the ANGEN, ACE inhibitors, and ANG II receptor (AT receptor) antagonists in vitro. Furthermore, human lung myofibroblast—derived inducers of alveolar epithelial apoptosis were identified as angiotensin peptides (Wang et al., 1999). These data suggest that ANG II is the “Master Switch” for apoptosis of AECs induced by Fas Ligand, TNF-alpha, amiodarone and bleomycin. This “Master Switch” 31 function of in fibrogene: fibrosis. 6.3.2 ANG II stimt receptor and t al.. 2000). A vitro via AT] function of ANG II for AECs apoptosis could account for the role of angiotensin system in fibrogenesis since apotosis of AECs is essential for the development of pulmonary fibrosis. 6. 3.2 Mitogenic for fibroblasts and Activate T GF beta expression: ANG II stimulates fetal and adult human lung fibroblast proliferation in vitro via the AT1 receptor and the autocrine action of transforming growth factor beta (TGF [3) (Marshall et al., 2000). ANG II also increased procollagen synthesis by human lung fibroblasts in vitro via AT] receptors (Marshall et al., 2000). 6.3.3 Inhibition of the fibrinolytic pathway: Mice with deficient fibrinolytic system developed pulmonary fibrosis in response to bleomycin, suggesting that IPF could result from compromised fibrolytic capability (Swaisgood et al., 2000). Plasminogen activator inhibitor 1 (PAI-l) inhibits plasmin and subsequently decreases the fibrinolytic capability promoting the deposition of collagen and other extracellular matrixs in bleomycin-induced lung injury in animal models (Eitzman et al., 1996; Swiderski et al., 1998). ANG II has been shown in rat microvessel endothelial cells to control thrombosis by inducing, PAI-l, the inhibitor for extracellular matrix turnover and fibrinolysis (N ishimura et al., 1997). In addition, in a rat model used to investigate the cardiac vasculopathy mediated by nuclear factor-KB and activator protein-1, ANG II was shown to regulate the thrombogenic pathway by increasing the expression of tissue factor which can convert fibrinogen into fibrin (Muller et al., 2000). 32 Hence. AN(' and induce a 6.3.4 HGF has bee suggesting t} Taniyama e1 prevented ms for local 11 dounregulatir 6.4 Angie The angiotens 6”ch5 and much evidenc 6.4.1. 1 A number of s Peptide Were 11 upregulated in increased in the Pulmonan. . fibr 1990). thcm Hence, ANG II can break the balance between the fibrolytic and antifibrolytic process and induce an environment favoring the deposition of collagen. 6.3.4 Downregulation of hepatocyte growth factor (H GP) expression: HGF has been shown to be important in suppressing bleomycin-induced fibrosis in mice, suggesting the antifibrotic function of HGF (Yaekashiwa et al., 1997). Furthermore, Taniyama et al. (2000) using a cardiomyopathic hamster model showed that HGF prevented myocardial fibrosis and that both ANG II and TGF-beta are strong inhibitors for local HGF production. Therefore, ANG 11 may have profibrotic role by downregulation of the hepatocyte growth factor (HGF) expression in the lung. 6.4 Angiotensin system is involved in pulmonary fibrosis: The angiotensin system consists of ANGEN, ANGI, ANG 11, corresponding converting enzymes and angiotensin 11 receptors (Proudn 199]; Filippatos et al., 200]). There is much evidence suggesting that angiotensin system is involved in pulmonary fibrosis. 6.4.1. Increased levels of converting enzymes and A T I receptors in IPF A number of studies show that enzymes required for the production of local angiotensin peptide were upregulated in pulmonary fibrosis. ACE which converts AN G1 to ANG II is upregulated in both human and animal fibrotic lungs. ACE activity was shown to be increased in the bronchoalveolar lavage fluid (BALF) of animals with bleomycin-induced pulmonary fibrosis (V enkatesan, et al.1997) and human patient with IPF (Specks et al., 1990). Furthermore, bleomycin upregulated gene expression and enzyme activity of ACE 33 in bovine pulmonary artery endothelial cells (BPAEC) (Day, et al., 2001). Those data suggest ACE may play a critical role in development of pulmonary fibrosis. In addition, the study to examine the incidence of D allele of the insertion/ deletion (I/D) polymorphism of ACE in patients with interstitial pneumonia and moderate to severe pulmonary fibrosis showed that the incidence of the D allele which confers a higher ACE expression was approximately 15% higher in the study population than in the general population (Morrison et al., 2001). Although the sample size is limited, these data indicate that polymorphisms that confer higher levels of ACE predispose patients to lung fibrosis and thus support the hypothesis that ACE and its product ANG II are involved in the pathogenesis of human pulmonary fibrosis. Cathepsin D (Cat D) is one of the enzymes capable of cleaving ANGEN to ANG 1. Cat D was upregulated in both animal and human fibrotic lung. Our lab showed that Cat D immunolabeling in alveolar wall cells morphologically consistent with type I and type 11 cells increased following intratracheal administration of bleomycin. Meanwhile, soluble Cat D enzymatic activity was elevated in cell-free bronchoalveolar lavage fluid (BALF) from the same lungs. Upregulation of Cat D was also been shown in human lungs from PF patients (Kasper et al., 1996) and was induced in L132 lung cells during apoptosis (Kasper et al., 1999). AT] receptors were also upregulated in animal fibrotic lung (Otsuka et al., 2004). AT] expression in the lung increased markedly after intratracheal administration of bleomycin, suggesting that angiotensin II as a ligand of AT] is involved in pulmonary fibrosis (Otsuka et al., 2004). 34 6.4.2. Blockade of experimental lung fibrosis by angiotensin system antagonists Application of ACE inhibitor, which presumably inhibits ANG 11 production, has been shown to attenuate experimental pulmonary fibrosis in animal models induced by various agents. For example, ACE inhibitor captopril exerted inhibitory effects on monocrotaline (Molteni et al, 1985) as well as y irradiation-induced lung fibrosis in rats (Ward et al, 1990). Captopril also inhibited the proliferation of human lung fibroblast in vitro (Nguyen et al., 1994). More recently, the AT] receptor-selective antagonists L158809 as well as the nonthiol ACE inhibitor enalapril were shown to have similar effects on radiation-induced pulmonary fibrosis in rats (Molteni et al., 2000). Uhal et al demonstrated that captopril prevented collagen deposition in bleomycin-treated rats (Wang et al., 2000). It was later shown that captopril and AT] selective antagonist losartan blocked the amiodarone induced pulmonary fibrosis in rats. (Uhal et al., 2002) These results combined together indicate that ANG 11 plays an important role in lung fibrogenesis via AT1 -receptor. 6. 4.3. Blockade of apoptosis by ACE inhibitor captopril or caspases inhibitors blocks pulmonary fibrosis Caspase is one of the key enzymes mediating apoptosis. Collagen deposition and epithelial apoptosis were blocked by both captopril and ZVADfink, a broad-spectrum inhibitor of caspases in intratracheally administered bleomycin-induced pulmonary fibrosis in rats (Wang et al., 2000). Another group confirmed this result by showing that the same caspase inhibitor delivered by inhalation attenuated bleomycin-induced 35 pulmonary fibrosis in mice (Kuwano et al., 2001). Those results suggest that both ANG II and epithelial apoptosis are required for lung fibrogenesis. 7. Working hypothesis: The pulmonary angiotensin system including its components angiotensinogen, cathepsin D and AT1 receptor plays an essential role in the development of bleomycin-induced pulmonary fibrosis at least in part through ANG II-ATl receptor pathway mediated apoptosis of alveolar epithelial cells. The role of pulmonary angiotensin system during PF can be summarized in the following figure (Fig. 1.1): Lung epithelium apoptosis inducers: Bleomycin _ An . t . Antisense of go ensmogen ANGEN mRNA CatD? 4+ Angiotensin I l ACE ACE inhibitors -> Angiotensin II /\ AT receptor a antagonists Apo ptosis > Fibrosis Bleomycin induces apoptosis of lung AECs by upregulation of ANGEN gene expression. Synthesized ANGEN can be converted to ANG I by cathepsin D (Cat D). ANG I can be further converted to ANG II by ACE. ANG II can induce apoptosis of the AECs through 36 AT1 receptor, which will cause excessive loss of AECs and insufficient epithelial repair. ANG 11 produced by apoptotic epithelial cells can also directly contribute to the fibroblast activation. Both ways will lead to lung fibrosis. Synthesis of ANGEN is the key upstream event in this process, which can be blocked by the antisense against ANGEN mRNA. 8. Significance: Our study is one of the first studies to determine the existence of intrinsic pulmonary angiotensin system and its role in the development of bleomycin induced pulmonary fibrosis. This study helps us to better understand the mechanism of lung fibrogenesis with regard to the pulmonary angiotensin system, which will likely lead us to the finding of effective therapeutic or preventive approaches for IPF. 37 Chapter 2 HYPOTHESIS AND SPECIFIC AIMS Treatment for IPF targeting the suppression of inflammation has not been successful, suggesting that inflammation is not the sole mechanism underlying lung fibrogenesis. Mortality of IPF patients is not dependent on severity of inflammation, but correlates well with the presence of “fibroblastic foci” and adjacent failure of reepithelization (Selman et al., 200]). The normal alveolar epithelium has “anti-fibrotic” functions including inhibiting fibroblast proliferation. Loss of alveolar epithelial cells (AECs) and failure of reepithelization characterized in pulmonary fibrosis can be considered as profibrotic and are believed to initiate the fibrotic lesion. The loss of AECs could result from necrosis and/ or apoptosis. Increased level of apoptosis was found in AECs in experimental and human pulmonary fibrosis. One indication that the angiotensin system is involved in fibrogenesis is findings that blockade of angiotensin systems blocked Pulmonary Fibrosis (PF) at least in several animal models. Our lab showed that angiotensin II (ANG II) induces apoptosis of the primary AECs through ANG 11 type I (AT1) receptor in vitro. Apoptosis of AECs induced by Fas Ligand, TNF-alpha, and amiodarone requires angiotensin synthesis de novo as AECs apoptosis can be blocked by the antisense oligonucleotide against the mRNA of the angiotensinogen (ANGEN), angiotensin converting enzyme (ACE) inhibitors, and ANG II receptor (AT receptor) antagonists in vitro. Taken together, those data suggest that ANG II, the processed product of ANGEN, is the key to regulate apoptosis of AECs and subsequent lung fibrosis. 38 The existence of “local angiotensin system” in the lung, which is independent of the endocrine RAS, is supported by studies demonstrating the expression of angiotensin system components in cultured primary rat AECs in response to Fas Ligand, TNF-alpha, and amiodarone and myofibroblasts from human fibrotic lungs. Bleomycin-induced rat and mouse pulmonary fibrosis model is a well-studied model for fibrogenesis. But there is much still unknown about the components and roles of pulmonary angiotensin system in bleomycin-induced pulmonary fibrosis. Overall Hypothesis: The pulmonary angiotensin system including its components angiotensinogen, cathepsin D and AT1 receptor plays an essential role in the development of bleomycin-induced pulmonary fibrosis at least in part through ANG II-ATl receptor pathway mediated apoptosis of alveolar epithelial cells. Specific Aims: 1) To examine if bleomycin-induced apoptosis of alveolar epithelial cells requires angiotensin synthesis de novo. Specific hypothesis: AEC apoptosis in response to bleomycin (BLEO) requires ANG synthesis and might be inhibited by ANG system antagonists. 2) To identify the primary aspartyl protease that could convert angiotensinogen to AN GI and contribute to pulmonary angiotensin system in vitro. Specific hypothesis: Cat D is required for ABC apoptosis in response to bleomycin at least in part by converting angiotensinogen to ANGI. 39 3) To examine if angiotensin receptor ATl is essential for AEC apoptosis and lung fibrosis in viva. Specific hypothesis: Administration of the ATl-selective receptor antagonist and deletion of the ATla receptor gene block BLEO-induced AEC apoptosis and lung fibrosis. 4) To identify the cellular sources of lung-derived ANGEN in situ: Specific hypothesis: The apoptotic type II alveolar epithelial cells and myofibroblasts are the major cellular sources of lung-derived ANGEN. 5) To examine the effect of blockade of lung-derived AN GEN on pulmonary fibrosis. Specific hypothesis: Administration of the antisense against ANGEN messenger RNA attenuates bleomycin-induced AEC apoptosis and lung fibrosis. 40 Chapter 3 BLEOMYCIN-INDUCED APOPTOSIS OF ALVEOLAR EPITHELIAL CELLS REQUIRES ANGIOTENSIN SYNTHESIS DE N0 V0 Xiaopeng Li, Huiying Zhang, Valerie Soledad-Conrad, J iaju Zhuang and Bruce D. Uhal Department of Physiology, Michigan State University, East Lansing, Michigan, 48824 This chapter is published in: Am J Physiol Lung Cell Mol Physiol. 2003, 284(3): L501-507 41 ABSTRACT Primary cultures of rat type II alveolar epithelial cells (AECs) or human AEC-derived A549 cells, when exposed to bleomycin (BLEO), exhibited dose-dependent apoptosis detected by altered nuclear morphology, fragmentation of DNA, activation of Caspase 3 and net cell loss over time. In both cell culture models, exposure to BLEO caused time- dependent increases in angiotensinogen (ANGEN) mRNA. Antisense oligonucleotides against ANGEN mRNA inhibited BLEO-induced apoptosis of rat AEC or A549 cells by 83% and 84%, respectively (p<0.01 and p<0.05) and prevented] BLEO-induced net cell loss. Apoptosis of rat AECs or A549 cells in response to BLEO was inhibited 91% by the ACE inhibitor captopril or by 82%, respectively, by neutralizing antibodies specific for ANGII (both p<0.01). Antagonists of ANG receptor AT] (losartan, L158809 or saralasin), but not an AT2-selective blocker (PD123319), inhibited BLEO-induced apoptosis of either rat AECs (79%, p<0.01) or A549 cells (83%, p<0.01) and also reduced the activity of Caspase 3 by 52% (p<0.05). These data indicate that BLEO, like FasL or TNF-a, induces transactivation of ANG synthesis de novo that is required for ABC apoptosis. They also support the theory that ANG system antagonists have potential for the blockade of ABC apoptosis in situ. 42 INTRODUCTION Alveolar epithelial cells (AECs) have many important roles that are critical to normal lung function (Mason and Williams, 1991). The death of AECs by apoptosis is now believed to be an important event in the pathogenesis of lung fibrosis (Haschek and Witschi, 1979) and in more acute lung injury (Matute-Bello et al., 2001; Uhal, 2001). A variety of investigations have implicated important roles for key molecules such as tumor necrosis factor alpha (TNF-a) and F as ligand (F asL), both known inducers of apoptosis in a variety of cell types, in the events that lead to fibrogenesis in the lung (Hagimoto et al., 1997a; Hagimoto et al., 1997b; Ortiz et al., 1998). In earlier work (Wang et al., 2000; Wang et al., 1999), we showed that exposure of either primary cultures of rat AECs or the human AEC-derived A549 cell line to FasL or TNF-a increases angiotensinogen (ANGEN) mRNA and protein, and evokes its subsequent conversion to angiotensin II (ANGII). Moreover, we found that transactivation of ANGII synthesis is required for ABC apoptosis in response to TNF-a or F as ligand. Thus, AEC death in response to these could agents could be blocked by ANG receptor antagonists or inhibitors of ANG converting enzyme (ACEis), at least in vitro. Studies of another inducer of ABC apoptosis, the antiarrythmic agent amiodarone, also showed that antagonists of ANG production or receptor interaction could prevent apoptosis of AECs in response to this benzofuran compound (Bargout et al., 2000). For these reasons we hypothesized that AEC apoptosis in response to bleomycin (BLEO) might also require ANG synthesis and might therefore be inhibitable by ANG system antagonists. We report here that bleomycin, if applied to rat or human AECs in vitro, induces the expression of angiotensinogen mRNA and subsequent apoptosis that can be 43 blocked by ANGEN antisense oligonucleotides, by ACEis or by ANG receptor antagonists of the ATl-selective subtype. 44 MATERIALS AND METHODS Reagents and materials: The ATl-selective antagonists L158809 and losartan were obtained from Merck and Co., West Point, PA. The AT2-selective antagonist PD123319 was obtained from Parke Davis Research Division, Ann Arbor, MI. The caspase inhibitor ZVAD-fink (N-benzylcarboxy-Val-Ala-Asp- [O-Me]-CH2F) was obtained from Kamiya Biomedical, Seattle, WA. DEVDfmk (Asp-Glu-Val-Asp- [O-Me]-CH2F) was obtained from Pharrningen, San Diego, CA. Alkaline phosphatase-conjugated streptavidin, digoxigenin-labeled deoxyuridine trisphosphate (dig-dUTP) and biotinylated deoxyuridine trisphosphate (bio-dUTP) were obtained from Boerhinger Mannhiem, Indianapolis, IN. Bleomycin (BLEO), anti-angiotensin antibodies, ATA, captopril and saralasin were obtained from Sigma Chemical Co., Saint Louis, MO. Reagents for detection of alkaline phosphatase and other secondary reagents for in situ end labeling of DNA or western blotting were from sources described earlier (Wang et al., 2000). All other materials were of reagent grade and were obtained from Sigma Chemical Co., Saint Louis, MO. Cell culture: The human lung adenocarcinoma cell line A549 was obtained from the American Type Cell Culture Collection and cultured in Ham’s F12 medium supplemented with 10% fetal bovine serum (FBS). Primary alveolar epithelial cells isolated from adult male Wistar rats as described earlier (Wang et al., 1999). The primary cells were studied at day two of culture, a time at which they are type II cell-like by accepted morphologic and biochemical criteria (Paine and Simon, 1996). Primary cell preparations were of better than 90% purity assessed by acridine orange staining as described previously (Wang et al., 2000; Wang et al., 1999). All cells were grown in 24- 45 or 6-well chambers and were analyzed at subconfluent densities of 80-90%. All subsequent incubations with BLEO and/or other test agents were performed in serum-free medium. The cells were exposed to caspase inhibitors or antagonists of the angiotensin system 30 minutes before exposure to BLEO for 1-20 hours as indicated. Quantitation of apoptosis and cell loss: Detection of apoptotic cells with propidium iodide (PI) was conducted as described earlier (Wang et al., 2000; Wang et al., 1999) following digestion of ethanol-fixed cells with DNase-free RNase in PBS containing 5ug/ml P1. In these assays, detached cells were retained by centrifugation of the 24-well culture vessels during fixation with 70% ethanol. Cells with discrete nuclear fragments containing condensed chromatin were scored as apoptotic. As in earlier publications, the induction of apoptosis was verified by in situ end labeling (ISEL) of fragmented DNA (Uhal et al., 1998; Wang et al., 2000; see Figure 3.1). Apoptotic cells were scored over a minimum of four separate microscopic fields from each of at least three culture vessels per treatment group. Cell loss over 20 hours of culture was quantitated by cell counts of the adherent plus detached cell populations. These were obtained following centrifiagation of the culture vessels as described in the preceding paragraph, without prior washing. Thus, detached cells (routinely less than 10% of the total cell number) were included in the cell loss data. Total cell counts (attached plus detached) were scored over a minimum of 200 nuclei per field, 4 fields per well with a minimum of 6 culture wells, or 4,800 nuclei, per treatment group. Data from each treatment group were compiled and analyzed by ANOVA followed by Student-Newman-Keul’s post hoc analysis. 46 Detection and quantitation of caspase 3 activity: Activation of Caspase 3 was detected through: a) immunolabeling of fixed cells adherent to plastic culture surfaces with an antibody that recognizes only the active form of the enzyme (Biovision, Mountain View, CA). The primary antibody was detected with an alkaline phosphatase-conjugated secondary antibody followed by nitro-blue tetrazolium. The enzymatic activity of Caspase 3 was measured in adherent cells incubated for 20 hours with the membrane permeable substrate Ac-DEVD-AMC (Upstate Biotech, Saranac Lake, NY) at SOuM. Quantitation of the fluorescent product was achieved with a Biotek FL600 fluorescence plate reader. Fluorescence values were normalized to cell number determined on the same culture well after cell fixing and staining of DNA with propidium iodide (Uhal and Rannels, 1991). RTPCR and antisense experiments: Semiquantitative reverse transcriptase polymerase chain reaction was performed as described earlier (Wang et al., 2000; Wang et al., 1999). The annealing temperatures for PCR reactions were optimized for each primer by preliminary trials. All PCR amplifications were terminated at or near the center of the linear range for each gene product analyzed, as determined by sequential withdrawal of sample at 5-cycle intervals between 20 and 40 cycles (not shown). The identity of expressed genes was determined by expected size ‘of the PCR product in 1.6% agarose gels. For RTPCR of rat-specific gene products, the following primers were used: for angiotensinogen, coding = 5'-CCTCGCTCTCTGGACTTATC-3', and uncoding = 5'- CAGACACTGAGGTGCTGTTG-3', which yields a PCR product of 226bp by single- step RTPCR (Pierzchalski et al., 1997). For El-microglobulin, the primers used were: 47 coding = 5' -CTCCCCAAA-TTCAAGTGTACTCTCG-3', and uncoding = 5'- GAGTGACGTGTTTAACTCTGCA-AGC-3', which yields a product of 249bp (Katwa, et al., 1995). For RT—PCR from human A549 cells, the following primers were used: for angiotensinogen, coding = 5'GCTTTC-AACACCTACGTCCA3', and uncoding = 5'AGCTGTTGGGTAGACTCTGT3'. These primers yield a final PCR product of 509bp (Lai et al., 1998). For [II-actin, single-step RTPCR was used with the primers: coding = 5'AGG-CCAACCGCGAGAAGATGACC3', and uncoding = 5'GAAGTCCAGGGCGACGT-AGC3', which produces a PCR product of 332bp (Ponte et al., 1984). For antisense studies, phosphorothioated control and antisense oligonucleotides against angiotensinogen (l8-mers) were synthesized and transfected into A549 cells or primary rat AEC (both at 4011M final concentration) using the lipofectin reagent OligofectAMINE (Invitrogen Life Technologies, Grand Island, NY) at 4ul/ml as the vehicle, diluted in the OPTIMEM medium accompanying the lipofectin. The control nucleotides were of the same length and base composition as the antisense, but with scrambled sequence. The oligonucleotide: lipofectin ratio was optimized (over a 4hr tranfection) to yield transfection efficiencies of 50-75% with no apparent cell loss or detachment. Transfection efficiency was monitored with F ITC-labeled 25-mer oligonucleotide for luciferase (not shown). Transfections were conducted for 4 hours followed by 5 times washing with serum-free cell culture medium; immediately thereafter, BLEO or vehicle was applied as described above for 20 hours. The transfection protocol itself had no significant effect on basal or BLEO-induced apoptosis (see Results). Phosphorothioated oligonucleotides used for transfection were: (ANGEN antisense) 5- 48 CCGTGGGAGTCATCACGG-3', and (ANGEN scramble) 5'- CAGGGATCTCTGGCGGAC-3' as described by Phillips et al. (Phillips et al., 1994). Attention: Images in this dissertation are presented in color. 49 RESULTS Exposure of primary cultures of rat AECs or A549 cells to BLEO for 20 hours caused apoptosis detectable by nuclear fragmentation, DNA fragmentation and by immunolabeling of the active form of Caspase 3 (Figure 3.1). Although these markers were detected in a minor fraction of the cells, the apoptosis induced was sufficient to reduce the total cell number significantly over time (see Figure 3.1D-F and quantitation in Figure 3.7B). Scoring of fragmented nuclei revealed dose-dependent apoptosis that reached statistical significance beginning at 0.5mU/ml in primary AECs (Figure 3.2) and at lmU/ml in A549 cells (not shown). The nuclear fragmentation was blocked by the broad-spectrum caspase inhibitor ZVAD-fink (60uM) or by the endonuclease inhibitor aurintricarboxylic acid (ATA, IOuM), confirming the specificity of the assay for apoptosis. Apoptosis of the rat AECs also was blocked by the ACE inhibitor captopril (CAPTO, SOOng/ml) and by the nonselective ANG receptor antagonist saralasin (SARAL, 50ug/ml), in agreement with earlier studies of F as L and TNFa-induced AEC apoptosis (Wang et al., 2000; Wang et al., 1999). In a separate experiment (Figure 3.3), BLEO-induced apoptosis of primary AECs was blocked by the Caspase 3-selective blocker DEVD-fmk (60uM) and by the ANG receptor subtype ATl-selective blocker losartan (10'6M, p<0.01), suggesting that subtype AT] mediates BLEO-induced apoptosis as it does AEC apoptosis in response to ANGII (Papp et al., 2002). This was found to be the case in human AECs as well (Figure 3.4); BLEO- induced apoptosis of A549 cells was inhibited 83% by the ATl-selective blocker L158809, but was not reduced by the AT2—selective antagonist PD123319. Moreover, BLEO-induced apoptosis was also prevented by a neutralizing antibody specific for 50 ANGII (anti-ANGII), but not by an isotype-matched nonimmune immunoglobulin (N .S.IgG). Further, the total enzymatic activity of Caspase 3 was elevated by exposure of A549 cells to BLEO (Figure 3.5) but the increase was inhibited 52% by saralasin. These data suggested that BLEO induces ANGII synthesis de novo in primary AECs and A549 cells. Consistent with this theory, semiquantitative RTPCR for angiotensinogen (ANGEN) revealed more abundant ANGEN mRNA in primary AECs at 3 hours and especially at 20 hours after challenge with 25mU/ml BLEO (Figure 3.6A). In A549 cells (Figure 3.6B), 25mU/ml BLEO stimulated a significant increase in ANGEN mRNA that was detectable at 1 hour after addition of BLEO and increased by 7 hours. To determine if functional ANGEN mRNA is required for the apoptotic response to BLEO, phosphorothioated antisense or scrambled-sequence control oligonucleotides against ANGEN mRNA were transfected into rat AECs and A549 cells immediately before challenge with BLEO for an additional 20 hours. As shown in Figure 3.7, BLEO- induced apoptosis of primary AECs (Panel A) was inhibited by 83% by the ANGEN antisense but not by the scrambled control oligonucleotides (+scram). In A549 cells (B), the antisense also reduced BLEO-induced apoptosis by 84% but the scrambled oligonucleotide had no significant effect. Moreover, exposure to BLEO for 20 hours reduced the total cell number of A549 cells (attached plus detached, bottom panel) by 43%, but the ANGEN antisense prevented the BLEO-induced cell loss. 51 52 ow. Figure 3 . 1: Detection of apoptosis in primary AECs and A549 cells. A, B and C: Primary cultures of AECs were exposed to vehicle (A) or BLEO (B and C) at 25mU/ml for 20hr and were then fixed in 70% ethanol without washing (see Methods). Cells exhibiting chromatin condensation and nuclear fragmentation with propidium iodide (arrowheads, B) were scored as described earlier (27 and Methods). C: BLEO-exposed cells were fixed and prepared for in situ end labeling (ISEL, left) or TUNEL (right) of fragmented DNA (25); note colocalization of label (blue or brown, respectively) in nuclear fragments (arrowheads) identified by propidium in B. D, E and F: A549 cells exposed to vehicle (D) or BLEO at 25mU/ml (E) or lOOmU/ml (F) for 20hrs were fixed and prepared for immunolabeling for the active form of Caspase 3 (see Methods). Note labeling of active Caspase 3 (purple) in cells with either normal morphology (arrowheads, E) or in cells with condensed cytoplasm and nucleus (arrows, E and F). Note also reduced total cell number with increasing BLEO doses (E and F). 53 * AEC °/o of TOTAL FRAGMENT ED NUCLEI, Figure 3.2: Dose-dependent induction of nuclear fragmentation by bleomycin (BLEO) in primary rat AECs and blockade by inhibitors of caspases, endonucleases, ANG converting enzyme (ACE) and ANG-receptor interaction. Rat AECs were isolated and challenged with the indicated concentrations of BLEO on Day 2 of primary culture (see Methods). Putative inhibitors were added 30 minutes prior to addition of BLEO; nuclear fragmentation was scored as described in Fig.3.lB and Methods. ZVAD = ZVAD-fmk (N-benzylcarboxy—Val-Ala-Asp-[O-Me]-CH2F, 60uM); ATA = aurintricarboxylic acid lOuM); CAPTO = captopril (SOOng/ml); SARAL = saralasin (50ug/ml). Bars are the mean i S.E.M. of at least 4 observations; * = p<0.05 versus control (0.0 BLEO). 54 400 * AEC APOPTOTIC CELLS, % of CONTROL CTL BLEO BLEO BLEO 25mU/ml +DEVD +LOS Figure 3.3: Blockade of bleomycin-induced apoptosis in primary AECs by selective caspase or ANG receptor blockers. Rat AECs were isolated and challenged with 25mU/ml BLEO alone or in the presence of the Caspase 3-selective inhibitor DEVD—fmk (60uM) or the ANG receptor ATl-selective antagonist losartan (LOS, 10'6M). Control cultures (CTL) received BLEO and blocker vehicles only. Nuclear fragmentation was scored as described in Fig.1 and Methods. Bars are the mean i S.E.M. of at least 4 observations; * = p<0.05 versus control. 55 ‘AJ 3 500- * A549 g8 * * W , om 4°° BE 300m 88 EM 200' O 8:100- < 0- BLEOmU/ml: 0 25 25 25 25 25 25 25 X X. X X X X ‘72-. 06}, ‘99,}, ‘le <{g’60 70 o ‘74,. fiat, d’ 00 Figure 3.4: Inhibition of bleomycin-induced apoptosis of A549 cells by inhibitors of caspases or ANGII-receptor interaction. A549 cells were cultured to 8% confluence as described in Methods and were challenged with 25mU/ml BLEO in the presence or absence of the indicated compounds. Anti-ANGII = neutralizing antibody to ANGII (lug/ml); N.S.IgG = isotype matched nonimmune immunoglobulin (lug/ml); L158809 = ANG receptor ATl-selective antagonist (10'6 M); PD123319 = ANG receptor AT2- selective antagonist (lO‘6M). Other abbreviations and concentrations are the same as in Figs. 2 and 3. Nuclear fragmentation was scored as described in Fig.3.l and Methods. Bars are the mean : S.E.M. of at least 4 observations; * = p<0.05 versus control (0 dose). 56 25 * A549 * N O CASPASE 3 ACTIVITY, F.U./10‘ CELLS a: 10 ‘ 5 . o . CTL BLEO BLEO +SARAL Figure 3.5. Induction of caspase 3 activity by bleomycin and inhibition by an ANG receptor antagonist. A549 cells were challenged with BLEO (25mU/ml) for 20 hours in the presence and absence of the nonselective ANG receptor antagonist saralasin (SARAL, SOug/ml). Assay of Caspase 3 was conducted on adherent cells as described in Methods. * = p<0.05 versus control (CTL) and ** = p<0.05 versus BLEO. A AECS CT. 3hr 20hr p-MG . . B A549: CTL 1hr 3hr 7hr Figure 3.6. Semiquantitative RTPCR of angiotensinogen (ANGEN) mRNA in AECs afier bleomycin exposure. Primary cultures of rat AECs (A) and A549 cells (B) were exposed to BLEO (25mU/ml) for the indicated times and total RNA was isolated. RTPCR was performed as described before with primers specific for rat or human angiotensinogen (ANGEN), B-microglobulin (B-MG) or a-actin as control mRNAs. 58 AEC A MW nu nu Qv at 1 4019750.? .x. .mddmu 05.9—50.3. nu nu nu +anti- +scram sense +lipo CTL BLE CTL BLE BLE BLE 25mU +lipo /ml * A549 B 1 it I - nu nu nu nu nu nu nu nu no 00 AW at w—OMHZOU .«o .x. .mAAHU U—POPa—Om< nu * * * * Tu . w- m m m m o Emmogmnammoghé o\o .MHEDZ AAHU A . CTL CTL CTL +LIPO +AS +SCR °\o 500 3 400 d 300 U o 200 H 8 100 E 0 O CTL BLEO BLEO % +LIPO +LIPO +AS Figure 4.6. Antisense oligonucleotides reduce CatD immunoreactivity and inhibit bleomycin-induced apoptosis of AECs in vitro. A: Antisense (AS) or scrambled-sequence oligonucleotides (SCR) were transfected into primary cultures of rat AECs in the presence of lipofectin (LIPO, see Methods), without challenge with bleomycin (CTL). Western blotting of concentrated cell culture media was performed with CatD-specific antibodies; note decrease in immunoreactive CatD by AS but not SCR oligonucleotides. B: After antisense oligonucleotide transfection as in panel A, AECs were challenged with BLEO (25mU/ml) and harvested for detection of fragmented nuclei as in Figure 5. Bars are the mean i: S.E.M. of n = 3; * = p<0.01 versus untreated control (CTL) and ** = p<0.05 versus BLEO by ANOVA and Student-Newman-Keul’s test. 83 so-A AN GH produced, pmol/ml Fl-l4 Fl-14 F1-14 F1-14 AECs, AECS-l- +ACE +CatD +CatD no F 1-14 +ACE Fl-14 Figure 4.7. Production of ANGII from angiotensinogen fragment 1-14 in vitro. A: Angiotensinogen fragment 1-14 (F l-l4, 5uM) was incubated in vitro (without cells) with the indicated purified enzymes; ANGII was measured in the reaction buffer by specific ELISA (see Methods for details). Note production of ANGII by the combination of purified CatD + purified angiotensin converting enzyme (ACE), but not by either enzyme alone. B: Primary cultures of AECs were exposed to 5uM F1-l4, and ANGII was measured in the serum-free cell culture medium; note production of ANGII by AECs challenged with F1-14, but not by untreated AECs. 84 APOPTOTIC AECS, % 0 50 100 150 200 250 300 Figure 4.8. CatD-dependent induction of ABC apoptosis by angiotensinogen fragment 1- 14. Primary cultures of AEC were incubated with F1-14 as in Figure 4.7, in the presence or absence of pepstatin A (pepA, luM); CatD-specific neutralizing antibodies (CatD AB, 1:100) and the ANG receptor antagonists saralasin (SARAL, 50ug/ml) or L158809 (10'6 M). See Methods for details. Bars are the mean :1: S.E.M. of n = 3; * = p<0.001 versus untreated control (CTL) and ** = p<0.001 versus F1-14 by ANOVA and Student- Newman-Keul’s test. DISCUSSION A role for the aspartyl protease CatD in apoptosis has been shown previously in HeLa cells exposed to interferon-y, Fas ligand or TNF ~01 (Deiss et al., 1996) and in PA] ovarian cancer cells (Wu et al., 1998). The activity of CatD is upregulated by the apoptosis inducer adriamycin in PA] cells and in MLl leukemia cells and U1752 lung cancer cells (Wu et al., 1998). Although the aspartyl protease inhibitor pepstatin A could block apoptosis in these cell types, the exact mechanism(s) by which CatD participates in the execution of apoptosis is unclear. In accord with the known ubiquitous expression of CatD as a lysosomal protease (U chiyama et al., 2001), it has been suggested that this and other lysosomal proteases might be involved in the production of a bioactive molecule required for apoptosis of PC12 cells in response to trophic withdrawal (Isahara et al., 1999) Cathepsin D also is known to be one of the enzymes capable of proteolytically processing the liver-derived and serum-borne protein angiotensinogen to the peptide angiotensin I, a function normally performed in the serum by the kidney—derived enzyme renin (Filippatos et al., 2001). On the other hand, evidence from several nonpulmonary cell types has established CatD as the primary enzyme that converts angiotensinogen to ANGI within local “intrinsic” angiotensin systems, independently of renin (F ilippatos et al., 2001; Weber et al., 1995). Recent studies from this laboratory have shown that bleomycin-induced apoptosis of alveolar epithelial cells requires the autocrine synthesis of angiotensinogen, angiotensin II and it’s binding to ANG receptor ATl (Li et al., 2003). Those data were consistent with related studies showing that purified AN GII itself was a potent inducer of apoptosis 86 in ABC (Wang et al., 1999), and implied that AEC express enzymes capable of converting angiotensinogen to ANGII. Although the same study showed constitutive expression of angiotensin converting enzyme (ACE) by alveolar epithelial cells, the aspartyl protease required for providing the substrate for ACE (ANGI) in ABC was unknown. The data herein strongly suggest that CatD functions in this capacity in ABC; bleomycin- induced nuclear fragmentation and caspase 3 activity were significantly reduced by the aspartyl protease inhibitor pepstatin A (Figure 4.5) or by antisense oligonucleotides against CatD mRNA (Figure 4.6). In earlier investigations, bleomycin-induced apoptosis of AEC was competely blocked by specific angiotensin receptor antagonists or ANG- neutralizing antibodies (Li et al., 2003); this finding lead to the theory that autocrine generation of ANGII is required for ABC apoptosis regardless of the initiating stimulus (Uhal, 2002). In the light of those results, the finding that CatD antisense treatment did not completely block bleomycin-induced nuclear fragmentation (48%, Figure 4.6B) might indicate a potential role for additional protease(s) in angiotensinogen processing and subsequent AEC apoptosis. This interpretation is consistent with the finding that the protease inhibitor pepstatin A, which blocks all aspartyl proteases, also was incapable of complete blockade of nuclear fragmentation (76%, Figure 4.5) despite complete inhibition of CatD enzyme activity in AEC lysates (Figure 4.2). On the other hand, the antisense treatment, which is at least theoretically specific, did not completely eliminate immunoreactive CatD detected by western blotting (Figure 4.6A). Thus, it is difficult to know with certainty if the incomplete blockage of apoptosis is due to inefficient CatD knockdown or additional proteases activities. 87 Regardless, studies of angiotensinogen fragment 1-14 (Figures 4.7&8) are consistent with the theory that CatD is required for the conversion of angiotensinogen to ANGII by ABC, and with earlier work. For example, the finding that incubation of primary rat AECs with the fragment F1-l4 alone in serum-free culture medium (but without added enzymes) yielded significant production of ANGII (Figure 4.7) is consistent with the earlier demonstration of constitutive, albeit low, expression of both ACE and an unidentified aspartyl protease by primary AECs (Wang et al., 1999). Moreover, the complete abrogation of ABC apoptosis in response to angiotensinogen fragment F 1-14 by the nonselective and ATl-selective ANG receptor antagonists saralasin and losartan (Figure 4.8) confirmed that the induction of apoptosis was dependent on both the generation of ANGII from F1-14 and the binding of ANGII to receptor ATl. Those results also are consistent with our earlier demonstrations that AT1 receptor mediates AEC apoptosis in response to bleomycin (Li et al., 2003), amiodarone (Filippatos and Uhal, 2003; Uhal et al., 2003) or purified ANGII (Papp et al., 2002). Most important, the findings that ABC apoptosis in response to F1-14 was essentially abrogated by either pepstatin A or by CatD antibodies (Figure 4.8) strongly suggest that the conversion of angiotensinogen to ANGI, and subsequently ANGII to induce AEC apoptosis, is dependent on CatD activity. The upregulation of CatD activity by bleomycin in this study is consistent with the earlier findings that CatD is upregulated in alveolar epithelial cells in fibrotic human lung (Kasper et al., 1996) and is induced in the L132 lung cell line during apoptosis in vitro (Kasper et al., 1999). In other cell types, apoptosis inducers upregulate both CatD protein and mRNA, which suggests control of activation at the level of RNA (Wu et al., 1998). In contrast, RTPCR studies of ABC transcripts after bleomycin treatment failed to detect 88 changes in CatD mRNA (Figure 4.3) despite significant increases in CatD activity (Figure 4.2) and immunoreactive protein by western blotting (Figure 4.4). It is possible that the relatively few sampling times chosen for realtime analyses of CatD mRNA may have missed a transient but shortlived increase in the mRNA that might be revealed by a more exhaustive timecourse study. On the other hand, CatD is known to undergo activation by proteolytic mechanisms as well; in human U937 cells, CatD was shown to undergo processing of the inactive prepro- isoform (52kdal) to the active proCatD (48kdal) and an active 32kdal isoform, in response to autocatalysis of the enzyme induced by the direct binding of the apoptosis mediator ceramide (Heinrich et al., 1999). Consistent with those findings, western blotting of rat AEC lysates did reveal bleomycin- induced increases in several isoforms of apparent MW 44-52kdal. However, two of the isoforms shown to be increased in ABC media (52 and 48kdal, see “medium” in Figure 4.4) are larger than the primary isoform detected intracellularly in ABC (44kdal, “monolayer” in Figure 4.4). This finding argues against proteolytic processing alone as a mechanism of CatD activation in AEC. Thus, the exact mechanism(s) by which bleomycin upregulates CatD in ABC is unknown, but will pose an interesting problem for future studies. Pepstatin A-inhibitable CatD activity also was upregulated by amiodarone (Uhal et al., 2003) and TNF-alpha (Wang et al., 2000), both of which induce apoptosis in AECs (Uhal et al., 2003; Wang et al., 2000), but a determination of whether the requirement for CatD is universal to all proapoptotic stimuli for AEC will require further investigation. In summary, bleomycin upregulated CatD enzymatic activity and immunoreactive protein in primary cultures of rat alveolar epithelial cells (AEC). Apoptosis of cultured AEC in 89 response to bleomycin was significantly inhibited by the aspartyl protease inhibitor pepstatin A or by antisense oligonucleotides against CatD mRNA. The same inhibitors also prevented the enzymatic processing of a synthetic fragment of angiotensinogen (amino acids 1-14), and completely blocked AEC apoptosis in response to the same peptide. These data are consistent with earlier studies showing that apoptosis of AEC in response to bleomycin requires the autocrine synthesis and proteolytic processing of angiotensinogen to angiotensin II, and suggest that the proteolytic processing requires CatD. The data herein also suggest that blockade of CatD and other aspartyl proteases might provide a potential strategy for preventing AEC apoptosis and lung injuries that involve this mode of cell death. 90 Chapter 5 ESSENTIAL ROLES FOR ANGIOTENSIN RECEPTOR ATla in BLEOMYCIN- INDUCED APOPTOSIS AND LUNG F IBROSIS IN MICE Xiaopeng Li, Heather Rayford, and Bruce D. Uhal From the Department of Physiology, Michigan State University, East Lansing, Michigan This chapter is published in: Am J Pathol 2003, 163(6): 2523-2530 91 ABSTRACT: Apoptosis of alveolar epithelial cells (AECs) has been implicated as a key event in the pathogenesis of lung fibrosis. Recent studies demonstrated a role for the synthesis and binding of angiotensin II to receptor ATl in the induction of AEC apoptosis by bleomycin (BLEO) and other proapoptotic stimuli. On this basis we hypothesized that BLEO-induced apoptosis and lung fibrosis in mice would be inhibited by the ATI antagonist losartan (LOS) or by targeted deletion of the AT1 gene. Lung fibrosis was induced by intratracheal administration of BLEO (1 U/kg) to wild-type C57BL/6J mice. Co-administration of LOS abrogated BLEO-induced increases in total lung caspase 3 activity detected 6 hours after in vivo administration and reduced by 57% BLEO-induced caspase 3 activity in blood-depleted lung explants exposed to BLEO ex vivo (both P < 0.05). Co-administration of LOS in vivo reduced DNA fragmentation and immunoreactive caspase 3 (active form) in AECs, measured at 14 days after intratracheal BLEO, by 66% and 74%, respectively (both P < 0.05). LOS also inhibited the accumulation of lung hydroxyproline by 45%. The same three measures of apoptosis and lung fibrosis were reduced by 89%, 85%, and 75%, respectively (all P < 0.01), in mice with a targeted disruption of the ATla receptor gene (C57BL/6J-Agtrla‘mlunc). These data indicate an essential role for angiotensin receptor ATla in the pathogenesis of BLEO-induced lung fibrosis in mice and suggest that AT1 receptor signaling is required for BLEO-induced apoptosis of AECs in mice as it is in rat and human AECs. 92 INTRODUCTION: Idiopathic pulmonary fibrosis is a progressive and often fatal human disease characterized by infiltration of inflammatory cells into interstitial and alveolar spaces, ongoing damage to the lung parenchyma, fibroblast proliferation, and accumulation of interstitial collagens (Selman et al., 2001). Ongoing evaluations of both older and newer data have lead to the recent characterization of idiopathic pulmonary fibrosis as a disease of abnormal wound repair, in which abnormalities in epithelial-mesenchymal interactions are of key importance (Selman et al., 2001; Gauldie et al., 2002). This evolving theory about the pathogenesis of idiopathic pulmonary fibrosis is, in some respects, a revival of the hypothesis first put forth by Haschek and Witschi (Haschek and Witschi, 1979) and Adamson and colleagues (Adamson et al., 1988) that the severity of the fibrogenic response in the lung is directly related to the severity of epithelial injury. A growing body of evidence suggests that alveolar epithelial cell (AEC) death by apoptosis is a key event in the initiation and progression of lung fibrosis. In mice exposed to bleomycin (BLEO) by intratracheal instillation, up-regulation of the receptor Fas on lung epithelial cells and F as ligand on infiltrating lymphocytes were associated with DNA fragmentation in epithelia and subsequent accumulation of collagens (Hagimoto et al., 1997a). Intratracheal instillation of Fas—activating antibodies caused epithelial cell apoptosis and subsequent collagen accumulation, the severity of which was proportional to the amount of Fas-activating antibody instilled (Hagimoto et al., 1997b). Other investigators have shown that BLEO itself induces apoptosis of AECs, which precedes the deposition of collagens (Wang et al., 2000). More importantly, several groups have found that blockage of epithelial apoptosis with caspase inhibitors administered in vivo can prevent 93 BLEO-induced lung cell apoptosis and the subsequent accumulation of lung collagens (Wang et al., 2000; Kuwano et al., 2001) Recent work from this laboratory has shown that exposure of cultured AECs to F as ligand (Wang et al., 1999), tumor necrosis factor-a (Wang et al. R, 2000), or BLEO (Li et al., 2003) all induce expression of angiotensinogen mRNA and protein, and its cleavage to the peptide angiotensin II (ANGII). Moreover, apoptosis of cultured AECs in response to these apoptosis inducers was abrogated by antagonists of ANG receptor AT1, such as losartan (LOS) or L158809 (Li et al., 2003; Uhal et al., 2003; Filippatos and Uhal, 2003) For all these reasons, it was hypothesized that angiotensin receptor AT1 is essential for ABC apoptosis and lung fibrosis in vivo. To test this theory, normal mice and mice deficient in ANG receptor ATla, the AT1 subtype expressed in lung (Burson et al., 1994), were subjected to intratracheal BLEO administration and quantitation of apoptosis and lung collagens. We report here the prevention of both BLEO-induced AEC apoptosis and lung collagen accumulation in mice by administration of the AT1-selective receptor antagonist LOS or by targeted deletion of the ATla receptor gene. 94 MATERIALS AND METHODS Reagents and Materials The AT1-selective antagonist LOS was obtained from Merck and Co., West Point, PA. Alkaline phosphataseconjugated streptavidin, digoxigenin-labeled deoxyuridine trisphosphate (dig-dUTP), and biotinylated deoxyuridine trisphosphate (bio—dUTP) were obtained from Boehringer Mannheim, Indianapolis, IN. BLEO was obtained from Sigma Chemical Co., Saint Louis, MO. Reagents for detection of alkaline phosphatase and other secondary reagents for in situ end labeling (ISEL) of DNA or Western blotting were from sources described earlier (Wang et al., 2000). All other materials were of reagent grade and were obtained from Sigma Chemical Co. Animals, Induction of Pulmonary Fibrosis, and Surgical Procedures All mice were obtained from The Jackson Laboratories, Bar Harbor, ME, and were housed in a satellite facility of University Laboratory Animal Resources, Michigan State University. Control animals were wild-type C57BL/6J mice used at 7 to 8 weeks of age. Some experiments also used mice of the same genetic background but with a targeted disruption in the ANG receptor ATla gene (C57BL/6J-Agtr1a’mwm) that removes a portion of the coding region sufficient to eliminate specific binding of AT1-selective agonists in all organs tested (Ito et al., 1995). Heterozygous animals were used on the basis of availability at the same age and body weight as wild types. Induction of Lung Injury and Fibrosis 95 Animals under pentobarbital anesthesia received a single intratracheal instillation of bleomycin sulfate (BLEO) at 1 U/ kg body weight, in 50 pl of sterile saline. The 50-111 dose was instilled at end-expiration, and the liquid was followed immediately by 300 pl of air to ensure delivery to the distal airways. Control animals were instilled with an equal volume of sterile saline. In some studies the AT1 receptor antagonist LOS was added to the intratracheal instillate at 20 umol/L; the same animals also received daily intraperitoneal injections of LOS at 10 mg/kg in sterile saline throughout the test interval. Other treatment groups received daily intraperitoneal sham injections of the saline alone. LOS or sham injections were continued for 14 days after instillation of BLEO, at which point all animals were sacrificed for histology, detection of collagen or DNA fragmentation, and caspase-3 activation in epithelial cells. Surgical Procedures Immediately before sacrifice, animals were given intraperitoneal injections of sodium pentobarbital and the trachea was cannulated. The left lung was ligated at the hilus, excised distal to the ligation, and immediately frozen in liquid N2 for hydroxyproline assay of total collagen (see below). The remaining lung tissues were carefully removed and were instilled with 4% paraforrnaldehyde in phosphate-buffered saline (PBS) at 20 cm of H20 constant pressure, then immersed in the same fixative for 30 minutes followed by storage in 70% ethanol. The fixed tissues were washed with PBS three times for 15 minutes and were then embedded in paraffin. Five 11m sections of lung were deparaffinized by passing through xylene, xylene: alcohol 1:], 100% alcohol, and 70% alcohol for 10 minutes each. Ethanol was removing by rinsing with distilled water. 96 Lung Explant Culture Explants of 1 mm2 were prepared by mincing of blood depleted (PBS-perfused) mouse lung, and were cultured in Transwell polycarbonate inserts (3.0um pore; Costar, Corning, NY) under a thin layer (1 mm) of Dulbecco’s modified Eagle’s medium cell culture medium to facilitate gas exchange (Taylor et al., 2000). All explants were obtained from normal mouse lung that was PBS-perfused in situ before excision of the lungs. After excision of the lungs, treatment with BLEO or LOS was initiated ex vivo by intratracheal instillation of BLEO at 25 mU/ml in 300 pl of sterile Dulbecco’s modified Eagle’s medium (+/- LOS at 10’6 mol/L). The culture medium for explants also contained BLEO at 25 mU/ml, +/- LOS at 10'6 mol/L. Explants were harvested by transfer into liquid N2 and storage at -80°C until assay. Identification and Quantitation of Apoptotic Cells and Total Lung Caspase 3 Activity Localization of DNA Fragmentation ISEL of fragmented DNA was conducted by a modification of the method of Mundle and colleagues (Mundle et al., 1994). Briefly, ethanol was removed from deparaffinized lung sections by rinsing in distilled water for at least 10 minutes. The slides were then placed in 3% hydrogen peroxide (Sigma Chemical Co.) for 30 minutes at 20°C, rinsed with PBS, and incubated with Proteinase K (Sigma) in standard saline citrate for 15 minutes at 37°C. Samples were rinsed once in water, three times in 0.15 mol/L PBS for 4 minutes each, and were then incubated in standard saline citrate (0.3 mol/L NaCl and 30 mmol/L 97 sodium citrate in water, pH 7.0) at 80°C for 20 minutes. After four rinses in PBS and four rinses in buffer A (50 mmol/L Tris/HCl, 5 mmol/L MgCl, 10 mmol/L B- mercaptoethanol, and 0.005% bovine serum albumin in water, pH 7.5), the sections were incubated at 18°C for 2 hours with ISEL solution (0.001mmol/L digoxigenin-dUTP; 20 U/ml DNA Polymerase I; and 0.01 mmol/L each of dATP, dCTP, and dGTP in buffer A). Afterward the sections were rinsed thoroughly five times with buffer A and three additional times in PBS. Detection of incorporated dUTP was achieved with by incubation for 2 hours at 37°C with AP-conjugated antidigoxigenin (Boehringer Mannheim) at 1/400 dilution. Bound AP-antibody was then detected with the Fast Blue chromogen system and the sections were mounted with F luoromount solution (Southern Biotechnology, Birmingham, AL). Immunohistochemistry (IHC) for Activated Caspase 3 IHC was performed with an antibody that recognizes only the active form of the enzyme (BioVision, MountainView, CA). Deparaffinized lung sections were blocked with a solution of 3% bovine serum albumin in PBS for 1 hour; the primary antibody was then applied overnight at 4°C in 3% bovine serum albumin/PBS. After washing in PBS, the antibody was detected with a biotin-conjugated secondary antibody and avidin-linked chromogen system. Type II pneumocytes were identified with the anticytokeratin antibody MNF 116, an established marker of type II cells (Fehrenbach et al., 2000). Detection of mouse lung antigens with this mouse monoclonal antibody was achieved with the Mouse-on-Mouse Iso-IHC kit (InnoGenex, San Ramon, CA) according to the manufacturer’s instructions. 98 For quantitation of ISEL- or caspase 3-positive epithelial cells, the number of positive cells within the surfaces of the alveolar walls was counted in a minimum of six randomly selected x 400 microscopic fields per lung section. Positive cells within the alveolar airspaces, or otherwise clearly not within the surface of the alveolar wall, were not scored. The counts of positive nuclei per field were expressed as a percentage of the total number of nuclei in the same microscopic field. Sections from each of at least five mice per treatment group were analyzed by an investigator blinded to sample identity. Caspase 3 Enzyme Activity Assay of total lung caspase-3 enzyme activity was conducted with a commercially available kit (Molecular Probes, Eugene, OR). Fast-frozen lung was homogenized in assay kit buffer and was analyzed according to the manufacturer’s instructions on a Biotek FL600 fluorescence plate reader. In all samples, specificity of the reaction for caspase 3 was verified by abrogation of the signal with a caspase 3-selective irreversible inhibitor (data not shown). Quantitation of Lung Collagen For quantitation of total lung collagen, tissues frozen in liquid N2 were dried to constant weight in preweighed tubes at 80°C. The weighed dry tissue was hydrolyzed in 6 N HCl and was subjected to determination of hydroxyproline as described earlier by Woessner (Woessner, 1961) The efficiency of the hydrolysis was verified with rat-tail collagen by comparison to standard hydroxyproline (Sigma Chemical Co.). Attention: Images in this dissertation are presented in color. 99 RESULTS Quantitation of Apoptosis and Lung Injury On the basis of earlier work with rat models (Wang et al., 2000; Li et al., 2003), we hypothesized that BLEO would induce apoptosis of mouse lung AECs that might be detected in situ by end labeling of fragmented DNA or by IHC for the active form of caspase 3. Consistent with this expectation, intratracheal instillation of BLEO caused increased ISEL and positive caspase 3 IHC within cells in the surfaces of alveolar corners, the expected locations of type II pneumocytes (Figure 5.1; B, F, and G, arrows; see subsequent figures for quantitation). Many ISEL- or caspase 3-positive cells colocalized with positive immunoreactivity to monoclonal antibody MNF116 (Figure 5.1C), an established marker of type II pneumocytes (Fehrenbach et al., 2000). Interestingly, MNFl l6 immunoreactivity was observed in relatively normal regions of BLEO-exposed lung (Figure 5.1D, right) but not in more severely affected regions (Figure 5.1D, left), consistent with the elimination of type II cells in these areas. Co- administration of the AT1 receptor antagonist LOS with the BLEO (Figure 5.1H) significantly reduced caspase 3 IHC (see below for quantitation). The instillation of BLEO also resulted in histological changes typical of BLEO-induced lung fibrosis by day 14 after instillation (Figure 5.2). These include the infiltration of inflammatory cells, thickening of alveolar walls, and collagen accumulation (Figure 5.2B); for quantitation, see Figures 5.5 and 5.7 below. Again, the co-administration of LOS with the BLEO (Figure 5.2C) significantly reduced the alterations in lung morphology and collagen deposition (see below). 100 Inhibition of Apoptosis and Collagen Deposition by an AT1 Antagonist Apoptosis also was detected as an increase in the total activity of caspase 3 in lung tissue, measured by enzyme assay of lung homogenates. As early as 6 hours after instillation of BLEO intratracheally (Figure 5.3A), lung caspase-3 activity was increased 100%, but the increase was prevented by co-administration of the AT1 receptor antagonist LOS (see Materials and Methods). In Figure 5.3B, BLEO also increased caspase-3 activity when applied in vitro at 25 mU/ml to mouse lung explants that were depleted of blood before explant culture. Application of BLEO in vitro increased caspase-3 activity in the explants by nearly 100% in 24 hours (P <0.05), but LOS (10'6 mol/L) inhibited the increase by 57%. The ability of LOS to inhibit lung epithelial apoptosis was also observed in vivo at 14 days after BLEO administration. In Figure 5.4A, intratracheal BLEO increased the abundance of ISEL-positive cells by ll-fold (P < 0.01), but LOS inhibited the increase by 66% (P < 0.05). Similarly, intratracheal BLEO increased the number of caspase 3- positive cells by 25-fold (Figure 5.4B, P < 0.01), but LOS blocked the increase by 74% (P < 0.05). Measurement of lung collagen accumulation in the same animals by hydroxyproline assay (Figure 5.5) revealed an increase in total lung collagen of 56% by 14 days afier intratracheal BLEO, but LOS reduced the increase by 45%, to a value not significantly different from the control (CTL). Inhibition of Apoptosis and Collagen Deposition by AT1 Gene Deletion Receptor subtype ATla is the AT1 isoform expressed in the lungs of mice (Burson et al., 1994). To test the hypothesis that angiotensin receptor AT1 is essential for BLEO- induced epithelial apoptosis and lung fibrogenesis, heterozygous ATla-null mice were 101 exposed to intratracheal BLEO in the same manner as wild-type mice of the same genetic background. In Figure 5.6, the deletion of one allele of the ATla gene (+/-) reduced BLEO-induced ISEL by 89% (Figure 5.6A) and inhibited BLEO-induced caspase 3 IHC by 85% (Figure 5.6B), both relative to the response in wild-type mice (**, P <0.01). When the susceptibility of the same mice to BLEOinduced fibrosis was measured, heterozygous ATla-null mice did not exhibit a statistically significant increase in lung hydroxyproline at 14 days after intratracheal BLEO (Figure 5.7A), in contrast to wild- type mice. Expression of the hydroxyproline data as the absolute amount of collagen per left lung (Figure 5.7B) suggested that unchallenged ATla +/- mice, of the same age and body weight as the wild types, have more total collagen per lefi lung at baseline relative to wild-type mice, but the difference was not statistically significant. 102 103 Figure 5.1. Detection of DNA fragmentation, activation of caspase 3, and alveolar type II pneumocytes in mouse lung. Deparaffinized lung sections were prepared from mice instilled intratracheally 14 days earlier with sterile saline (A, C, and E) or BLEO (B, D, F, and G). The sections were subjected to ISEL of fragmented DNA (A and B) or IHC with antibodies against the active form of caspase 3 (E—H) or with the type II cell- specific antibodyMNFll6 (C and D). G: Higher magnification of active caspase 3 labeling in F. H: Active caspase 3 labeling in mice treated with BLEO and LOS, an AT1 receptor antagonist. Note ISEL and active caspase 3 labeling in cells in the comers of alveolar walls in the lungs of BLEO-treated mice (B, F, and G, arrowheads) but not in saline-treated mice (A and E) or in mice treated with BLEO and LOS (H). Note also the co-localization of MNF116 (C) with anti-caspase 3 IHC (G) or ISEL (B) in BLEO- treated lungs. D reveals labeling of the type 11 cell marker MNF116 in relatively normal regions of BLEOtreated lung (D, right) but not in more severely affected regions (D, lefi). See text for details. Original magnifications: X400 (A, B, C, G); X200 (D); X100 (E, F, H). 104 Figure 5.2. Histology of mouse lungs at 14 days after instillation of BLEO. A—C: Hematoxylin and eosin preparations of mouse lung instilled intrauacheallyl4 days earlier with sterile saline (A), BLEO (B), or BLEO and LOS (C). See text and Materials and Methods section for details. Original magnifications X200. 105 mU/mg protein Caspase 3 Activity, Caspase 3 Activity, mU/mg protein CTL BLEO BLEO +LOS Figure 5.3. The AT1—selective receptor antagonist LOS inhibits BLEO-induced activation of caspase 3. A: BLEO was instilled intratracheally into normal mice with and without LOS in the intratracheal instillate (see Materials and Methods). Six hours later, the lungs were perfused to remove blood, excised, and the enzymatic activity of caspase 3 was measured in lung homogenates. B: Lung explants were prepared from normal mouse lung tissue perfused before excision (see Materials and Methods). BLEO (25 mU/ml) was applied in serum-free culture medium for 24 hours in the presence or absence of LOS (10’6mol/L). Bars are the means 3: SEM ofn = 6; *, P < 0.05 versus control (CTL); ** , P < 0.05 versus BLEOiLOS by analysis of variance and Student-Newman-Keul’s test. 106 3518 of Alveolar Walls,% III 6 Active Caspase 3+ cells ISEL+ cells in Surface in Alveolar Walls,% C TL BLE 0 BLE 0+L0 S Figure 5.4. AT1 receptor blockade inhibits DNA fragmentation and caspase-3 activation in lung epithelial cells 14 days after BLEO instillation. Normal mice were given a single intratracheal instillation of BLEO in the presence or absence of LOS in the instillate. LOS also was administered thereafter daily intraperitoneally. Fourteen days later, lung sections were prepared and labeled by ISEL (A) or by IHC for the active form of caspase 3 (B). Labeling was quantitated in cells within the alveolar surfaces (see Figure 1C). Bars are the means :1: SEM of n = 6; *, P < 0.01 versus control (CTL); **, P < 0.05 versus BLEO by analysis of variance and Student-Newman-Keul’s test. 107 i—s {xi-36%: GGOGG HP/Left Lung, 11g 6 CTL BLEO BLEO +LOS Figure 5.5. AT1 receptor blockade inhibits lung collagen accumulation at 14 days after BLEO instillation. Normal mice were administered BLEO in the presence or absence of LOS as described in Figure 5.3. Fourteen days later, total lung collagen was determined by assay of total hydroxyproline (HP) in hydrolyzed lung tissue (see Materials and Methods). Bars are the means i SEM of n = 6; *, P < 0.05 versus control (CTL) by analysis of variance and Student-Newman-Keul’s test. 108 OF ALVEOLAR WALLS, % CASP3+ CELLS IN SURFACE ISEL+ CELLS IN SURFACES OF ALVEO LAR WALLS, “/0 Bleo: - + - + Figure 5.6. Mice deficient in angiotensin receptor ATla exhibit reduced DNA fragmentation and caspase-3 activation in lung epithelial cells 14 days after BLEO instillation. Normal [wild type (w.t.)] or heterozygous ATla knockout mice (+/-) were administered BLEO intratracheally as in Figure 5.3. Fourteen days later, lung sections were prepared and labeled by ISEL (A) or by IHC for the active form of caspase 3 (B), which were quantitated as described in Figure 5.4 and Materials and Methods. Bars are the means i SEM of n = 5; *, P < 0.001 versus wild-type unchallenged (w.t. - BLEO); **, P < 0.01 versus wild-type challenged (w.t. + BLEO) by analysis of variance and Student-Newman-Keul’s test. 109 w.t. w.t. +/- +/- .A * § E % of Control g I-IP/Left Lung, pg HP/Left Lung é O Bleo: - + - + Figure 5.7. Mice deficient in angiotensin receptor ATla exhibit reduced lung collagen accumulation in response to BLEO instillation. Normal [wild type (w.t.)] or heterozygous ATla knockout mice (+/-) were administered BLEO intratracheally as in Figure 5.3. Fourteen days later lung tissue was fast-frozen, hydrolyzed, and total collagen was measured by hydroxyproline assay (HP) as described in Materials and Methods. A: HP data are expressed as a percentage of the corresponding control (— BLEO). B: Data are expressed as the absolute amount of HP per left lung. Bars are the means i SEM of n = 5; *, P < 0.01 versus untreated (- BLEO) by analysis of variance and Student-Newman- Keul’s test. 110 DISCUSSION Inhibitors of angiotensin-converting enzyme (ACE) or antagonists of ANG receptor AT1 have been shown to have anti-apoptotic and anti-fibrotic effects in the heart (Sun and Weber, 1998), kidney (Mezzano et al., 2001), and liver (Yoshiji et al., 2001). The first reports of anti-fibrotic actions in the lung by the ACE inhibitor captopril, published many years ago (Molteni et al., 1985; Ward et al., 1990), were recently extended by the demonstration that the AT1 antagonists LOS and L158809 have even more potent anti- fibrotic potential in the lungs than do ACE inhibitors (Molteni et al., 2000). The present work extends those observations by showing that at least one of the mechanisms by which AT1 antagonists act is through the inhibition of apoptosis in AECs. Other potential mechanisms by which AT1 antagonists might act on the lung in vivo, a topic recently reviewed by Marshall (Marshall, 2003), include decreased vascular tone, decreased vascular permeability, and altered fibroblast activity. At least some of these actions could be envisioned to be related to the ability of ACE inhibitors or AT1 antagonists to reduce blood pressure. Indeed, the AT1-null mice used here were shown earlier to exhibit reductions in systemic blood pressure of about 12 mm Hg for heterozygous animals at baseline. Thus, it is possible that some of the anti-fibrotic actions of LOS or ATla deletion might be related to lowered systemic or pulmonary hydrostatic pressures, and the data herein do not strictly exclude that possibility. On the other hand, the ability of LOS to prevent caspase-3 activation by BLEO in cultured lung explants (Figure 5.3B) argues against the involvement of decreased blood pressure in the inhibition of apoptosis because no hydrostatic pressure changes occur in explants manipulated ex vivo. lll Moreover, the induction of LOS-inhibitable caspase 3 activities in lung explants, depleted of blood by previous PBS perfirsion, and argues against a primary role for blood-derived cells in the initiation of the apoptosis. This experiment also supports the theory that the inhibitory effect of LOS on apoptosis was not mediated by an indirect action on infiltrating inflammatory cells. The initial protocol for the reported in vivo studies was designed to determine whether blockade or deletion of the AT1 receptor, throughout the time course of the 14- day BLEO model, was capable of inhibiting or blocking the apoptotic and fibrotic responses. The success of this strategy, particularly in light of the many known functions of angiotensin discussed in preceding paragraphs, raises the interesting question of whether the blockage of fibrogenesis was due to the acute or delayed consequences of AT1 blockade. Although a time course study of various LOS administration protocols was not performed, quantitation of the number of erythrocytes reaching the alveolar airspaces by 6 hours afier BLEO (a crude index of lung barrier collapse) suggested that LOS did not prevent acute, transient barrier collapse (data not shown) despite its ability to reduce caspase 3 activation at the same sampling time (Figure 5.3). This observation, although very preliminary, is consistent with the theory that the blockage of apoptosis in AECs is a key to the subsequent blockade of collagen deposition. In contrast, blockage of receptor AT1 may also inhibit mitosis of lung fibroblasts (Marshall et al., 2000) and reduce collagen synthesis by the same cells (Marshall et al., 2004) relatively delayed effects that might be independent of AEC apoptosis at early time points. Moreover, endothelial cells also express receptor AT1 and undergo apoptosis in response to angiotensin, albeit at relatively high concentrations (Dimmeler et al., 1997; Li et al., 1999) and other cell types resident in the lung are known 112 to respond to angiotensin in ways currently under intense study (Harrison et al., 2003). Thus, it is possible that the acute early effects of AT1 blockade on AEC apoptosis are not necessary for inhibition of collagen deposition at later time points, and this study does not exclude that possibility. On the other hand, earlier work has shown that AECs undergoing apoptosis in response to F as ligand, tumor necrosis factor-a or BLEO begin secreting angiotensin into the extracellular space within hours of exposure (Wang et al., 1999; Wang et al., 2000; Li et al., 2003), at least in vitro. Those studies also showed that the autocrine production of angiotensin and its binding to receptor AT1 on AECs were required for apoptosis in response to these agents; this mechanism can explain the ability of LOS to block AEC apoptosis in vivo in the present study. Moreover, previous studies with apoptosis inhibitors support the contention that the acute apoptotic response is a pivotal event in the BLEO model. Wang and colleagues (Wang et al., 2000) showed that the ACE inhibitor captopril or the caspase inhibitor ZVAD-fmk had essentially equal ability to block the appearance of apoptotic epithelial cells in rats exposed to intratracheal BLEO and to prevent subsequent collagen deposition (Wang et al., 2000). That report, which was confirmed by Kuwano and colleagues (Kuwano et al., 2001) in studies of mice exposed to BLEO and/or ZVADfmk, suggested that the blockade of fibrogenesis by captopril was indeed related to inhibition of apoptosis, rather than the many other effects of ACE inhibition in vivo (Marshall, 2003). Later work confirmed that the ZVAD compound had no inhibitory effect on angiotensin converting enzyme itself (F ilippatos and Uhal, 2003). Thus, the present data are consistent with the ability of ACE inhibition by captopril to block both AEC apoptosis and collagen deposition in rats (Wang et al., 2000), and extend this concept to angiotensin receptor blockade in mice. The data herein ll3 also are in agreement with recent reports that LOS inhibits BLEO-induced collagen deposition in rat lung (Fang et al., 2002) and that ATla-null mice show reduced liver fibrosis in response to carbon tetrachloride (Kanno et al., 2003). The AT1 receptor is expressed as two isoforms, ATla and Ale, for which no selective antagonists have yet been developed (Filippatos et al., 2001). Subtype ATla is known to be expressed in the lungs of mice, but Ale was not detected in mouse lung by reverse transcriptase- polymerase chain reaction (Burson et al., 1994). Although it is possible that cells of minor abundance in the lung, such as type II cells, might express Ale in quantities not detected in earlier studies, the primary isolates of type II pneumocytes from Wistar rats did not reveal Ale expression by reverse transcriptase- polymerase chain reaction despite the use of two different primer sets and high-amplification cycle numbers (data not shown). In any case, the finding that deletion of only one allele of the ATla gene significantly supports the notion that ATla is the only active AT1 receptor subtype on the alveolar epithelium of mice. In an earlier report describing the mechanisms by which angiotensin induces apoptosis in primary cultures of AECs, Papp and colleagues (Papp et al., 2002) showed that blockage of AT1 signaling through protein kinase C (PKC) with the specific PKC inhibitor chelerythrin could attenuate the apoptotic response to angiotensin. This finding is consistent with the known role of PKC in AT1 signaling in a variety of cell types (Li et al., 1999), but the pathways from PKC to the effector caspase 3, which is also required for this response (Papp et al., 2002), are currently unknown. Given that ABC apoptosis in response to Fas ligand, tumor necrosis factor- a, or BLEO all require the autocrine production and binding of angiotensin to AT] (Wang et al., 1999; Wang et al., 2000; Li “4 et al., 2003), the report of Papp and colleagues (Papp et al., 2002) suggests that PKC inhibitors would also block AEC apoptosis in response to these agents in vivo as well. This prediction was not tested in the present study, but will be an interesting topic for future inquiry. In summary, BLEO-induced apoptosis of lung epithelial cells in mice was significantly inhibited by the AT1- selective angiotensin receptor antagonist LOS or by targeted deletion of the gene for angiotensin receptor subtype ATla. Both methods of reducing AT1 action also reduced or abrogated lung collagen accumulation in response to BLEO challenge. These data agree with earlier demonstrations of the anti-fibrotic action of ACE inhibitors and AT1-selective antagonists in rat models of lung fibrosis, and with in vitro studies showing a role for receptor AT1 in mediating apoptosis of AECS. They also suggest the possibility that AT1 antagonists may hold potential for the treatment of lung fibrosis in humans; this possibility is supported by the recent finding that patients with pulmonary fibrosis have a higher frequency of the D allele of angiotensin-converting enzyme (Morrison et al., 2001), a deletion polymorphism that confers higher levels of ACE. 115 Chapter 6 ATTENUATION OF BLEOMYCIN-INDUCED PULMONARY FIBROSIS BY INTRATRACHEAL ADMINISTRATION OF ANTISENSE OLlGO- NUCLEOTIDES AGAINST ANGIOTENSINOGEN mRN A Xiaopeng Li, J iaju Zhuang, Heather Rayford, Huiying Zhang, Ruijie Shu and Bruce D. Uhal Department of Physiology, Michigan State University, East Lansing, Michigan, USA 116 ABSTRACT: Apoptosis of alveolar epithelial cells (AECs) is believed to be critical for the development of bleomycin-induced pulmonary fibrosis. Angiotensin II is generated by AECs undergoing apoptosis and by human lung myofibroblasts isolated from IPF patient biopsies (Am J Physiol. 277:L1245-L1250, 1999; Am J Physiol. 277:L1158-L1164, 1999). Previous studies showed that apoptosis of alveolar epithelial cells in response to bleomycin could be abrogated by antisense oligonucleotides against angiotensinogen (ANGEN) mRNA and requires angiotensin II synthesis de novo (Am J Physiol Lung Cell Mol Physiol 284: L501-L507, 2003). Here we hypothesized that blockade of local pulmonary ANG II synthesis by intratracheal administration of antisense oligonucleotides against ANGEN mRNA might attenuate bleomycin-induced apoptosis of AECs and pulmonary fibrosis. Male Wistar rats received 8 U/kg of bleomycin sulfate or vehicle intratracheally. Endogenous lung ANGEN was upregulated in vivo as early as 3 hours after bleomycin instillation in rat lungs by RT-PCR, in situ hybridization and immunohistochemistry staining methods. ANGEN mRN A and angiotensin peptides were localized in alveolar wall cells in the alveolar comers, tentatively identified as type II cells, and also colocalized with alpha-Smooth Muscle Actin (a-SMA). Labeled antisense administered by intratracheal instillation was specifically accumulated in the lung compared to liver and kidney, and localized primarily in the epithelium of airways and cells within alveolar walls. Intratracheal instillation of 75ug antisense reduced bleomycin—induced pulmonary fibrosis detected by hydroxyproline assay; decreased ANGEN and active caspase-3 protein detected by western blot and reduced the ISEL positive cells, but had no effect on the serum ANG II level. These data are consistent with 117 the hypothesis that lung-derived ANGEN is involved in bleomycin-induced pulmonary fibrosis. 118 INTRODUCTION : Idiopathic Pulmonary fibrosis (IPF) is a pathological condition resulting from injury to the lung and an ensuing fibrotic response leading to thickening of the alveolar walls and the obliteration of the alveolar space without known etiology (F onseca C et al., 2000). In IPF the normal architecture and functional integrity of the lung are destroyed. The main histological features of the fibrotic lung are persistent and unrepaired epithelial damage, proliferation and accumulation of fibroblast/ myofibroblast cells, and increased collagen deposition (Selman et al., 2001). Unfortunately, despite years of research on its pathogenesis, the treatment of IPF has not been successful. The traditional therapeutic approaches using potent anti-inflammatory agents supplemented with an immunosuppressive agent could only offer a marginal benefit at best (King et al., 2000). Recent NHLBI Workshops have concluded that our current understanding of the pathogenesis of the IPF is incomplete (Mason et al., 1999; Crystal et al., 2002). Current evolving hypothesis about pathogenesis is that IPF results from the epithelial microfoci injury and a failure of reepithelization (Selman et al., 2001, Gauldie et al., 2002). Haschek and Witschi first proposed this hypothesis more than 20 years ago, who believe that epithelial damage drives fibrogenesis and efficient epithelialization would prevent fibrogenesis (Haschek et al., 1979; Witschi H., 1990). Recent studies in our lab and others using bleomycin-treated rat and mouse models strongly suggested a role of epithelial apoptosis as the profibrotic event in fibrogenesis. The evidences include: First, apoptosis of AECs was found in both patients with IPF (Uhal, et al., 1998) and animal models (Hagimoto, et al., 1997). Second, induction of 119 apoptosis in the epithelium is sufficient to initiate a fibrotic response (Hagimoto N. et al., 1997). Third, several labs showed that blockade of the apoptosis could prevent fibrotic response (Wang, et al., 2000; Kuwano et al., 2001). These studies support the “Witschi Hypothesis” regarding the pathogenesis of pulmonary fibrosis that is PF results from the epithelial injury and failure to reepithelization. Results obtained from studies on human lungs are consistent with those on animal models. Fragmented DNA, the hallmark of apoptosis, in bronchiolar and AECs were found in lung biopsies from patients with IPF (Kuwano et a1, 1996). Our lab confirmed this result by showing the simultaneous double labeling of fragmented DNA and alpha-smooth muscle actin (a-SMA), the marker for myofibroblast, in biopsies from patients with IPF (Uhal et al., 1998). Fragmented DNA in the alveolar epithelium was found frequently and immediately adjacent to a-SMA- positive interstitial cells. Thus, epithelial apoptosis colocalizes with myofibroblast where collagen deposition is severe, at least in patients with IPF. Recent work from this laboratory has shown that exposure of cultured AECs to Fas ligand (Wang et al., 1999), tumor necrosis factor-a (Wang et al., 2000), or BLEO (Li et al., 2003) all induce expression of angiotensinogen mRNA and protein, and its cleavage to the peptide angiotensin II (ANGII). Moreover, apoptosis of cultured AECS in response to these apoptosis inducers was abrogated by antisense oligonucleotides against ANGEN mRNA (Li et al., 2003; Uhal et al., 2003; Filippatos and Uhal, 2003). For all these reasons, it was hypothesized that blockade of local pulmonary ANG II synthesis by administration of antisense oligonucleotides against ANGEN mRNA might attenuate bleomycin-induced apoptosis of AECs and pulmonary fibrosis. To test this theory, normal rats and explants were subjected to intratracheal BLEO administration and 120 quantitation of apoptosis and lung collagens. We report here the prevention of both BLEO-induced AEC apoptosis and lung collagen accumulation in whole animals and explants by administration of the antisense oligonucleotides against ANGEN mRNA. 121 MATERIALS AND METHODS Reagents and Materials Phosphorothioated control and antisense oligonucleotides against angiotensinogen (18- mers) were synthesized and purchased from Genemed Synthesis (San Francisco, CA). Alkaline phosphataseconjugated streptavidin, digoxigenin-labeled deoxyuridine trisphosphate (dig-dUTP), and biotinylated deoxyuridine trisphosphate (bio-dUTP) were obtained from Boehringer Mannheim, Indianapolis, IN. BLEO was obtained from Sigma Chemical Co., Saint Louis, MO. Reagents for detection of alkaline phosphatase and other secondary reagents for in situ end labeling (ISEL) of DNA or Western blotting were from sources described earlier (Wang et al., 2000). All other materials were of reagent grade and were obtained from Sigma Chemical Co. Animals, Induction of Pulmonary Fibrosis, and Surgical Procedures: Induction of Lung Injury and Fibrosis Adult male Wistar rats, 150-200 g, were housed in a satellite facility of University Laboratory Animal Resources, Michigan State University. Animals under pentobarbital anesthesia received a single intratracheal instillation of bleomycin sulfate (BLEO) at 8 U/ kg body weight, in 400 pl of sterile saline. The 400-111 dose was instilled at end- expiration, and the liquid was followed immediately by 2ml of air to ensure delivery to the distal airways. Control animals were instilled with an equal volume of sterile saline by using previously published protocols (Wang et al., 2000). And lung tissue was perfused with PBS and harvested at 3 hours, 6 hours, 24 hours, or 2,7,14 days after instillation of bleomycin. ANGEN mRNA was detected by semiquantitative RTPCR and 122 in situ hybridization. Double immunolabeling was performed by using Anti-ANG I, lectin or Anti-alpha smooth muscle actin (a-SMA) antibody to identify the cellular source of ANGEN/ANG 1 protein. ANG II in perfused lung tissue was measured by ELISA kit specific for ANG II. Intratracheal delivery of antisense in vivo Fluorescent (BODIPY)-labeled 18-mer antisense phosphorothioated oligonucleotides against ANGEN were synthesized and used in a series of instillations of bare oligonucleotide at 4 different doses: 150, 75, 25 and 10ug in 400ul PBS, each of which were instilled one time I.T. into Wistar rats. 2 hours later, the lungs, livers and kidneys were excised. One half tissue was immediately frozen in liquid nitrogen and the other half was processed by a standard protocol for the preparation of frozen sections. Frozen tissues in dry ice were sent to the histology lab at Michigan State University for frozen cryostat sectioning. In order to demonstrate individual cells, all sections were incubated with P1 to stain the cell nuclei. Sections then were examined under fluorescence microscope. In some studies antisense oligonucleotides against ANGEN mRNA and scramble oligonucleotides was added to the intratracheal instillate at the dose of 75ug/rat. 14 days after instillation of BLEO with or without oligonucleotides, all animals were sacrificed for histology, detection of collagen or DNA fragmentation, and caspase 3 activation in epithelial cells. Surgical Procedures Immediately before sacrifice, animals were given intraperitoneal injections of sodium pentobarbital and the trachea was cannulated. Afier perfusion by PBS, the left lung was 123 ligated at the hilus, excised distal to the ligation, and immediately frozen in liquid N2 for hydroxyproline assay of total collagen (see below). The remaining lung tissues were carefully removed and were instilled with 4% parafonnaldehyde in phosphate-buffered saline (PBS) at 20 cm of H20 constant pressure, then immersed in the same fixative for 30 minutes followed by storage in 70% ethanol. The fixed tissues were washed with PBS three times for 15 minutes and were then embedded in paraffin. Five um sections of lung were deparaffinized by passing through xylene, xylene: alcohol 1:1, 100% alcohol, and 70% alcohol for 10 minutes each. Ethanol was removing by rinsing with distilled water. Lung Explant Culture Normal rat lungs were removed and perfused with PBS to flush the blood out. Tracheas were cannulated and DMEM contains bleomycin (25mU/ml) alone or with oligonuclitides (40nm) were instilled. Medium filled lungs were then cut into 1 mm square pieces. As suggested in other lung tissue culture study (Taylor et al., 2000), the minced lung chunks were plated into transwell polycarbonate inserts (0.4 um pore) and covered by a thin layer (1 mm) of culture medium with 10% fetal bovine serum (FBS) or 1% insulin-transferrin-selenitun (ITS) to facilitate gas exchange. We used two different culture medium to test whether culture medium with ITS has the same effect of that with F BS which could contain angiotensin system components like ANGEN. The explants were exposed to bleomycin (25mU/ml), BLEO plus saralasin (SAR: 50ug/ml) or BLEO plus ANGEN antisense oligonucleotides (AS: 40nM) and Vit C (0.1mM). The culture medium was changed every other day and at the same time added fresh reagents. The explants were cultured in a 5% C02 incubator at 37 °C for 24 hours 124 or 14 days, and then harvested by transfer into liquid N2 and storage at -80°C until for hydroxyproline assay to quantitate collagen amount (Woessner, 1961) and caspase-3 enzyme assay. Identification and Quantitation of Apoptotic Cells and Total Lung Caspase 3 Activity Localization of DNA Fragmentation ISEL of fragmented DNA was conducted by a modification of the method of Mundle and colleagues (Mundle et al., 1994). Briefly, ethanol was removed from deparaffinized lung sections by rinsing in distilled water for at least 10 minutes. The slides were then placed in 3% hydrogen peroxide (Sigma Chemical Co.) for 30 minutes at 20°C, rinsed with PBS, and incubated with Proteinase K (Sigma) in standard saline citrate for 15 minutes at 37°C. Samples were rinsed once in water, three times in 0.15mol/L PBS for 4 minutes each, and were then incubated in standard saline citrate (0.3 mol/L NaCl and 30 mmol/L sodium citrate in water, pH 7.0) at 80°C for 20 minutes. After four rinses in PBS and four rinses in buffer A (50 mmol/L Tris/HCl, 5 mmol/L MgCl, 10 mmol/L B- mercaptoethanol, and 0.005% bovine serum albumin in water, pH 7.5), the sections were incubated at 18°C for 2 hours with ISEL solution (0.001mmol/L digoxigenin-dUTP; 20 U/ml DNA Polymerase I; and 0.01 mmol/L each of dATP, dCTP, and dGTP in buffer A). Afterward the sections were rinsed thoroughly five times with buffer A and three additional times in PBS. Detection of incorporated dUTP was achieved with by incubation for 2 hours at 37°C with AP-conjugated antidigoxigenin (Boehringer Mannheim) at 1/400 dilution. Bound AP-antibody was then detected with the Fast Blue 125 chromogen system and the sections were mounted with Fluoromount solution (Southern Biotechnology, Birmingham, AL). For quantitation of ISEL- positive epithelial cells, the number of positive cells within the surfaces of the alveolar walls was counted in a minimum of six randomly selected x 400 microscopic fields per lung section. Positive cells within the alveolar airspaces, or otherwise clearly not within the surface of the alveolar wall, were not scored. The counts of positive nuclei per field were expressed as a percentage of the total number of nuclei in the same microscopic field. Sections from each of at least five rats per treatment group were analyzed by an investigator blinded to sample identity. Caspase 3 Enzyme Activity Assay of total lung caspase-3 enzyme activity was conducted with a commercially available kit (Molecular Probes, Eugene, OR). Fast-frozen lung was homogenized in assay kit buffer and was analyzed according to the manufacturer’s instructions on a Biotek FL600 fluorescence plate reader. In all samples, specificity of the reaction for caspase 3 was verified by abrogation of the signal with a caspase-3 selective irreversible inhibitor (data not shown). Quantitation of Lung Collagen For quantitation of total lung collagen, tissues frozen in liquid N2 were dried to constant weight in preweighed tubes at 80°C. The weighed dry tissue was hydrolyzed in 6 N HCl and was subjected to determination of hydroxyproline as described earlier by Woessner 126 (Woessner, 1961) The efficiency of the hydrolysis was verified with rat-tail collagen by comparison to standard hydroxyproline (Sigma Chemical Co.). Detection of AN GEN mRNA in lung section by In Situ Hybridization (ISH) Based on published methods (Panoskaltisis-mortari et al., 1995), modification was done so that digoxigenin-labled DNA probes will be substituted for riboprobes. For this reason, denaturation, hybridization and wash conditions was modified to favor RNA/DNA hybridization rather than RNA/RNA. Then lung sections were incubated with biotin- conjugated anti-digoxigenin antibodies, followed by using the streptavidin- AP and NBT/BCIP detection system to amplify the chromogen signal. Immunohistochemistry (IHC) for Activated Caspase 3, ANGEN/ANGI, lectin, alpha- smooth muscle actin (aSMA). IHC was performed with an antibody that recognizes only the active form of caspase 3 (BioVision, MountainView, CA) and anti- ANGEN/ANGI antibody (Santa Cruz, CA). Deparaffinized lung sections were blocked with a solution of 3% bovine serum albumin in PBS for 1 hour; the primary antibody was then applied overnight at 4°C in 3% bovine serum albumin/PBS. After washing in PBS, the antibody was detected with a biotin- conjugated secondary antibody and avidin-linked chromogen system. For double labeling, lectin (Vector Laboratories, CA) and alpha- smooth muscle actin (Sigma, Missouri) antibody directly conjugated with FITC were applied on some sections. 127 Western blot analysis to detect and quantitate pulmonary angiotensinogen and active caspase 3 The lungs were perfused and snap-frozen in liquid nitrogen and stored at -80°C until protein extraction. Lung protein extraction was performed as following. Briefly, the frozen lungs were pulverized in a chilled mortar and placed in NP40 tissue lysis buffer contain protease inhibitors cocktail. The samples were firrther centrifuged at 15,800 g for 10 min at 4°C. The supernatants were collected, and protein levels were determined using a Bio-Rad protein assay (Bio-Rad Laboratories, Hercules, CA). The protein samples (60 ug) were electrophoresed on 12% SDS-PAGE in a Mini-Protean II Electrophoresis Cell (Bio-Rad). Protein molecular weight markers (Invitrogen) were run parallel to each blot as an indicator of the molecular weight. The separated proteins were transferred at 150 V for 1.5 hours onto PVDF membrane (Bio-Rad) in a Mini Trans-Blot chamber with transfer buffer (25 mM Tris-HCI, 192 mM glycine, and 20% methanol). The PVDF membrane was blocked for l h using 5% no fat dry milk in Tris-buffered saline (TBS). For detection of angiotensinogen, a 1:100 dilution of ANG I/II antibody (Santa cruz, CA) as used and for detection of active caspase-3, 1 ug/ml antibody (Bio Vision, CA) as used. After being washed, the membrane was incubated with horseradish peroxidase linked with the secondary antibody (anti-goat immunoglobulin G for ANGEN; anti-rabbit immunoglobulin G for active caspase-3), as recommended by the manufacturer. Finally, the washed blots were exposed to an enhanced chemiluminescence (ECL) detection system (Amersham) and recorded on an autoradiograph (Amersham film). Attention: Images in this dissertation are presented in color. 128 RESULTS: Local pulmonary angiotensin synthesis was upregulated in vivo after bleomycin instillation. Semiquantitative RTPCR (Fig. 6.1) showed upregulation of ANGEN mRNA in the lungs as early as 3h after i.t. instillation of bleomycin. In situ hybridization of ANGEN mRNA (Fig. 6.2) demonstrated that positive labeling (dark purple, shown by arrowhead) increased 6h (Fig. 6.2: Bx200 magnification, Cx400 magnification) and 14 days (F ig.6.2: E, Fx200 magnification) after BLEO instillation compared to the corresponding controls (Fig. 6.2: A, Dx200 magnification). 6h after BLEO instillation, the positive labeling is mainly localized at the corners of the alveolar, which typically are the positions for type II alveolar cells (Fig. 6.ZBC). 14 days after BLEO instillation, the positive labeling is mainly localized at fibrotic foci (Fig. 6.2E) where myofibroblast / fibroblast accumulate and the comers of the alveolar (Fig. 6.2F) near the fibrotic region. Immunohistochemistry (IHC) staining (Fig. 6.3) by using anti-ANG I antibody, which cross-reacted with ANGEN [see Fig. 6.10 western blot: rat liver ANGEN (first lane) was recognized by anti-ANG I Ab], showed that at 24h after i.t. BLEO ANGEN /ANG I (purple) was found in alveolar walls cell at the comers that did not label with lectin (compare Fig 6.3 A &B x400, same filed with matched arrowhead), consistent with the identity of those alveolar wall cells as type 11 cells because lectin labels bronchial epithelium and type I alveolar epithelial cells but not type 11 cells (Fehrenbach et al., 2000). In severely affected areas of the parenchyma of bleomycin- induced fibrotic rat lungs, regions of ANGEN / ANG I labeling (Fig. 6.3 Cx200, black box) were observed to coincide with a loss of lectin labeling (Fig. 6.3 Dx200, white box: double label of same 129 section as C), consistent with our theory that alveolar epithelium dies in regions rich in ANG peptides. Enlargement of the white boxed region (Fig. 6.3 Ex400) reveals a-SMA immunoreactivity in the middle of the box on the adjacent serial section, suggestive of myofibroblasts. Western blot for ANGEN showed that ANGEN protein in the lungs which were perfused by PBS, 14 days after bleomycin instillation were significantly higher than that in control lung instilled with saline (Fig. 6.10). (Bars are the meansi SEM of n=5, p<0.05 vs. CTL by t test). A specific ELISA for ANG II detected that the pulmonary concentrations of ANGII in the lungs which were perfused by PBS, 6h and 14 days after bleomycin instillation were significantly higher than that in control lung instilled with saline (Fig. 6.4). (Bars are the meansi SEM of n=5, p<0.05 vs. CTL by t test). Antisense oligonucleotides against ANGEN mRNA blocked bleomycin induced apoptosis and collagen accumulation in lung explants (ex vivo). BLEO increased caspase 3 activity (P < 0.05), when applied in vitro at 25 mU/ml for 24 hours to rat lung explants that were depleted of blood before explant culture. Fig 6.5 showed that application of BLEO in vitro increased caspase-3 activity in the explants in 24 hours but Antisense oligonucleotides against ANGEN mRNA (40nM) inhibited the increase. Fig. 6.6A showed that there was more collagen accumulation in explants cultured in 10% F BS, treated with bleomycin compared with control group treated without bleomycin [Results were shown as pg hydroxyproline (HP) per mg of dry lung tissue. Bars are the mean 1 SEM of at least 4 separated samples; * = p<0.05 versus 130 control]. Moreover, Fig 6.6B showed that ANGEN antisense oligonucleotides reduced BLEO-induced collagen accumulation in explants cultured in 1% ITS (Bars are the mean 35 SEM; * = p<0.01 versus BLEO+AS). Blockade of lung-derived ANGEN suppressed apoptosis of AECs and pulmonary fibrosis in vivo. The feasibility of intratracheal delivery of antisense oligos specifically to the lung: Figure 6.7 shows the distribution of BODIPY fluorescence in homogenates of the frozen lung, liver and kidneys after normalization to total tissue protein. At the dose of 150ug, lung tissue retained 30-fold more fluorescence, per unit protein, than liver and 37-fold more than kidney. At the dose of 75ug, the lungs retained 6-fold more than liver and 13- fold more than kidney. Sections from the rat treated with 75ug of oligonucleotide revealed that BODIPY fluorescence (green) was localized primarily to the epithelium of airways (Fig. 6.8B, arrow) and in isolated cells of the alveolar walls (Fig. 6.8B, arrowhead). At higher magnification (Fig. 6.8C, D), some alveolar wall cells were found to concentrate the BODIPY-oligonucleotide (arrows) while others were stained with very little of the BODIPY-oligonucleotide (arrowheads). Inhibition of Apoptosis and Collagen Deposition by Antisense in vivo: The ability of Antisense to inhibit lung epithelial apoptosis was also observed in vivo at 14 days after BLEO administration. In Figure 6.12, intratracheal BLEO increased the abundance of ISEL-positive cells, but AS inhibited the increase. Measurement of lung collagen accumulation in the same animals by hydroxyproline assay (Figure 6.9B) 131 revealed an increase in total lung collagen by 14 days after intratracheal BLEO, but AS reduced the increase to a value not significantly different from the control (CTL). Fig. 6.9 A showed macroscopic photographs of the lungs 14 days after different treatment. Note the almost normal appearance of the lung in the Bleo +AS treated rat compared with the Bleo- and bleo+SCR treated rat lungs, which are smaller with many patchy bleeding spots. Histology of the whole lung sections obtained 14 days alter intra tracheal bleomycin administration revealed typical changes of PF in bleomycin treated rats without antisense administration. The normal alveolar architecture appeared to be distorted with collapsed alveolar spaces and thickening of nearby alveolar septa. In . antisense treated rats challenged with bleomycin, fibrosis—like changes were less extensively present. In rats treated with sterile saline, the histology was normal. Active caspase-3 was detected by immunohistochemistry (IHC) and western blots (WB) 14 days after bleomycin instillation. Active caspase-3 shown by WB by using anti-active caspsase-3 (p17 fragment) antibody was induced by bleomycin and suppressed by antisense treatment (Figure 6.11, upper panel). Active caspase—3 was localized in alveolar epithelial cells and alveolar macrophages (Figure 6.11, lower panel). The Antisense oligonucleotides against ANGEN mRNA inhibited DNA fragmentation detected by in situ end labeling (ISEL) of fragmented DNA in lung epithelial cells 14 days after BLEO instillation (Figure 6.12, Figure 6.13). ISEL positive cells are blue. In CTL group, ISEL-positive nuclei were not observed (Figure6.l2A). In bleomycin alone treated group, ISEL-positive nuclei were observed in cells within the alveolar walls, many ISEL-positive nuclei were observed in septal wall cells at the alveolar corners 132 (Figure6.lZB). Administration of antisense decreased ISEL positive cells in response to bleomycin, but scramble did not (Figure6. 12 C, D). Effect of antisense treatment on angiotensin peptides: Intratracheal instillation of 75ug antisense decreased pulmonary ANGEN expression (fig 6.10) but had no effect on the serum ANG II level. The plasma ANGEN protein did not decreased in response to intratracheal instillation of antisense. 133 CTL BLEO Fig. 6.1: Semiquantitative RT-PCR of angiotensinogen (ANGEN) mRNA in lungs 3hours after Bleo exposure. Normal rats were intraltracheeally instilled with Bleo (8U/Kg) or saline (CTL) for 3hours and total RNA was isolated. RT-PCR was performed as described before (see Material and Methods) with primers specific for rat ANGEN, 13 -Microglobulin ( B -MG) as control mRNAs. 134 Fig. 6.2: In situ hybridization of ANGEN mRNA demonstrated that positive labeling (dark purple, shown by arrowhead) increased 6h (B x 200 magnification, C x 400 magnification) and 14 days (E, Fx200 magnification) after BLEO instillation compared to the corresponding controls (A, D x 200 magnification). 6h aficr BLEO instillation, the positive labeling is mainly localized at the corners of the alveoli, which typically are the positions for type II alveolar cells (BC). 14 days after BLEO instillation, the positive labeling is mainly localized at fibrotic foci (E) where myofibroblast / fibroblast accumulate and the comers of the alveolar (F) near the fibrotic region. ANTI-ANGI: crr. ANT"A”G'=FL30 ' 1_I"('l|\ \\ll;rS\1\ Fig. 6.3: The sections were subjected to IHC with antibodies against the ANGI, lectin and a-SMA 24 hours (B, C) and 7 days (D, E, F) after intratracheal instillation of bleomycin. 24h alter BLEO instillation ANGEN /ANG I (purple) was found in alveolar walls cell at the corners that did not label with lectin (compare B&C x400, same field with matched arrowhead). In seveme affected areas of the parenchyma of bleomycin- induced fibrotic rat lungs, regions of ANGEN / ANG I labeling (Dx200, black box) were observed to coincide with a loss of lectin labeling (Ex200, white box: double label of same section as D). Enlargement of the white-boxed region (F x400) revealed a-SMA immunoreactivity in the middle of the box on the adjacent serial section, suggestive of myofibroblasts. 136 “' rill yup; 20 a =p<0.05 vs. CTL .3. 0| ..‘L O 01 [ANG II] IN PERFUSED LUNG (ng/ mg DNA) C TL BLEO 6h BLEO 14D Fig. 6.4 Pulmonary angiotensin II (ANG II) increased in bleo-induced lung injury. 6 hours and 14 days after bleo instillation, lungs were perfused, homogenized, and analyzed by ELISA specific for ANG 11. Values are means of at least 5 separate determinations. * Significantly different from CTL, P < 0.05 (by Student's t-test). 137 Caspase 3 Activety (uMol/g protein) CTL BLEO BLEO+ BLEO+ +lipo +lipo antisense scramble Fig. 6.5 The Antisense oligonucleotides against ANGEN mRNA inhibit BLEO-induced activation of caspase 3. Lung explants were prepared from normal rat lung tissue perfused before excision (see Materials and Methods). BLEO (25 mU/ml) was applied in serrun-free culture medium for 24 hours in the presence or absence of antisense (40nM). Bars are the means :1: SEM of n = 3; *, P < 0.05 versus control (CTL+lipo) by analysis of variance and Student-Newman-Keul’s test. 138 w e AzFBS u—t—NN UIOUIOUI pg HP/ mg dry weight *=p<0.05 CT'L BLEO BLEO+ SAR 30 25 20 15 10 B: ITS a: =p<0.01 CT L BLEO BLEO+AS Fig. 6.6 The Antisense oligonucleotides against ANGEN mRNA and non-selective AT receptor antagonist saralasin inhibit BLEO-induced collagen accumulation in lung explants. Lung explants were prepared from normal mouse lung tissue perfused before excision (see Materials and Methods). BLEO (25 mU/ml) was applied in 10% FBS (A) or 1% ITS (B) for 14 days in the presence or absence of SAR (50ug/ml) or antisense (40nM). Bars are the means :1: SEM of n = 3; *, P < 0.05 versus control (CTL) in A; *, P < 0.01 versus CTL in B by analysis of variance and Student-Newman-Keul’s test. 139 [:1 (D 5515- 150ugl.T. -* 75ugI.T. ‘ m8 HO an- -. . 1321’ >\ B3 5. -. . gs LUNG LIVER KIDNEY LUNG LIVER KIDNEY Fig. 6.7 Distribution of fluorescence in lung, liver and kidney 2 hous after instillation of BODIPY labeled oligonucleotides. Normal rats were instilled with different dose of BODIPY labeled oligonucleotides. Fluorescence intensity was measured in the homogenates of the frozen lung, liver and kidneys after normalization to total tissue protein. 140 Fig. 6.8 Localization of the intratracheal instilled fluoseence-labelled antisense against ANGEN mRNA on the lung sections fi'om rats treated with 75ug of oligonucleotides. Panels with red fluorescence show PI staining, and panels with green fluorescence show BODIPY staining. Panel A, B (10 x10) show the same field from the same section. And so do panel C, D (40 x 10). Panel A, B revealed that BODIPY fluorescence was localized primarily to the epithelium of airways (arrow) and in isolated cells of the alveolar walls (arrowhead). Panel C, D revealed that some alveolar wall cells were stained with high intensity of BODIPY-oligonucleotides (arrows) while others were stained with very little of the oligonucleotides (arrowheads). 141 12 ”6‘0 %10- * 5 w 8“ a: .g 6_ E 2- E o- CTL BLEO BLEO+AS BLEO+SCR Fig. 6.9 A: Macroscopic photographs of the lungs 14 days after different treatment. Note the almost normal appearance of the lung in the Bleo +AS treated rat compared with the Bleo- and bleo+SCR treated rat lungs, which are smaller with many patchy bleeding spots. B: Quantitation of total lung collagen by hydroxyproline (HP) assay. At 14 days post-Bleo, collagen was quantitated by HP assay applied to hydrolyzed lung tissue. See METHODS for details. * P < 0.05 vs. CTL by ANOVA. 142 . Lena-we - n -. nu --. rn-I .- s-.- -a--. Z 1. . ‘ ll ' I i . - . , . . . . 7 I l 1.. . . a ’ I . . ’ h n- . - .. .---h. u..- .n v. ' x .a'. J 74 .. . . r- ' . 1'. .1 . '0 . n.— m . 0‘ ‘H i ‘ 1‘) , «u a ' ‘ V“. .-. pa. --. ”a g- l- I.“ I ’1 . n-a .‘f t . . . r | r . .' . , I: . . . f' ‘. " , . A: Liver CTL BLEO BLEO+AS u 0'! 3.0 - 2.5 . 2.0 - .x 01 1.0 ‘ O 01 0.0 . Relative Density Intensi CTL BLEO BLEO+AS Fig. 6.10 Bleomycin increased angiotensinogen protein expression, which was suppressed by the treatment of antisense in perfused lung tissues. 14 days after instillation of bleomycin or vechicle, lungs were perfused and homogenized for the western blot analysis. (A) Representative imrnunoblot is shown and (B) densitometric evaluation of blot data. Data are presented as mean iSE (n=5). *: P<0.05 versus CTL 143 WB: Active Caspase 3 (P17) . I M flan-1‘. 4.91“" CTL BLEO BLEO+AS BLEO+SCR Fig 6.11. Detection of active caspase-3 by immunohistochemistry (IHC) and western blots (WB) 14 days after described treatment. Male Wistar rats received indicated treatment intratracheally. Lung tissues were harvested 14 days after treatment and subjected to IHC and WB by using anti-active caspsase-3 (p17 fragment) antibody. Upper penal (WB): Active caspase-3 was induced by bleomycin and suppressed by antisense treatment. Lower penal (IHC): active caspase—3 was localized in alveolar epithelial cells and alveolar macrophages. sqwznw aka: . 1 garages” at; Fig 6.12. Detection of apoptotic cells by in situ end labeling (ISEL) of fragmented DNA 14 days after described treatment. Male Wistar rats received indicated treatment intratracheally; lung tissues were harvested 14 days after treatment and subjected to ISEL coupled to a fast blue detection system (see METHODS). Positive reaction is blue. A: in CTL group, ISEL-positive nuclei were not observed. B: in bleo group, ISEL-positive nuclei were observed in cells within the alveolar walls, many ISEL-positive nuclei were observed in septal wall cells at the alveolar corners. C: administration of antisense decreased ISEL positive cells in response to Bleo. D: administration of scramble nucleotides did not decreased ISEL positive cells in response to Bleo. 145 .3 O * p<0.01 Vs.CTL 8 **p<0.01 Vs. BLEO or B+SCR o . CTL BLEO BLEO+AS BLEO+SCR ISEL positive cells in total nuclei, % Fig. 6.13 The Antisense oligonucleotides against ANGEN mRNA inhibited DNA fragmentation in lung epithelial cells 14 days after BLEO instillation. Rats were given a single intratracheal instillation of BLEO in the presence antisense or scramble ONT in the instillate. Fourteen days later, lung sections were prepared and labeled by ISEL. Labeling was quantitated in cells within the alveolar surfaces. Bars are the means i SEM ofn = 5; *, P < 0.01 versus control (CTL); **, P < 0.01 versus BLEO or BLEO +SCR by analysis of variance and Student-Newman—Keul’s test. 146 DISCUSSION: Recently a few studies suggested the existence of “local” angiotensin systems in various organs and tissues. For example, the ANG II concentrations in the interstitial compartment of heart and eye were found to be 5-100 fold higher (about 50 —500pM) than that in plasma (~5-10pM) (van Kats et al., 1998; Danser et al., 1994). The higher interstitial levels of ANG 11 compared to the circulating level could not be explained by diffusion and/or receptor-mediated uptake of circulating angiotensin 11. These results thereby suggest that tissue angiotensin II is largely, if not completely, synthesized locally. Furthermore, cultured cells from various organs including heart (Lindpaintner et al., 1988), vascular endothelium (Li et al., 1999), brain (Campbell et al., 1986; Dzau et al., 1982; Ohkubo et al., 1986) and lung (Filippatos et al., 2001) were shown to express the RAS components. In contrast to the classical endocrine system of RAS in which angiotensin II is delivered to tissues via circulating blood, local angiotensin systems can be from either “intrinsic “(independent of the endocrine RAS) or “extrinsic” (relying on the endocrine RAS as its components sources) sources. The evidence that local angiotensin system is involved in pulmonary fibrosis is as follows: 1) Enzymes needed for the production of local angiotensin peptide were upregulated in pulmonary fibrosis. Polymorphisms that confer higher levels of ACE predispose patients to lung fibrosis (Morrison et al., 2001). Cathepsin D was upregulated in both animal and human fibrotic lung (Kasper et al., 1996; Koslowski et al., 2003). ACE is upregulated in both human and animal fibrotic lungs (V enkatesan et al., 1997; Specks et al., 1990). 2) Angiotensin system antagonists block experimental lung fibrosis (Ward et al., 1990; Uhal 147 et al., 2000 and 2002; Molteni et al., 1985 and’2000; Fang et al., 2002; Li et al., 2003; Marshall et al., 2004; Otsuka et al., 2004). For the pulmonary angiotensin system, where the ANGEN, the only precursor of angiotensin 11, comes from is unknown. Two cells types in the lung under certain conditions, so far, have been found to be able to produce ANGEN in vitro. In vitro primary AECs could synthesize and secrete ANGEN that is converted to ANG II when undergoing apoptosis induced by Fas ligand (Wang et al., 1999), TNF-alpha (Wang et al., 2000), amiodarone (Bargout et al., 2000) and bleomycin (Li et al., 2003). Studies done in our lab showed that primary myofibroblasts isolated from fibrotic human lungs (IPF biopsies) expressed the ANG II precursor ANGEN mRNA and protein (Wang et al., 1999), suggesting that human lung myofibroblast can synthesize ANGEN in culture. Furthermore, the concentration of ANG II detected by Enzyme-Linked Immunosorbent Assay (ELISA) was found to be significantly higher in culture medium of myofibroblast isolated from fibrotic human lungs than that from normal lungs. Preincubation of the medium with purified renin and ACE increased the ELISA-detectable ANG 11 concentration roughly eight fold, indicating that there are abundant ANGEN synthesized constitutively which waits to be converted to ANG 11. However, no in vivo studies have been published to directly prove the existence of a local angiotensin system produced in the lung independently of the endocrine angiotensin system during the development of pulmonary fibrosis. Therefore, the present study was performed to answer a part of unknown question. We showed that endogenous lung ANGEN was upregulated in vivo as early as 3 hours after bleomycin instillation in rat lungs by RT-PCR, in situ hybridization and immunohistochemistry staining methods. ANGEN mRNA and 148 angiotensin peptides were localized in alveolar wall cells in the alveolar comers, tentatively identified as type 11 cells, and also colocalized with alpha-Smooth Muscle Actin (aSMA)(Fig 6.3). Those data suggested that bleomycin activated the production of the local angiotensin systems in vivo. Moreover, those data are consistent with our earlier finding of epithelial cell death in the vicinity of alpha- SMA-positive myofibroblast in fibrotic human lung and support the theory that alveolar epithelial death is caused by the ANG peptide. We hypothesized that lung derived angiotensinogen is required for development of PF. Using whole animal models of bleomycin— induced PF, we cannot discriminate the lung- derived angiotensin system from the endocrine angiotensin system. However, using explants as the ex vivo model we can rule out the contributions of endocrine angiotensin system and hemodynamics to the development of bleomycin-induced PF as blood circulation is removed in explants. Furthermore, as lung explants provide an in vitro condition at the organ level, it will provide a better mimic of the in vivo conditions compared to cell cultures. This study showed that bleomycin could induce apoptosis and fibrosis in lung explants, which could be blocked by antisense oligonucleotides against ANGEN mRNA (Fig. 6.6), suggesting lung-derived ANGEN is required for bleomycin- induced pulmonary fibrosis in serum free condition. The previous studies in our lab demonstrate that apoptosis of AECs in response to BLEO can be abrogated by antisense oligonucleotides against angiotensinogen (ANGEN) mRNA. We use the same oligonucleotide sequences, delivered by intratracheal (I.T.) instillation, for in vivo experiments to attempt blockade of BLEO-induced apoptosis and lung fibrosis in the intact animal. The success of such an experiment will depend on 149 specific delivery to the lung, so we sought to determine the distribution of the oligonucleotides in the lungs and other tissues after I.T. instillation. Our data showed that a single I.T. dose of 150ug antisense oligonucleotide can provide delivery primarily to lung with relatively little accumulation in liver or kidney. Moreover, the intratracheal instillation of antisense at dose of 75ug provided delivery of oligonucleotides primarily to lining cells of the airways and alveolar walls, suggesting that this is a feasible approach for in vivo studies. The initial concern was that a single dose of the antisense oligonucleotides against ANGEN mRNA is not sufficient to exert its effects on pulmonary fibrosis considering the decay of its efficacy over time in vivo although a single dose of the same antisense oligonucleotides against angiotensinogen mRNA reduces hypertension for a long time in various hypertension animal model (Gyurko et al., 1993; Wielbo et al., 1995; Wielbo et al., 1996; Peng et al., 1998; Kagiyama et al., 2001; Phillips, 2001). The half-life of ANGEN antisense oligonuclotide is not known although there are a few reports regarding the half-lives of similar oligonucleotides available. Some studies showed that the half-life of a 15-mer phosphorothioate oligonucleotide was 9 hours in human serum, 4 days in tissue culture media (Li et al., 1996) and 19 hours in cerebrospinal fluid (Campbell et al., 1990). Other studies reported that the half-life of phosphorothioate oligonucleotide following inhalation delivery to lung was more than 20 hours in mice (Templin et al., 2000) and 30 hours in rabbit (Ali et al., 2001). Regardless of this concern, intratracheal instillation of 75ug antisense once reduced bleomycin—induced pulmonary fibrosis detected by hydroxyproline assay, decreased pulmonary ANGEN and active caspase-3 level detected by western blot and ISEL positive cells, but had no effect on the serum ANG 11 level. These data are 150 consistent with the hypothesis that lung-derived ANGEN is involved in bleomycin- induced pulmonary fibrosis. Summary and Conclusion The present study demonstrated that the upregulation of de novo ANGEN synthesis found in vitro occurs in vivo. Administration of antisense oligos against ANGEN mRN A, presumably by blockade the synthesis of lung-derived ANGEN, blocked BLEO-induced apoptosis and lung fibrosis. Those data also suggest that lung-derived ANGEN is the additional therapeutic target within the pulmonary angiotensisn system and antisense oligonucleotides against ANGEN mRNA may hold potential for the treatment of lung fibrosis in humans. 151 Chapter 7 GENERAL DISSCUSSION l. Angiotensin Receptor AT1 Mediates Bleomycin-induced Apoptosis and Lung Fibrosis Our study demonstrated at various levels including cellular, organ and whole animal level that AT1 receptor mediates bleomycin-induced apoptosis and lung fibrosis. 1.1 An Essential Role by Angiotensin Receptor AT1 in Bleomycin-Induced Apoptosis in Alveolar Epithelial Cells (AECs): We demonstrated that BLEO induced apoptosis of primary AECS in a dose dependent manner in vitro. This result is consistent with the in vivo studies from other labs showing that AECs apoptosis is found in bleomycin-induced pulmonary fibrosis model (Hagimoto et al., 1997). To confirm the nature of the cell death induced by BLEO was apoptosis, our study also showed that fragmented DNA induced by bleomycin was blocked by the broad-spectrum caspase inhibitor ZVAD-fink, the caspase 3-selective inhibitor DEVD-fmk and endonuclease inhibitor ATA. F urtherrnore, we found that the ANG receptor subtype AT1-selective blocker losartan blocked nuclear fragmentation induced by bleomycin, suggesting that angiotensin receptor subtype AT1 mediates BLEO-induced apoptosis as it does in ANGII-induced apoptosis (Papp et al., 2002). Similarly, our study using the human AECs showed that BLEO-induced apoptosis of A549 cells was inhibited by the AT1-selective blocker L158809 but not by the AT2- selective antagonist PD123319 (Li et al., 2003). Moreover, BLEO-induced apoptosis was 152 also prevented by a neutralizing ANGII antibody (anti-ANGII). Furthermore, the total enzymatic activity of caspase 3 was elevated by exposure of A549 cells to BLEO and the increase was inhibited by non-selective AT receptor antagonist saralasin (Li et al., 2003). The findings that bleomycin-induced apoptosis of AECS could be blocked by the AT1- selective antagonists but not by the AT2-selective antagonist support the contention that there is an essential role by AT1 receptor in Bleomycin-induced apoptosis in AECs. 1.2 An Essential Role of Angiotensin Receptor AT1 in Bleomycin-induced Apoptosis and Fibrosis in Lung Explants: We cannot discriminate the lung-derived angiotensin system from the endocrine angiotensin system using whole animal models of bleomycin- induced PF. However, using explants as the ex vivo model the contributions of endocrine angiotensin system and hemodynamics to the development of bleomycin-induced PF can be ruled out as blood circulation is removed in explants. Furthermore, as lung explants provide an in vitro condition at the organ level, it offers a better mimic of the in vivo conditions compared to cell cultures. Our studies showed that bleomycin could induce apoptosis and fibrosis in lung explants, which could be blocked by AT] selective antagonists losartan, suggesting that AT1 receptor is required for bleomycin-induced apoptosis and pulmonary fibrosis in serum free condition. The ability of LOS to prevent caspase-3 activation by BLEO in cultured lung explants argues against the involvement of decreased blood pressure in the inhibition of apoptosis as no hydrostatic pressure is there in explants. Moreover, the induction of LOS- inhibitable caspase 3 activities in lung explants argues against a primary role for blood- 153 derived cells in the initiation of the apoptosis as blood was removed from the explants. Hence, this result also suggests that the inhibitory effect of LOS on apoptosis was not mediated by an indirect action on infiltrating inflammatory cells. 1.3 In vivo Inhibition of Apoptosis and Collagen Deposition by an AT1 Antagonist and AT1 Gene Deletion: Our study demonstrated that losartan, the AT1 receptor antagonist, inhibited lung epithelial apoptosis and fibrosis in vivo 14 days after BLEO administration. The specific effect of inhibition of apoptosis was confirmed by using two different assays: ISEL labeling to mark the fragmented DNA and active caspase 3 labeling to show the activat- ion of caspase 3. Meanwhile, measurement of lung collagen accumulation in the same animals by hydroxyproline assay revealed an increase in total lung collagen after intratracheally delivery of BLEO, which was reduced by LOS to a level not significantly different from the control. The AT1 receptor is expressed in two isoforms, ATla and Ale, for which no selective antagonists have yet been developed (Filippatos et al., 2001). Subtype ATla is known to be expressed in the lungs of mice, but Ale was not detected in mouse lung by reverse transcriptase-polymerase chain reaction (Burson et al., 1994). Although it is possible that cells of minor abundance in the lung, such as type 11 cells, express Ale in quantities not detected in earlier studies (Burson et al., 1994), the primary isolates of type II pneumocytes from Wistar rats did not reveal Ale expression by reverse transcriptase- polymerase chain reaction despite the use of two different primer sets and high- amplification cycle numbers (from unpublished observation). Based on those studies, we 154 used ATla-null mice to test the hypothesis that AT1 receptor is essential for the development of pulmonary fibrosis. Our study demonstrated that deletion of the AT1 receptor, throughout the time course of the 14-day BLEO model, was capable of inhibiting or blocking apoptotic and fibrotic responses. Furthermore, the finding that deletion of only one allele of the ATla gene significantly reduced Bleo-induced apoptosis and collagen accumulation supports the notion that ATla is the only active AT1 receptor subtype on the alveolar epithelium of mice. One concern was raised when we were using ATla deletion mice. That is whether the ANG II dependent caspase-3 activation cascade is still intact in ATla deletion mice since we detected less active caspase-3 in response to instillation of bleomycin. The chance that caspase-3 activation cascade has been compromised is small since those mice have normal phenotype except that 12-mmHg lower blood pressure is observed. If caspase-3 activation cascade were damaged in those mice, they would have a much higher chance to develop tumors, which has not been reported in those mice. In addition, in a recently published study, Ohashi et al. (2004) demonstrated that ANG II dependent caspase-3 activity is increased in ATla deletion mice, strongly supporting that AN G 11 dependent caspase 3 activation cascades is intact in ATla deletion mice. 1.4 AT1 receptor blockade block fibrosis via suppression of apoptosis: Considering the many known functions of angiotensin 11, these results above raise the question of whether the inhibition of fibrogenesis by AT1 blockade was due to its suppression of apoptosis. For example, blockade of AT1 receptor may also inhibit mitosis of lung fibroblasts (Marshall et al., 2000) and reduce collagen synthesis by lung 155 fibroblasts (Marshall et al., 2004). Both effects are relatively delayed consequences of AT1 blockade that might be independent of ABC apoptosis occurring at earlier time. Moreover, endothelial cells also express AT1 receptor and undergo apoptosis in response to angiotensin, although at relatively high concentrations (Dimmeler et al., 1997; Li et al., 1999). There are also other cell types residing in the lung known to respond to angiotensin by mechanisms currently under intensive study (Harrison et al., 2003). Thus, it is possible that the acute early effects of AT1 blockade on ABC apoptosis are not required for inhibition of collagen deposition later. Quantification of erythrocytes reaching the alveolar airspaces 6 hours after BLEO administration (a crude index of lung barrier collapse) suggested that LOS did not prevent acute, transient barrier collapse despite its ability to reduce caspase-3 activation at the same time. This observation is in agreement with the theory that the blockade of apoptosis in AECs is a key to the subsequent blockade of collagen deposition. Moreover, previous studies in our lab with apoptosis inhibitors support the contention that the acute apoptotic response is a crucial event in the BLEO model of PF (Wang et al., 2000). Wang and colleagues showed that the ACE inhibitor captopril or the caspase inhibitor ZVAD- fmk had essentially equal ability to block the appearance of apoptotic epithelial cells in rats exposed to intratracheal BLEO and to prevent subsequent collagen deposition (Wang et al., 2000). That report, which was confirmed by Kuwano and colleagues (Kuwano et al., 2001) in studies of mice exposed to BLEO and/or ZVADfmk, suggested that the blockade of fibrogenesis by captopril was indeed related to inhibition of apoptosis, rather than the many other effects of ACE inhibition in vivo (Marshall, 2003). Later work in our 156 lab confirmed that the ZVAD compound had no inhibitory effect on angiotensin converting enzyme itself (Filippatos and Uhal, 2003). On the other hand, earlier work has shown that AECs undergoing apoptosis in response to F as ligand, tumor necrosis factor-a or BLEO begin secreting angiotensin into the extracellular space within hours of exposure (Wang et al., 1999; Wang et al., 2000; Li et al., 2003). Those studies also showed that the autocrine production of angiotensin and its binding to AT1 receptor on AECs were required for apoptosis in response to those agents. This mechanism can explain the ability of LOS to block AEC apoptosis in vivo in the present study. My data also are in agreement with recent reports that LOS inhibits BLEO-induced collagen deposition in rat lung (Fang et al., 2002) and that ATla-null mice show reduced liver fibrosis in response to carbon tetrachloride (Kanno et al., 2003). Thus, the present data are consistent with the ability of ACE inhibition by captopril to block both AEC apoptosis and collagen deposition in rats (Wang et al., 2000), and extend this concept to angiotensin receptor blockade in mice. 1.5 Potential intracellular mechanisms underlying AT1 mediated apoptosis: In an earlier report describing the mechanisms by which angiotensin induces apoptosis in primary cultures of AECs, Papp and colleagues (Papp et al., 2002) showed that blockage of AT1 signaling through protein kinase C (PKC) with the specific PKC inhibitor chelerythrin could attenuate the apoptotic response to angiotensin. This finding is consistent with the known role of PKC in AT1 signaling in a variety of cell types such as human coronary artery endothelial cells (Li et al., 1999) but the pathways from PKC to the effector caspase 3, which is also required for this response (Papp et al., 2002), are 157 currently unknown. Given that ABC apoptosis in response to F as ligand, tumor necrosis factor- a, or BLEO all require the autocrine production and binding of angiotensin to AT1, (Wang et al., 1999; Wang et al., 2000; Li et al., 2003) the report of Papp and colleagues (Papp et al., 2002) suggests that PKC inhibitors would also block AEC apoptosis in response to these agents in vivo as well. This prediction was not tested in the present study, but will be a worthwhile topic for future inquiry. 2. Essential role for cathepsin D in bleomycin-induced apoptosis of alveolar epithelial cells CatD is a lysosomal protease known to be ubiquitously expressed (Uchiyama et al., 2001). It has been suggested that this and other lysosomal proteases might be involved in the production of a bioactive molecule required for apoptosis of PC12 cells in response to trophic withdrawal (Isahara et al., 1999). Particularly, a role for the aspartyl protease CatD in apoptosis has been shown previously in HeLa cells exposed to interferon-y, F as ligand or TNF-or (Deiss et al., 1996) and in PA] ovarian cancer cells (Wu et al., 1998). Furthermore, the activity of CatD is upregulated by the apoptosis inducer adriaourcin in PA], MLl leukemia cells and U1752 lung cancer cells (Wu et al., 1998). Although the aspartyl protease inhibitor pepstatin A could block apoptosis in these cell types, the exact mechanism(s) by which CatD participates in the execution of apoptosis is unclear. Cathepsin D also is known to be one of the enzymes capable of proteolytically processing the liver-derived and serum-borne protein angiotensinogen to the peptide angiotensin I, a function normally performed in the serum by the kidney-derived enzyme renin 158 (F ilippatos et al., 2001). Evidence from several nonpulmonary cell types has established CatD as the primary enzyme that converts angiotensinogen to ANGI within local “intrinsic” angiotensin systems, independently of renin (F ilippatos et al., 2001; Weber et al., 1995). 2.1 CatD activity is upregulated in AECs apoptosis response to bleomycin and in fibrotic lungs: It was shown that CatD activity is upregulated in AECs in fibrotic human lung (Kasper et al., 1996) and induced in the L132 lung cell line during apoptosis in vitro (Kasper et al., 1999). Consistent with those findings, in our study, western blotting of conditioned medium from rat AECs exposed to bleomycin reveals bleomycin-induced increases in several isoforms of CatD. Hence, we demonstrated that bleomycin upregulated CatD activity and release from AECs. To investigate the mechanisms underlying the increase of CatD activity in response to bleomycin, we performed the time course study to monitor the change of the level of CatD mRNA. In other cell types such as PAl human ovarian cancer cells, it was shown that apoptosis inducers upregulate both CatD protein and mRNA, suggesting control of activation at the level of mRNA (Wu et al., 1998). However, in our study, RT-PCR studies of ABC transcripts after bleomycin treatment failed to detect changes in CatD mRNA despite significant increases in CatD activity and’immunoreactive protein by western blotting. It is likely that the relatively few sampling times chosen for realtime analyses of CatD mRNA may have missed a transient but shortlived increase in the mRNA that might be revealed by a more exhaustive time course study. On the other 159 hand, CatD is known to undergo activation by proteolytic mechanisms as well; in human U937 cells, CatD was shown to undergo processing from the inactive prepro-isoform (52kdal) to the active proCatD (48kdal) and an active 32kdal isoform, in response to autocatalysis of the enzyme induced by the direct binding of the apoptosis mediator ceramide (Heinrich et al., 1999). However, two of the isoforms shown to be increased in ABC media (52 and 48kda1) are larger than the primary isoform detected intracellularly in ABC (44kdal). This finding argues against proteolytic processing alone as a mechanism of CatD activation in AEC. Thus, the exact mechanism(s) by which bleomycin upregulates CatD in ABC is unknown, but will pose an interesting problem for future studies. Pepstatin A-inhibitable CatD activity also was upregulated by amiodarone and TNF- alpha, both of which induce apoptosis in AECS (Uhal et al., 2003; Wang et al., 2000). However to determine whether the requirement for CatD is universal to all proapoptotic stimuli for ABC will require further investigation. 2.2 Bleomycin-induced apoptosis of AECs is reduced by blockade of CatD activity or synthesis: Our data showed that bleomycin-induced nuclear fragmentation and caspase 3 activity were significantly reduced by the aspartyl protease inhibitor pepstatin A and by antisense oligonucleotides against CatD mRNA, strongly suggesting a role played by CatD in AEC apoptosis. The other finding from our study that CatD antisense treatment did not completely block bleomycin-induced nuclear fragmentation indicates a potential role for additional 160 protease(s) in angiotensinogen processing and subsequent AEC apoptosis. This interpretation is reflected by the result the protease inhibitor pepstatin A, which blocks all aspartyl proteases, was also incapable of complete blockade of nuclear fragmentation despite complete inhibition of CatD enzyme activity in AEC lysates. On the other hand, the antisense treatment for CatD, which is at least theoretically specific, did not completely eliminate immunoreactive CatD detected by western blotting. Thus, it is difficult to determine whether the incomplete blockade of apoptosis is due to insufficient CatD knockdown or additional proteases activities. 2.3 CatD is involved in conversion of angiotensinogen to ANG I during bleomycin -induced AECs apoptosis. Our previous study demonstrated that bleomycin-induced apoptosis of AECs was competely blocked by specific angiotensin receptor antagonists or ANG-neutralizing antibodies or antisense oligonucleotides against ANGEN mRNA (Li et al., 2003). Therefore, bleomycin-induced apoptosis of AECs requires the autocrine synthesis of angiotensinogen, angiotensin II and it’s binding to ANG receptor AT1 (Li et al., 2003). This finding support the hypothesis that autocrine generation of ANGII is required for ABC apoptosis regardless of the initiating stimulus (Uhal, 2002). This result is also consistent with data from previous studies in our lab showing that purified AN GII itself was a potent inducer of apoptosis in ABC (Wang et al., 1999). Those data imply that AECs express enzymes capable of converting angiotensinogen to ANGII. Although Wang et al. (Wang et al., 1999) showed constitutive expression of angiotensin converting 161 enzyme (ACE) by AECs, the aspartyl protease required for providing the substrate for ACE (ANGI) in AEC was unknown. As noted before, apoptosis of cultured AECs in response to bleomycin was significantly inhibited by the aspartyl protease inhibitor pepstatin A and by antisense oligonucleotides against CatD mRNA. The same inhibitors also prevented the enzymatic processing of a synthetic fragment of angiotensinogen (amino acids 1-14) containing the CatD and ACE cleavage sites, and completely blocked AEC apoptosis in response to the same peptide. For example, incubation of primary rat AECs with the fi'agment F1-14 alone in serum- free culture ‘medium without CatD yielded significant production of ANGII. Our result from study using the fragment 1-14 of angiotensinogen containing the Cat D and ACE cleavage sites supports the theory that CatD is required for the conversion of angiotensinogen to ANGII by AEC. This result is also consistent with the earlier demonstration of constitutive, although low, expression of both ACE and an unidentified aspartyl protease by primary AECs (Wang et al., 1999). Moreover, the complete abrogation of AEC apoptosis in response to angiotensinogen fragment F1-14 by the nonselective and AT1-selective AN G receptor antagonists saralasin and losartan, confirmed that the induction of apoptosis was dependent on both the generation of ANGII from F1-14 and the binding of ANGII to receptor AT1. Most importantly, the findings that ABC apoptosis in response to F1-14 was essentially abrogated by either pepstatin A or by CatD antibodies, strongly suggest that the conversion of angiotensinogen to ANGI, and subsequently ANGII to induce AEC apoptosis, is dependent on CatD activity. 162 In summary, bleomycin upregulated CatD enzymatic activity and expression in primary cultures of rat AECS. Apoptosis of cultured AECs in response to bleomycin was significantly inhibited by the aspartyl protease inhibitor pepstatin A and by antisense oligonucleotides against CatD mRNA. The same inhibitors also prevented the enzymatic processing of a synthetic fragment of angiotensinogen (amino acids 1-14), and completely blocked AEC apoptosis in response to the same peptide. These data are consistent with earlier studies showing that apoptosis of AECs in response to bleomycin requires the autocrine synthesis and proteolytic processing of angiotensinogen to angiotensin II, and suggest that the proteolytic processing requires CatD. Therefore, our data suggest that blockade of CatD and other aspartyl proteases might provide a potential therapeutic strategy for pulmonary fibrosis by preventing AECS apoptosis and for lung injuries involving this mode of cell death. 3. Essential Roles for Angiotensinogen in Bleomycin-Induced Apoptosis and Lung Fibrosis In our study, the same strategies were used to demonstrate the essential roles for angiotensinogen in bleomycin-induced apoptosis and lung fibrosis as for AT1 receptor. Our studies demonstrated that lung-derived angiotensinogen is involved in bleomycin- induced apoptosis and lung fibrosis at various levels including cellular, organ and whole animal level. 3.1 Essential Roles for Angiotensinogen in Bleomycin-Induced Apoptosis in Alveolar Epithelial Cells: 163 Our data demonstrated that BLEO induces ANGEN expression in primary AECs and A549 cells. To determine if functional ANGEN mRNA is required for the apoptotic response to BLEO, phosphorothioated antisense or scrambled-sequence control oligonucleotides against ANGEN mRNA were transfected into rat AECs and A549 cells. Our studies showed that BLEO-induced apoptosis of primary AECs and A549 cells was inhibited by the ANGEN antisense but not by the scrambled control oligonucleotides. Moreover, exposure to BLEO for 20 hours reduced the total cell number of A549 cells (attached plus detached, bottom panel), which was prevented by the ANGEN antisense. Although the mechanism by which bleomycin upregulates ANGEN mRNA was not addressed in this study, the findings that BLEO, Fas L and TNF-a all increased ANGEN mRNA (Wang et al., 1999; Wang et al., 2000) imply the involvement of a pathway common to these inducers. Regulation of ANGEN expression is mostly studied in the hepatocyte, in which the stimulatory effects of TNF-a and interleukin—6 have been shown to be mediated through the interaction of transcription factors NF-kB and STAT-3 with the Acute Phase Response Element (APR) of the ANGEN promoter (Brasier et al., 1994; Sherman and Brasier, 2001). In contrast, studies of the regulation of ANGEN expression by the cardiac ourocyte have shown that p53 is a key regulator of its expression in response to a variety of stimuli (Leri et al., 1998; Pierzchalski et al., 1997). Whether this difference reflects the distinct developmental lineages of these cell types or other factors is unknown. From this point of view, future investigations of the regulation of ANGEN expression by cells of the lung shall be quite worthwhile, particularly in light of the fact that the lung contains many different cell types that are in very close proximity but of distinct embryologic origins. As an example, the results reported here that ANGEN 164 expression is required for apoptosis of AECs compliment our earlier report of ANGEN expression by human lung myofibroblasts (Wang, et al., 1999), which reside immediately adjacent to apoptotic AECs in the fibrotic lung in situ. 3.2 Essential Roles for Angiotensinogen in Bleomycin-Induced Apoptosis and Lung Fibrosis in explants: Lung-derived angiotensin system can not be discriminated from the endocrine angiotensin system if a whole animal model is used to study the role of angiotensin system in bleomycin- induced PF. However, using explants as the ex vivo model we can rule out the contributions of endocrine angiotensin system and hemodynamics to the development of bleomycin-induced PF as blood circulation is removed in explants. Furthermore, providing an in vitro condition at the organ level, lung explants will offer a better mimic of the in vivo conditions compared to cell cultures. Our studies showed that BLEO increased caspase 3 activity when applied in vitro to rat lung explants that were depleted of blood before explant culture. Application of BLEO in vitro increased caspase—3 activity in the explants, which was inhibited by antisense oligonucleotides against ANGEN mRNA. F uthemmore, we showed that there was more collagen accumulation in explants treated with bleomycin compared with control group without bleomycin. Moreover, we showed that ANGEN antisense oligonucleotides reduced BLEO-induced collagen accumulation in explants cultured in 1% ITS. Hence, this study demonstrated that bleomycin could induce apoptosis and fibrosis in lung explants, which could be blocked by antisense oligonucleotides against 165 ANGEN mRNA, suggesting lung-derived ANGEN is required for bleomycin-induced pulmonary fibrosis in serum free condition. 3.3 Essential Roles for Angiotensinogen in Bleomycin-Induced Apoptosis and Lung Fibrosis in vivo: The previous studies in our lab demonstrated that apoptosis of AECs in response to BLEO was abrogated by antisense oligonucleotides against angiotensinogen (ANGEN) mRNA (Li et al., 2003). We used the same oligonucleotide sequences delivered by intratracheal (I.T.) instillation for in vivo experiments to block BLEO-induced apoptosis and lung fibrosis in the intact animal. To ensure specific delivery of the oligonucleotide to the lung, the distribution of the oligonucleotides in the lungs and other tissues after I.T. instillation was determined. Our data showed that a single I.T. dose of 150ug antisense oligonucleotide provided delivery primarily to lung with relatively little accumulation in liver or kidney. Moreover, the intratracheal instillation of antisense at close of 75ug provided delivery of oligonucleotides primarily to lining cells of the airways and alveolar walls. Those results suggest that IT. instillation of the oligonucleotide is a feasible approach for in vivo studies. Intratracheal instillation of 75ug antisense once along with bleomycin reduced bleomycin—induced pulmonary fibrosis detected by hydroxyproline assay or picrosirius red staining, decreased ANGEN protein expression and active caspase-3 labeling and ISEL positive cells, but had no effect on the serum ANG II level. These data are consistent with the hypothesis that lung-derived ANGEN is involved in bleomycin- induced pulmonary fibrosis. Those data also suggest that lung-derived ANGEN is the 166 additional therapeutic targets within the pulmonary angiotensisn system and antisense oligonucleotides against ANGEN mRNA may hold potential for the treatment of lung fibrosis in humans. 4. Roles of Pulmonary Angiotensin System in Development of Bleomycin-induced Pulmonary Fibrosis: [Figure 7.1 modified from (Selman et al., 2001)] ® ANGEN 1‘ HM ~ ’1 ..‘“\‘ Roles of pulmonary angiotensin system in development of bleomycin-induced pulmonary fibrosis can be summarized in the above figure, described as following: 1) Induce epithelial damage and activation: Bleomycin induces apoptosis of lung AECs by upregulation of AN GEN gene expression. Synthesized ANGEN, which can be converted to ANG I by cathepsin D (Cat D). ANG I can be further converted to ANG II by ACE. ANG II can induce apoptosis of the AECs 167 through AT1 receptor, which will cause excessive loss of AECs and insufficient epithelial repair. 2) Cause fibroblast migration and proliferation: ANG II synthesized by epithelial cells can cause proliferation and transdifferentiation of fibroblasts into myofibroblasts, which are the most important source of collagen in pulmonary fibrosis through upregulation of TGF-beta gene expression. In addition, TGF beta can induce ANGEN gene expression in lung fibroblast (unpublished data from Uhal lab). There is an ANG II- TGF beta autocrine loop in the lung fibroblasts, which can amplify the profibrotic of ANG II and cause myofibroblast foci formation. 3) Cause impaired reepithelialization and subsequent pulmonary fibrosis Myofibroblasts in myofibroblast foci can synthesize ANGEN, which can be further converted to ANG II. ANG 11 derived form myofibroblast can cause the apoptosis of AECs and contribute the development of pulmonary fibrosis. 5. Rate-limiting step of pulmonary angiotensin [1 generation in bleomycin—induced pulmonary fibrosis: There are several potential rate limiting steps for pulmonary angiotensin II generation in bleomycin induced pulmonary fibrosis including: 5.1 Renin and/or Cat D: In other organ systems such as the cardiovascular system, renin has been shown to be the major determinant for local ANG II production, in which renin derived from kidney is taken up by the tissues via diffusion and binding to the (pro)renin receptors ( Danser, 2004). However, there is no evidence suggesting that similar mechanism for the 168 production of the pulmonary angiotensin system exists since the local renin synthesis has never been demonstrated in the lung (Re, 2004). In our study (unpublished data), when a non-specific aspartyl protease inhibitor Pepstatin A, also an inhibitor for renin, was systemically administered, bleomycin induced pulmonary fibrosis was more severe in those animals. Our study demonstrated that Cat D is responsible for the conversion of AN GEN to Ang I in the primary AECs. However, it is probably not the only enzyme capable of doing so as the complete inhibition of CatD by non-selective protease inhibitor pepstatin A did not completely block bleomycin-induced apoptosis in AECs. Hence, additional protease(s) may be involved in angiotensinogen processing and subsequent AEC apoptosis. Although Cat D activity and protein expression was increased in bleomycin- induced pulmonary fibrosis model in rats (Koslowski et al., 2003; Uhal et al., unpublished data), its roles in pulmonary fibrosis and pulmonary angiotensin peptides generation is still unclear in vivo. Futhermore, in bleomycin induced rat pulmonary fibrosis model, systemically administrated Pepstatin A did not protect the animals from bleomycin induced pulmonary fibrosis argues against that conversion of ANGEN to ANGI by Cat D is the rate limiting step in pulmonary angiotensin synthesis. 5.2 ACE ACE is expressed predominantly by the pulmonary endothelial cells in a membrane- bound form and mainly responsible for generating circulating ANG II from angiotensin I. ACE inhibitors completely blocked apoptosis of AECs induced by Fas L, TNF a, bleomycin and Amiodarone, suggesting that ACE is the only enzyme responsible for converting angiotensin I to angiotensin II in vitro. The in vivo condition could be 169 different though ACE activity was elevated in bronchoalveolar lavage fluid (BALF) and/or serum in many potentially fibrotic lung diseases including idiopathic pulmonary fibrosis (Specks et al, 1990). However, the functional significance of increased ACE activity in those fluids is not clear. It is speculated that increased ACE activity indicates endothelial cell damage causing ACE to dissociate from the surface of injured endothelial cells (Marshall, 2003). There is evidence supporting that ACE is functionally relevant to pulmonary angiotensin generation although pulmonary tissue ACE activity remain unchanged in bleomycin induced pulmonary fibrosis model (Marshall et al., 2004). For example, ACE inhibitor Ramipril decreased pulmonary ANG 11 concentration and reduced bleomycin-induced pulmonary fibrosis in rats. This result is consistent with our previous finding that Captopril, another ACE inhibitor, attenuated bleomycin-induced pulmonary fibrosis (Wang et al., 2000). Although Ramipril completely inhibited ACE activity in the lung, pulmonary ANG II concentration only decreased by half, suggesting that ACE activity represents approximately half of the AN G II-generating capacity in the lung. Other non-ACE enzyme such as chymases could be responsible for a significant proportion of ANG Il-generating activity in the lung as shown in the cardiac tissue (Urata et al., 1990). In addition, chymase was shown to be activated in the pulmonary inflammation and fibrosis induced by paraquat in hamsters (Orito et al., 2004). Taken together, those data argue against that ACE activity underlies the rate-limiting step in ANG II generation within the lung. 5.3 AN GEN and angiotensin peptides: 170 ANGEN is the only known precursor for the synthesis of angiotensin peptides. Our studies demonstrated that AN GEN mRNA expression in primary AECs and A549 cells is upregulated and functional ANGEN mRNA is required for the apoptotic response to F as L, TNF or, bleomycin and Amiodarone as apoptosis of primary AECS and A549 cells was inhibited by the ANGEN antisense but not by the scrambled control oligonucleotides (Li etaL,2003) In lung explant where systemic angiotensin system was removed, bleomycin —induced collagen accumulation ex vivo requires the ANGEN gene expression as ANGEN antisense blocked the collagen deposition in explants. In whole animal model, circulating levels of angiotensinogen are approximately equal to the Michaelis Constant (Km) of renin for its substrate (about 1 uM) (Gould and Green, 1971). Therefore, the rate of angiotensin II synthesis can be regulated by changes in angiotensinogen levels. Since the normal concentration of ANGEN is near the Km for its reaction with renin (Gould and Green 1971), one would expect any change in ANGEN levels to be accompanied by parallel changes in the formation and actions of Ang II. For instance, transgenic mice expressing the rat angiotensinogen gene are hypertensive (Kimura et al., 1992) and mice without angiotensinogen gene expression are hypotensive (Tanimoto et al., 1994). In addition, for any given level of renin activity angiotensin II synthesis can be altered by changes in the concentration of available angiotensinogen (Poulsen and Jacobson, 1993). For example, up-regulation of ANGEN in tissue can alter tissue concentrations of angiotensin II, particularly, in tissues where are not subjected to systemic short loop or long loop feedback control (Poulsen and Jacobson, 1993). In bleomycin induced pulmonary fibrosis model, ANGEN expression is upregulated in the lung shown in 171 chapter 6. Elevation of local pulmonary ANGEN concentration increased pulmonary ANG II concentration. Furthermore, intratracheal delivery of antisense against ANGEN mRNA blocked the apoptosis of alveolar epithelial cells and subsequent fibrosis, which is consistent with the result that local ANG II generation is decreased. Whether the circulating ANGI and ANGII contribute to the pulmonary ANGII is unknown. There is a possibility that tissue can uptake circulating angiotensins through AT1 receptor binding and internalization (Danser, 2004). Uptake from the circulation can be quantified by measuring steady state tissue and plasma levels of 1251—Labelled Ang I and 11 during 1251‘-ANGI and II infusion. The results obtained fiom pigs (van Kats et al. 1997, 1998, 2001) showed that 1251-Ang II, but not 1251-Ang I, accumulated in tissues like heart, kidney and adrenal gland. The absence of significant tissue 125 l-Ang I accumulation indicated that the presence of tissue ANG I cannot be attributed to uptake from circulation. If the same thing occurs in the lung, circulating ANGI does not contribute to the local pulmonary AN GII generation. Accumulation of 125 I-Ang II at tissue sites was largely prevented by pretreatment with an AT 1 receptor antagonist (Danser, 2004), suggesting that uptake of Ang II is mediated via AT 1 receptor-dependent endocytosis. It is unlikely that AT 2 receptors play a role in this process since AT 2 receptors do not internalize Ang II (Matsubara 1998). Similar conclusions about AT1 receptor mediates internalization of ANG II were drawn from Ang II infusion studies in rats (Zou et al. 1996a,b). However, comparison of the 125 l- labelled and endogenous angiotensin levels revealed that, despite the significant uptake of 1251-Ang II in various tissues, the majority (>90%) of tissue Ang II is not derived from circulation. Instead, it is locally synthesized from locally generated Ang I (Danser, 2004). 172 In the lung AN GII uptake may not exist as treatment with losartan increased rather than decreased pulmonary ANG 11 concentration (Marshall, 2004). Taken together, our results together with others’ indicate that elevation of ANGEN concentration in the lung is the most likely rate-limiting step for the pulmonary angiotensin II generation. 6. Therapeutic Implications: 6.1 Existing clinical trials: There have been several clinical trials to examine the therapeutic potentials of angiotensin system antagonists, especially the ACE inhibitors, on IPF as well as other fibrotic lung diseases. The results obtained from those trials, however, are still controversial and inconclusive. For example, Kyung et a1 (2002) conducted a small size clinical study to investigate the clinical efficacies of angiotensin receptor antagonist on treating IPF. Fourteen patients diagnosed with IPF by open lung biopsy and by arnerican thoracic sociatey (ATS) criteria were divided into two groups; the first group were given AT1 antagonist losartan (n=8), the second group were given placebo without losartan (n=6). Although there was no significant difference in serum angiotensin II level between the two groups, patients treated by losartan showed increased Forced Vital capacity (FVC) (by 12%). These results suggest that angiotensin II receptor antagonist may be an effective agent for treating IPF. Uhal et al. evaluated the effects of angiotensin system antagonists on pulmonary fibrosis induced by amiodarone, an antiarrhythmic reagent in which angiotensin system is also considered to play an important role in the pathogenesis of this disease (Uhal et al., 173 unpublished observation). After re-analyzing the CHF-STAT study (Survival Trial of Antiarrythmic Therapy in CHF) (Singh et al., 1997), we found that compared with those who did not receive the ACE inhibitors, there was a higher percentage without lung fibrosis (90.5% vs. 83.3%) and a lower percentage (0.98 vs. 3.3%) with severe pulmonary fibrosis of patients receiving either ACEis or ANG receptor antagonists although the difference was not statistically significant due to the low statistics power. Based on these results, we speculate that there is a “trend” for patients who did not receive ACEis drugs to develop more frequent and severe lung fibrosis. Similarly, we observed that with concurrent administration of the ACE inhibitors there was a trend toward improved survival in patients with long-terrn administration of amiodarone although the difference was not statistically significant clue to the low statistics power (p=0.57). That the CHF-STAT study did not demonstrate statistical differences in pulmonary toxicity of patients receiving ACE inhibitors versus placebo (Boutitie et al., 1999) may be due to the fact that the number of patients receiving placebo was too low to reach sufficient statistical power as they consist only 9% of the total patients. Hence, a larger population of patients treated with amiodarone is needed to determine the protective effects of ACEiS and angiotensin receptors blocker on amiodarone induced pulmonary toxicity (Flaherty et al., 2002). Contradictory to those above findings, results from two other clinical trials performed by different investigators do not suggest the beneficial role of angiotension-converting enzyme inhibitors in treating IPF. The first study by Raghu et al. (Raghu et al., ATS 2004 Mtg, Orlando, FL) employed prospective, randomized, double blind trials to investigate the association of ACE inhibitors with survival and disease progression in patients with 174 IPF. Data were collected from 168 patients, of which 20 were on ACEis and 146 patients were not. The other recently published clinical study by Nadrous et a1 (Nadrous et al., 2004) retrospectively reviewed the effects of ACEis on survival of 478 patients with IPF who visited Mayo Clinic Rochester from 1994 through 1996. 57 patients (12%) received ACEI and the rest of them were not. Both studies found that the mortality rates and rates of disease progression were similar in patients with or without ACEiS treatment, thus suggesting that there is no beneficial effect of ACEis on survival of patients with IPF. However, there are some pitfalls in those two studies. For example, the first study was not specifically designed for evaluating the effects of ACEis on IPF. Instead, the major purpose was to evaluate the effects of interferon gamma-1 b on treatment of IPF. As a retrospective study, the limitations in the second study include lack of blinded randomization for ACEis use, variations in the specific types and doses of ACEis prescribed, varying concurrent therapies for IPF, reliance on clinicoradiologic parameters for case definition in most patients, and incomplete information on duration of ACEis use before establishment of the diagnosis of IPF. In addition, in order to detect a 20% survival difference in a trial evaluating an agent for IPF, the minimum sample size is 700 patients (Mapel et al., 1996). In both studies, the number of patients who received ACEis was relatively small; therefore, those studies may be statistically underpowered to reliably detect a survival benefit of ACEis. In conclusion, the results from clinical trials regarding whether the angiotensin system antagonists have therapeutic effects on IPF are still controversial. Clinical trials involving larger population with randomized and double blind designs need to be done to determine their therapeutic potentials for IPF. 175 6.2 AT1 antagonist may be more efficient than ACE inhibitors in term of antifibrotic effect in vivo AT1 antagonist and ACE inhibitors both completely blocked AECs apoptosis induced by Fas L, TNF alpha and amiodarone, bleomycin in vitro. And we were not able to detect the difference between those two lines of reagents in inhibiting apoptosis. However, there are some in vivo studies suggesting that the AT1 antagonist may be a more efficient antifibrotic drug than ACE inhibitors. For example, it was shown that L-158, 809, a AT1 receptor antagonist, L-158, 809, was more effective in preventing radiation-induced pneumopathy and lung fibrosis than angiotensin-converting enzyme inhibitors captopril and enalapril (Moltine et al., 2000), the mechanism of which, however, was unknown. Theoretically speaking, AT1 receptor antagonist should be a more efficient anti-fibrotic drug than ACE inhibitors in the lung due to the following reasons: 1) AT 1 receptors mediate most of the profibrotic effects in the lung. Therefore, inhibition of AT1 receptors should exert more direct effects than ACE on AT1 receptor-mediated signal transduction. 2) AT 1 receptor antagonists increase the local concentration of ANG 11 (Marshall et al., 2004), which may act through AT2 receptor to exert antifibrotic function as shown in the heart (Danser, 2004). 3) ACE is not the only enzyme to convert ANG I to ANG II in the lung. Thus, ACE inhibitors might not completely block the profibrotic effect of ANG II converted by other enzymes such as chymase 4) ACE inhibitors disturb the bradykinin system, the function of which is unknown in pulmonary fibrosis. 5) Long —-term administration of ACE inhibitors can induce the production of ACE, which will counteract the antifibrotic effects of ACE inhibitors (Boomsma et al., 1981). 176 6.3 Potential therapeutic approaches for IPF: Local administration of ANGEN antisense or AT1 antagonist for IPF: Our studies together with others’ demonstrated that activated pulmonary angiotensin system plays an essential role in the development of pulmonary fibrosis. The potential rate-limiting step in the production of the pulmonary angiotensin system is the local synthesis of angiotensinogen. The AT 1 receptor plays a major role in mediating the profibrotic effect of local ANG II in the lung. Those findings indicate that local administration of ANGEN antisense or AT1 antagonists are potential therapeutic approaches for IPF. The benefits of local administration of those regents are as follows. 6. 3. 1. Local administration does not disturb the systemic angiotensin system Lung is easier to be accessed compared to most of the other organ systems and local delivery of drugs via the airway is feasible as nebulization or aerosolation of medications via inhalation are commonly used clinically. Furthermore, in our study, intratracheal instillation of antisense oligonucleotides against ANGEN mRNA achieved lung specific delivery of the drug as the instilled antisense were contained in the lung and did not affect plasma ANGEN and ANG 11 level. Yet, it is notable that, there exist regional differences in alveolar ventilation resulting in the uneven delivery of the inhaled drugs. Hence, this could cause insufficient delivery to most severely fibrotic areas within the lung (Marshall et al., 2003). Direct instillation through the endo-tracheal tube or via a bronchoscope in patients with IPF could be an alternative approach to compensate for the non-selective delivery of nebulized drug in the lung. 177 6.3.2 Local administration is a more effective approach with fewer side effects High tissue specificity ensures that much lower doses are needed to achieve the same effects for locally administered drugs compared to systemically administered counterparts. In addition, locally administration will cause much fewer side effects. This has been proven by the locally administered steroids for asthma treatment (V ervereli et al., 2004). 7. General Conclusions: (Fig. 7.2) Lung epithelium apoptosis inducer: Blerimycin — Angiotensinogen .L I Antisense of I | ANGEN mRNA 1 CatD * -> Angiotensin I l ACE : @E inhibi® -> Angiotensin II /\ AT receptor antagonists Apoptosis > Fibrosis Pulmonary angiotensin system is activated in bleomycin induced pulmonary fibrosis model. During the process of bleomycin-induced AECs apoptosis, AECs synthesize ANGEN, which is converted to ANG I by cathepsin D (Cat D). ANG I is further converted to ANG II by ACE. Locally produced ANG II mediates apoptosis of the AECs 178 leading to excessive loss of AECS and insufficient epithelial repair. ANG 11 produced by apoptotic epithelial cells also directly activates the fibroblasts. Both effects lead to pulmonary fibrosis. Synthesis of ANGEN is the key upstream event in this process, which can be blocked by the antisense oligonucletides against ANGEN mRNA. Local administration of AN GEN antisense can be used, as a potential therapeutic approach for treating PF. AT1 receptor is required for bleomycin- induced apoptosis and fibrosis and thus can also be a potential therapeutic target for PF. 179 REFERENCE Adamson IYR and Bowden DH. (1976) Pulmonary injury and repair: organ culture studies of murine lung after oxygen. Arch Pathol Lab Med 100: 640-643 Adamson IYR, Young L, et al. (1988) Relationship of alveolar epithelial injury and repair to the induction of pulmonary fibrosis Am J Pathol 130:377—3 83 Adler V, Schaffer A, et al. (1995) UV irradiation and heat shock mediate JNK activation via alternate pathways J Biol Chem 270: 26071-26077 Admiraal PJ, Danser AH, et al. (1993) Regional angiotensin 11 production in essential hypertension and renal artery stenosis Hypertension 21 : 173-84 Ali S, Leonard SA, et al. (2001) Absorption, distribution, metabolism, and excretion of a respirable antisense oligonucleotide for asthma Am J Respir Crit Care Med 163: 989-993 Allen RT, William III JH, et al. (1997) Morphological and biochemical characterization of apoptosis J Pharm Toxicol Meth 37: 215—228 American Thoracic Society (ATS), and the European Respiratory Society (ERS). (2000) Idiopathic pulmonary fibrosis: diagnosis and treatment. International consensus statement Am J Respir Crit Care Med 161: 646-664 Antoniades HN, Avila RB, et al. (1990) Platelet-derived growth factor in idiopathic pulmonary fibrosis. J Clin Invest 86: 1055-1064 Antus B, Mucsi I, et al. (2000) Apoptosis and inhibition of cellular proliferation by ANG 11: possible implication and perspective. Acta Physiol Hung 87: 5—24 Aoshiba K, Yasui S, et al. (2000) The Fas/Fas-ligand system is not required for bleomycin-induced pulmonary fibrosis in mice. Am J Respir Crit Care Med 162: 695-700 Bader M, Paul M, et al. (1994) Renin -angiotensin system A: molecular biology of the renin- angiotensin system. In: Swales JD, ed. Textbook o f Hypertension. Oxford: Blackwell Scientific Publications, 214-232 Barbas-Filho J, Ferreira M, et al. (2001) Evidence of type II pneumocyte apoptosis in the pathogenesis of idiopathic pulmonary fibrosis (IPF)/usual interstitial pneumonia (UIP) J Clin Pathol 54:132-138 Bargout R, Jankov A, et al. (2000) Amiodarone induces apoptosis of human and rat AECS in vitro Am J Physiol Lung Cell Mol Physiol 278: L1039-1044 180 Basset F, Soler P, et al. (1986) Importance of cellular data from broncho-alveolar lavage in interstitial pulmonary pathology Bull Acad Natl Med 170: 525-529 Basset G, Crone C, et al. (1987) Significance of active ion transport in transalveolar water absorption: a study on isolated rat lung J Physiol 384: 31 1-3 24 Battegay EJ, Colbert T, et al. (1995) TNF-alpha stimulation of fibroblast proliferation: Dependence on platelet-derived growth factor (PDGF) secretion and alteration of PDGF receptor expression J Immunol 154: 6040-6047 Baumgartner KB, Samet JM, et al. (2000) Occupational and environmental risk factors for idiopathic pulmonary fibrosis: a multicenter case-control study. Collaborating Centers Am J Epidemiol 152(4): 307-315 Bhat GJ, Thekkurnkara TJ, et al. (1994) Angiotensin II stimulates sis-inducing factor-like DNA binding activity. Evidence that the ATlA receptor activates transcription factor- Stat9l and/or a related protein J Biol Chem 269(50): 31443-31449 Blum RH, Carter SK, et al. (1973) A clinical review of bleomycin--a new antineoplastic agent Cancer 31(4): 903-914 Boggs SS, Sartiano GP, et al. (1974) Minimal bone marrow damage in mice given bleomycin Cancer Res 34(8): 1938-1942 Boomsma F, de Bruyn JH, et al. (1981) Opposite effects of captopril on angiotensin I- converting enzyme 'activity' and 'concentration'; relation between enzyme inhibition and long-term blood pressure response Clin Sci (Lond) 60(5): 491-498 Borok Z, Gillissen A, et a1. (1991) Augmentation of functional prostaglandin E levels on the respiratory epithelial surface by aerosol administration of prostaglandin E Am Rev Respir Dis 144(5): 1080-1084 Brasier A, Junyi L, et a1. (1994) Transcription factors modulating angiotensinogen gene expression in hepatocytes Kidney International. 46: 1564-1566 Brown LF, Dvorak AM, et al. (1989) Leaky vessels, fibrin deposition, and fibrosis: a sequence of events common to solid tumors and to many other types of disease Am Rev Respir Dis 140(4): 1104-1107 Bullock GR, Steyaert I, et al. (2001) Distribution of type-l and type-2 angiotensin receptors in the normal human lung and in lungs from patients with chronic obstructive pulmonary disease Histochem Cell Biol 115(2): 117-1124 Burson JM, Aguilera G, et al. (1994) Differential expression of angiotensin receptor 1A and 1B in mouse. Am J Physiol 267(2 Pt 1): E260-267 181 Campbell JM, Bacon TA, et al. (1990) Oligodeoxynucleoside phosphorothioate stability in subcellular extracts, culture media, sera and cerebrospinal fluid J Biochem Biophys Methods 20: 259-267 Campbell DJ and Habener JF. (1986) ANGEN gene is expressed and differentially regulated in multiple tissues of the rat. J Clin Invest 78: 31-39 Canington C, Coutu R, et al. (1978) Natural history and treated course of usual and desquamative interstitial pneumonia. N Engl J Med 298: 801-809 Chapman HA, Allen CL, et al. (1986) Abnormalities in pathways of alveolar fibrin turnover among patients with interstitial lung disease. Am Rev Respir Dis 133(3): 437- 443 Cott GR, Sugahara K, et al. (1986) Stimulation of net active ion transport across alveolar type 11 cell monolayers Am J Physiol 250(2 Pt 1): C222-227 Coultas DB, Black WC, et a1. (1994) The epidemiology of interstitial lung disease. Am J Respir Crit Care Med 150:967-972 Crouch E. (1990) Pathobiology of pulmonary fibrosis. Am J Physiol 259(4 Pt 1): L159- 184. Crystal RG, Bitterrnan PB, et al. (2002) Future research directions in idiopathic PF: summary of a National Heart, Lung, and Blood Institute working group. Am J Respir Crit Care Med 166: 236-246 Crystal RG, Roberts WC, et al. (1976) Idiopathic pulmonary fibrosis: Clinical, histologic, radiographic, physiologic, scintigraphic, cytologic, and biochemical aspects. Ann Intern Med 85: 769-788 Danahay H, Giddings J, et al. (1999) Distribution of a 20-mer phosphorothioate oligonucleotide, CGP69846A (ISIS 5132), into airway leukocytes and epithelial cells following intratracheal delivery to brown-norway rats. Pharm Res 16:1542-1549 Danser AHJ (2004). Renin-angiotensin system: plasma versus tissue. In: Unger T and Schdlkens BA, ed. Angiotensin. Berlin, New York: Springer, 129-147 Danser AH, Derkx FH, et al. (1994) Angiotensin levels in the eye. Invest Ophthalmol Vis Sci 35: 1008-1018 Day RM, Yang Y, et al. (2001) Bleomycin upregulates gene expression of angiotensin- converting enzyme via mitogen-activated protein kinase and early growth response 1 transcription factor. Am J Respir Cell Mol Biol 25: 613-619 182 Dechend R, Wallukat G, et al. (2000). AT (1) receptor agonistic antibodies from preeclamptic patients cause vascular cells to express tissue factor Circulation 101 :2382- 2387 Deiss LP, Galinka H, et al. (1996) Cathepsin D protease mediates programmed cell death induced by interferon-gamma, Fas/APO-l and TNF-alpha EMBO J 15: 3861-3870 Deshmukh R, Smith A, et al. (1998) Hypertension. In: Lilly LS, ed. Pathophysiology of Heart Disease; Second edition. Baltinmore: Williams and Wilkins, 267-288 Deterding RR, Havill AM, et al. (1997) Prevention of bleomycin-induced lung injury in rats by keratinocyte growth factor Proc Assoc Am Physicians 109:254-268 Dimmeler S, Rippmann V, et al. (1997) Angiotensin II induces apoptosis of human endothelial cells, Protective effect of nitric oxide C irc Res 81 :970—976 Dlugi AM, Robie KM, et al. (1974) Failure of bleomycin to affect humoral or cell- mediated immunity in the mouse Cancer Res 34(10): 2504-2507 Dohi M, Hasegawa T, et a1. (2000) Hepatocyte growth factor attenuates collagen accumulation in a murine model of pulmonary fibrosis. Am J Respir Crit Care Med 162:2302-2307 Domagala-Kulawik J, Droszcz P, et al. (2000) Expression of Fas antigen in the cells from bronchoalveolar lavage fluid (BALF) Folia Histochem Cytobiol 38: 185-188 Douglas WW, Swensen SJ, et al. (1998) Colchicine versus prednisone in the treatment of idiopathic pulmonary fibrosis. A randomized prospective study. Members of the Lung Study Group Am J Respir Crit Care Med 158: 220-225 Dzau VJ, Brenner A, et al. (1982) Evidence for renin in rat brain: differentiation from other renin-like enzymes Am J Physiol 242: E292-297 Eggena P, Zhu J, et al. (1993) Nuclear angiotensin receptors induce transcription of renin and angiotensinogen mRN A Hypertension 22: 496-501 Eitzman DT, Zheng X, et al (1996) Bleomycin-induced pulmonary fibrosis in transgenic mice that either lack or overexpress the murine plasminogen activator inhibitor-1 gene. J Clin Invest 97:232-237 Erdos EG. (1990) Angiotensin I converting enzyme and the changes in our concepts through the years. Lewis K. Dahl memorial lecture Hypertension 16(4): 363-3 70 Fang X, Zhu Y, et al. (2002) Losartan in the rat model of bleomycin-induced pulmonary fibrosis and its impact on the expression of monocyte chemoattractant protein-1 and basic fibroblast growth factor. Zhonghua Jie He He Hu Xi Za Zhi 25:268—272 183 Fehrenbach H, Kasper M, et al. (2000) Alveolar epithelial type 11 cell apoptosis in vivo during resolution of keratinocyte growth factor-induced hyperplasia in the rat Histochem Cell Biol 114:49—61 Flaherty KR, Toews GB, et al. (2001) Steroids in idiopathic pulmonary fibrosis: a prospective assessment of adverse reactions, response to therapy, and survival Am J Med 110(4): 278-282 F laherty KR, Toews GB, et al. (2002) Clinical significance of histological classification of idiopathic interstitial pneumonia Eur Respir J 19(2): 275-283 Flaherty KR, Travis WD, et al. (2003) Fibroblastic foci in usual interstitial pneumonia: idiopathic versus collagen vascular disease. Am J Respir Crit Care Med 167:1410-1415 Filippatos GS, Gangopadhyay N, et a1. (2001) Regulation of apoptosis by vasoactive peptides Am J Physiol Lung Cell M01 Physiol 281: L749-761 Filippatos G, Tilak M, et al. (2001) Regulation of apoptosis by ANG II in the heart and lungs Int J Mol Med 7: 273-280 Filippatos G and Uhal BD. (2003) Blockade of apoptosis by ACE inhibitors and angiotensin receptor antagonists Curr Pharm Design 9:707—714 Fine A, Anderson NL, et al. (1997) Fas expression in pulmonary alveolar type 11 cells. Am J Physiol 273:L64-71 Fine A, Uhal BD, et al. (2000) Apoptosis in lung pathophysiology. Am J Physiol Lung Cell Mol Physiol 279(3): L423-427 Fonseca C, Abraham D, et a1. (2000) Lung fibrosis Springer Semin lmmunopathol 21:453-474 Gallin JI, Goldstein IM, et al. (1992) Inflammation: Basic principles and clinical correlates, Second edition, Raven press, Chapter 5: 991-997 Gauldie J, Kolb M, et al. A new direction in the pathogenesis of pulmonary fibrosis Respir Res 2002, 3: 1—3 Geisterfer AA, Peach MJ, et al. (1988) Angiotensin II induces hypertrophy, not hyperplasia, of cultured rat aortic smooth muscle cells Circ Res 62(4): 749-56 Giaid A, Hamid Q, et al. (1993) Expression of endothelin-1 in lungs of patients with cryptogenic fibrosing alveolitis Lancet 341: 1550-1554 184 Goldstein R and Polgar P. (1982) The effect and interaction of bradykinin and prostaglandins on protein and collagen production by lung fibroblasts J Biol Chem 257: 8630-8633 Goodman BE and Crandall ED. (1982) Dome formation in primary cultured monolayers of alveolar epithelial cells Am J Physiol 243(1): C96-100 Goodman BE, Fleischer RS, et al. (1983) Evidence for active Na+ transport by cultured monolayers of pulmonary alveolar epithelial cells Am J Physiol 245(1): C78-83 Gorin AB and Stewart PA (1979) Differential permeability of endothelial and epithelial barriers to albumin flux. J Appl Physiol 47(6): 1315-1324 Gould AB and Green D (1971) Kinetics of the human renin and human substrate reaction Cardiovasc Res 5(1): 86-89 Griendling K and Alexander RW. (1994) Renin —angiotensin system C: Cellular mechanisms of angiotensin 11 action. In: Swales JD, ed. Textbook o f Hypertension. Oxford: Blackwell Scientific Publications 214-232 Gross TJ, Simon RH, et al. (1991) Rat alveolar epithelial cells concomitantly express plasminogen activator inhibitor-1 and urokinase Am J Physiol 260(4 Pt 1): L286-295 Gross TJ, Simon RH, et al. (1990). Expression of urokinase-type plasminogen activator by rat pulmonary alveolar epithelial cells Am J Respir Cell Mol Biol 3(5): 449-456 Gross TJ and Sitrin RG. (1990) The THP-l cell line is a urokinase-secreting mononuclear phagocyte with a novel defect in the production of plasminogen activator inhibitor-2 J Immunol 144(5): 1873-1879 Gross TJ, Sitrin RG. (1990) Expression of urokinase-type plasminogen activator by rat pulmonary alveolar epithelial cells Am J Respir Cell Mol Biol; 3: 449-456 Guerry-Force ML, Wright JL, et a1. (1987) A comparison of bronchiolitis obliterans with organizing pneumonia, usual interstitial pneumonia, and small airways disease Am Rev Respir Dis 135: 705-712 Guinee D Jr, Brambilla E, et al. (1997) The potential role of BAX and BCL-2 expression in diffuse alveolar damage. Am J Pathol 151:999-1007 Guyton AC and Moffatt DS. (1981) Role of surface tension and surfactant in the transepithelial movement of fluid and in the development of pulmonary edema Prog Respir Res 15: 62-75 185 Gyurko R, Wielbo D, et al. (1993) Antisense inhibition of AT1 receptor mRNA and angiotensinogen mRNA in the brain of spontaneously hypertensive rats reduces hypertension of neurogenic origin Regul Pept 49(2): 167-174 Hagimoto N, Kuwano K, et al. (1997) Induction of apoptosis and pulmonary fibrosis in mice in response to ligation of FAS antigen Am J Respir Cell Mol Biol 17:272-278 Hagimoto N, Kuwano K, et al. (1997) Apoptosis and expression of Fas/F as ligand mRN A in bleomycin-induced pulmonary fibrosis in mice Am J Respir Cell Mol Biol 16291-101 Hamilton RF, Li L, et al. (1995) Bleomycin induces apoptosis in human alveolar macrophages. Am J Physiol Lung Cell Mol Physiol 269: L318-L325 Haller H, Lindschau C, et al. (1996) Effects of intracellular angiotensin II in vascular smooth muscle cells C irc Res 79: 765-772 Harrison DG, Cai H, et al. (2003) Interactions of angiotensin II with NAD (P) H oxidase, oxidant stress and cardiovascular disease J Renin Angiotensin Aldosterone Syst 4:51—61 Haschek WM and Witschi H. (1979) PF--a possible mechanism Toxicol Appl Pharmacol 51: 475-87 Hasegawa T, Marshall BC, et al. (1997) Induction of urokinase-type plasminogen activator receptor by IL-1 beta Am J Respir Cell Mol Biol 16: 683-692 Hasday JD, Bachwich PR, et al. (1988) Procoagulant and plasminogen activator activities of bronchoalveolar fluid in patients with pulmonary sarcoidosis Exp Lung Res 14(2): 261- 278 Hasegawa T, Sorensen L, et al. (1997) Induction of urokinase-type plasminogen activator receptor by IL-1 beta Am J Respir Cell Mol Biol 16(6): 683-692 He Y, Martin WJ, et al. (2001) Expression of yeast apurinic/apyridimidinic endonuclease (APNl) protects lung epithelial cells from bleomycin toxicity Am J Resp Cell Mol Biol 25:692-698 Heinrich M, Schutze S, et al. (1999) Cathepsin D targeted by acid sphingomyelinase- derived cerarnide EMBO J, 18:5252-5263 Hubbard R, Britton J, et a1. (1996) Occupational exposure to metal or wood dust and aetiology of cryptogenic fibrosing alveolitis Lancet 347: 284-289 Ichikawa T, Matsuda A, et al. (1967) Biological studies on bleomycin A J Antibiot (Tokyo) 20(3): 149-155 186 Ichikawa T, Nakano I, et al. (1969) Bleomycin treatment of the tumors of penis and scrotum J Urol 102(6): 699-707 Idell S, James K, et al. (1989) Local abnormalities in coagulation and fibrinolytic pathways predispose to alveolar fibrin deposition in the adult respiratory distress syndrome J Clin Invest 84(2): 695-705 Inoshima I, Kuwano K, et al. (2004) Anti-monocyte chemoattractant protein-1 gene therapy attenuates pulmonary fibrosis in mice. Am J Physiol Lung Cell Mol Physiol 286:L1038-1044 Ip YT and Davis RJ. (1998) Signal transduction by the c-Jun N-terminal kinase (JNK)- from inflammation to development Curr Opin Cell Biol 10: 205-219 Isahara K, Ohsawa Y, et al. (1999) Regulation of a novel pathway for cell death by lysosomal aspartic and cysteine proteinases Neuroscience 91 :233-249 Ito M, Oliverio MI, et al. (1995) Regulation of blood pressure by the type 1A ANG II receptor gene. Proc Natl Acad Sci U S A, 92: 3521-3525 Iwai K, Hosoda Y, et al. (1994) Idiopathic pulmonary fibrosis. Epidemiologic approaches to occupational exposure. Am J Respir Crit Care Med 150: 670-675 Janssen YM and Sen CK. (1999) Nuclear factor kappa B activity in response to oxidants and antioxidants Methods Enzymol 300: 363-374 Johnston IDA, Rudd RM, et al. (1997) British Thoracic Society study of cryptogenic fibrosing alveolitis: current presentation and initial management. Fibrosing Alveolitis Subcommittee of the Research Committee of the British Thoracic Society. Thorax. 52:38-44 Kagiyama S, Varela A, et al. (2001) Antisense inhibition of brain renin-angiotensin system decreased blood pressure in chronic 2-kidney, 1 clip hypertensive rats Hypertension 37(2 Part 2): 371-375 Kanno K, Chayama K, et al. (2003) ATlA-deficient mice Show less severe progression of liver fibrosis induced by CC1(4) Biochem Biophys Res Commun 308: 177—183 Kapanci Y, Gabbiani G, et al. (1995) Cytoskeletal protein modulation in pulmonary alveolar myofibroblasts during idiopathic pulmonary fibrosis. Possible role of transforming growth factor beta and tumor necrosis factor alpha Am J Respir Crit Care Med 152: 2163-2169 Kasper M, Lackie P, et al. (1996) Immunolocalization of cathepsin D in pneumocytes of normal human lung and in PF. Virchows Arch 428(4-5): 207-215 187 Kasper M, Schinzel R, et a1. (1999) Experimental induction of AGES in fetal L132 lung cells changes the level of intracellular cathepsin D. Biochem Biophys Res Commun 261: 175-182 Katwa LC, Ratajska A, et a1. (1995) Angiotensin converting enzyme and kininase lI-like activities in cultured valvular interstitial cells of the rat heart Cardiovasc Res 29:57-64 Kazerooni EA, Flint A, et al. (1997) Thin-section CT obtained at lO-mm increments versus limited three-level thin-section CT for idiopathic pulmonary fibrosis: correlation with pathologic scoring. Am J Roentgenol 169: 977-983 Kazufumi M, Sonoko N, et al. (1997) Expression of bcl-2 protein and APO-1 (Fas antigen) in the lung tissue from patients with idiopathic pulmonary fibrosis. Microsc Res Tech 38:480-487 Kelly BG, Hasleton PS, et al. (2002) A rearranged form of Epstein-Barr virus DNA is associated with idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 166:510-513 Khalil N, Unruh H, et al. (1996) TGF-beta l, but not TGF-beta 2 or TGF-beta 3, is differentially present in epithelial cells of advanced pulmonary fibrosis: an immunohistochemical study. Am J Respir Cell Mol Biol 14: 131-138 Kim HS, Krege JH, et al. (1995) Genetic control of blood pressure and the angiotensinogen locus. Proc Natl Acad Sci U S A 92: 2735-2739 Kirnura I, Onoshi T, et a1. (1972) Treatment of malignant lymphomas with bleomycin Cancer 29(1): 58-60 Kirnura S, Mullins JJ, et al. (1992) High blood pressure in transgenic mice carrying the rat angiotensinogen gene. Embo J 11(3): 821-827 King TE Jr., Brown K, et a1. (2001) Idiopathic pulmonary fibrosis: relationship between histopathologic features and mortality. Am J Respir Crit Care Med 164:1025-1032 King TE Jr., Costabel U, et al. (2000) Idiopathic Pulmonary Fibrosis: Diagnosis and Treatment lntemational Consensus Statement Am J Respir Crit Care Med 161: 646-664 Koslowski R, Knoch K, et al. (2003) Cathepsins in bleomycin induced lung injury in rat. Eur Respir J 22:427-435 Kotani I, Takada A, et al. (1995) Increased procoagulant and antifibrinolytic activities in the lungs with idiopathic pulmonary fibrosis Thromb Res 77: 493-504 Kuhn C., Boldt J, et al. (1989) An immunohistochemical study of architectural remodeling and connective tissue synthesis in pulmonary fibrosis Am Rev Respir Dis 140(6): 1693-703 188 Kuhn C and McDonald JA. (1991) The roles of the myofibroblast in idiopathic pulmonary fibrosis. Ultrastructural and immunohistochemical features of sites of active extracellular matrix synthesis Am J Pathol 138: 1257-1265 Kuwano K, Kunitake R, et al. (1996) P21 and p53 expression in association with DNA strand breaks in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 154: 477-483 Kuwano K, Hagimoto N, et al. (1999) Essential roles of the F as/F as ligand pathway in the development of pulmonary fibrosis. J Clin Invest 104: 13-19 Kuwano K, Hagimoto N, et al. (2000) Expression of apoptosis-regulatory genes in epithelial cells in pulmonary fibrosis in mice. J Pathol 190:221-229 Kuwano K, Kunitake R, et al. (2001) Attenuation of bleomycin-induced pneumopathy in mice by a caspase inhibitor. Am J Physiol Lung Cell Mol Physiol 280:L316-325 Kuwano K, Maeyama T, et al. (2002) Increased circulating levels of soluble Fas ligand are correlated with disease activity in patients with fibrosing lung diseases. Respirology 7:15-21 Kyung SY, lim YH, et a1. (2003) The clinical efficacy of angiotensin II receptor antagonist on idiopathic pulmonary fibrosis Am J Respir Crit Care Med 167: A166 (abstract) Lai K, Leung J, et a1. (1998) Gene expression of the renin-angiotensin system in human kidney. J Hypertens 16:91 -102 Lama V, Peters-Golden M, et al. (2002) Prostaglandin E2 synthesis and suppression of fibroblast proliferation by alveolar epithelial cells is cyclooxygenase-Z-dependent. Am J Respir Cell Mol Biol 27: 752-758 Lander HM, Jacovina AT, et al. (1996) Differential activation of mitogen-activated protein kinases by nitric oxide-related species. J Biol Chem 271: 19705-19709 Lee YC, Chen KT, et al. (2000) Gene polymorphisms of endothelial nitric oxide synthase and angiotensin-converting enzyme in patients with asthma Allergy 55:959—963 Lee MW and Severson DL. (1994) Signal transduction in vascular smooth muscle: diacylglycerol second messengers and PKC action. Am J Physiol 267(3 Pt 1): C659-678 Lehane DE, Hurd E, et al. (1975) The effects of bleomycin on immunocompetence in man. Cancer Res 35(10): 2724-2728 Leri A, Claudia PP, et al. (1998) Stretch-Mediated Release of Angiotensin II Induces Myocyte Apoptosis by Activating p53 That Enhances the Local Renin-Angiotensin 189 System and Decreases the Bcl-2-to-Bax Protein Ratio In the Cell. J Clin Invest 101:1326-1342 Li B, Philips MI, et al. (1997) Uptake and efflux of intact antisense phosphorothioate deoxyoligonucleotide directed against angiotensin receptors in bovine adrenal cells. Neurochem Int 31 :393-403 Li D, Philips MI, et al. (1999) Proapoptotic effects of ANG II in human coronary artery endothelial cells: role of AT1 receptor and PKC activation. Am J Physiol 276(3 Pt 2): H786-792 Li HP, Kaplan AP, et a1. (2004) The influence of dexamethasone on the proliferation and apoptosis of pulmonary inflammatory cells in bleomycin-induced pulmonary fibrosis in rats. Respirology 9:25-32 Li X, Uhal BD, et al. (2003) Essential roles for angiotensin receptor ATla in bleomycin- induced apoptosis and lung fibrosis in mice. Am J Pathol 163: 2523-2530 Li X, Uhal BD, et al. (2003) Bleomycin-induced apoptosis of alveolar epithelial cells requires angiotensin synthesis de novo. Am J Physiol Lung Cell Mol Physio 284: L501- 507, Lindpaintner K, Ganten D, et a1. (1988) Intracardiac generation of angiotensin and its physiologic role. Circulation 77(6 Pt 2): 118-23 Low RB. (1989) Bronchoalveolar lavage lipids in idiopathic pulmonary fibrosis Chest 95(1):3-5 Mannino DM, Parrish RG, et al. (1996) Pulmonary fibrosis deaths in the United States, 1979-1991. An analysis of multiple-cause mortality data. Am J Respir Crit Care Med. 153:1548-1552 Mapel DW, Coultas DB, et al. (1998) Idiopathic PF: survival in population based and hospital based cohorts. Thorax 53: 469-476 Mapel DW, Samet JM, et al. (1996). Corticosteroids and the treatment of idiopathic pulmonary fibrosis. Past, present, and future. Chest 110(4): 1058- 1067 Marshall RP. (2003) The pulmonary renin-angiotensin system. Curr Pharm Des 9(9): 715-722 Marshall R, Laurent G, et al. (2000) Angiotensin II is mitogenic for human lung fibroblasts via activation of the type I receptor. Am J Respir Crit Care Med 161:1999— 2004 190 Marshall RP, Laurent GJ, et al. (2004) Angiotensin II and the fibroproliferative response to acute lung injury Am J Physiol Lung Cell Mol Physiol. 286:L156-164 Martin I, Humbert M, et al. (2002) Plasminogen activation by blood monocytes and alveolar macrophages in primary pulmonary hypertension Blood Coagul Fibrinolysis 13(5): 417-22. Mason RJ, Williams MC, et al. (1982) Transepithelial transport by pulmonary alveolar type 11 cells in primary culture Proc Natl Acad Sci U S A 79(19): 6033-6037 Mason RJ, Schwarz MI, et al. (1999) NHLBI Workshop Summary. Pharmacological therapy for idiopathic PF. Past, present, and future Am J Respir Crit Care Med 160(5 Pt 1): 1771-1777 Mason RJ and Williams MC (1991) Type II alveolar epithelial cells In: The Lung: Scientific Foundations (Crystal, R. and West, J .B., Eds.) Raven Press 1: 235-241 Matsubara H. (1998) Pathophysiological role of angiotensin 11 type 2 receptor in cardiovascular and renal diseases Circ Res 83(12): 1182-1191 Matsuoka H, Arai T, et al. (2002) p38 MAPK inhibitor, FR-167653, ameliorates murine bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 283:L103- 112 Matute-Bello G, Liles WC, et al. (2001) Recombinat human Fas ligand induces alveolar epithelial cell apoptosis and lung injury in rabbits. Am J Physiol 281 :L328-L335 Matthay MA, and Wiener-Kronish JP (1990) Intact epithelial barrier function is critical for the resolution of alveolar edema in humans. Am Rev Respir Dis 142(6 Pt 1): 1250- 1257 McCormack F X, King Jr. TE, et al. (1991) Idiopathic pulmonary fibrosis. Abnormalities in the bronchoalveolar lavage content of surfactant protein A. Am Rev Respir Dis 144(1): 160-166 McGee MP and Rothberger H. (1985) Tissue factor in bronchoalveolar lavage fluids. Evidence for an alveolar macrophage source. Am Rev Respir Dis 131(3): 331-336 Mezzano S, Ruiz-Ortega M, et a1. (2001) Angiotensin II and renal fibrosis Hypertension 38:635—638 Molteni A, Moulder JE, et al. (2000) Control of radiation-induced pneumopathy and lung fibrosis by angiotensin-converting enzyme inhibitors and an ANG 11 type 1 receptor blocker. Int J Radiat Biol 76: 523-532 191 Molteni A, Ward WF, et al. (1985) Monocrotaline-induced PF in rats: amelioration by captopril and penicillamine. Proc Soc Exp Biol Med 180: 112-120 Morrison CD, Papp AC, et al. (2001) Increased D allele frequency of the angiotensin- converting enzyme gene in PF. Hum Pathol 32: 521-528 Mullen J, Hodgson MJ, et al. (1998) Case-control study of idiopathic pulmonary fibrosis and environmental exposures. J Occup Environ Med 40(4): 363-3 67 Muller DN, Dechend R, et al. (2000) Angiotensin II (AT(I)) receptor blockade reduces vascular tissue factor in angiotensin II-induced cardiac vasculopathy. Am J Patho 1157: 111-122 Mundle S, Iftikhar A, et al. (1994) In situ end labeling of DNA to detect apoptotic cell death in a variety of human tumors. Cell Death Difler 1: 117-122 Murphy TJ, Alexander RW, et al. (1991) Isolation of a cDNA encoding the vascular type-1 angiotensin II receptor. Nature 351(6323): 233-6 Nadrous HF, Ryu JH, et al. (2004) Impact of angiotensin-converting enzyme inhibitors and statins on survival in idiopathic pulmonary fibrosis. Chest 126(2): 438-446 Nash JR, Corrin B, et al. (1993) Expression of tumour necrosis factor-alpha in cryptogenic fibrosing alveolitis. Histopathology 22: 343-347 Ng K and Vane JR. (1968) The conversion of angiotensin I to angiotensin II in vivo Naunyn Schmiedebergs Arch Exp Pathol Pharmakol 259(2): 188-189 Nguyen L, Ward WF, et al. (1994) Captopril inhibits proliferation of human lung fibroblasts in culture: a potential antifibrotic mechanism. Proc Soc Exp Biol Med 205: 80- 84 Nicholson AG, Wells AU, et al. (2002) The relationship between individual histologic features and disease progression in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 166: 173-177 Nishimura H, Masuda H, et al. (1997) Angiotensin 11 increases plasminogen activator inhibitor-1 and tissue factor mRNA expression without changing that of tissue type plasminogen activator or tissue factor pathway inhibitor in cultured rat aortic endothelial cells. Thromb Haemost 77:1189-1195 Nobukuni S, Inoue J, et a1. (2002) Cigarette smoke inhibits the growth of lung fibroblasts from patients with pulmonary emphysema. Respirology 7:217-223 192 Ohashi H, Takagi H, et a1. (2004) Phosphatidylinositol 3-kinase/Akt regulates angiotensin II- induced inhibition of apoptosis in microvascular endothelial cells by governing survivin expression and suppression of caspase-3 activity. C irc Res 94(6): 785- 793 Ohkubo H, and Nakanishi S. (1986) Tissue distribution of rat ANGEN mRNA and structural analysis of its heterogeneity. J Biol Chem 261(1): 319-323 Oparil S, Tregear GW, et al. (1971) Mechanism of pulmonary conversion of angiotensin I to angiotensin II in the dog Circ Res 29(6): 682-690 Orito K, Akahori F, et a1. (2004) Chymase is activated in the pulmonary inflammation and fibrosis induced by paraquat in hamsters. Tohoku J Exp Med. 203(4): 287-294 Ortiz LA, Moroz K, et al. (1998) Alveolar macrophage apoptosis and TNF-E1, but not p53, correlate with murine response to bleomycin. Am J Physiol 275: L1208-L1218 Otsuka M, Takahashi H, et al. (2004) Reduction of bleomycin induced lung fibrosis by candesartan cilexetil, an angiotensin 11 type 1 receptor antagonist. Thorax 59(1): 31-38 Paine R, and Simon RA. (1996) Expanding the frontiers of lung biology through the creative use of alveolar epithelial cells in culture. Am J Physiol 270: L484-L486 Panos R, King T, et al. (1990) Clinical deterioration in patients with idiopathic pulmonary fibrosis: causes and assessment. Am J Med 88: 396-404 Pantelidis P, Du Bois RM, et a1. (2001) Analysis of tumor necrosis factor-alpha, lymphotoxin-alpha, tumor necrosis factor receptor 11, and interleukin-6 polymorphisms in patients with idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 163: 1432-1436 Panoskaltsis-Mortari A and Bucy RP. (1995) In situ hybridization with digoxigenin RNA probes: fact and artifacts. Biotechniques 18:300-3 07 Papp M, Li X, et al. (2002) Angiotensin receptor subtype AT(1) mediates AEC apoptosis in response to ANG 11. Am J Physiol Lung Cell Mol Physiol 282: L713-718 Pardo A, Selman M, et a1. (1997) Lung AECS synthesize interstitial collagenase and gelatinases A and B in vitro. Int J Biochem Cell Biol 29: 901-910 Peng JF, Kirnura B, et al. (1998) Reduction of cold-induced hypertension by antisense oligodeoxynucleotides to angiotensinogen mRNA and AT1-receptor mRNA in brain and blood Hypertension 31(6): 1317-1323 Phillips MI, Gyurko R, et al. (1994) Antisense inhibition of hypertension: a new strategy for renin-angiotensin candidate genes. Kidney International 46:1554-1556 193 Phillips MI. (2001) Gene therapy for hypertension: sense and antisense strategies. Expert Opin Biol Ther 1(4): 655-662 Pick R, Weber K, et al. (1989) The fibrillar nature and structure of isoproterenol-induced myocardial fibrosis in the rat. Am J Pathol 134: 365-371 Pierzchalski, Anversa P, et a1 (1997) p53 induces myocyte apoptosis via activation of the renin-angiotensin system. Exp Cell Res 234:57-65 Ponte P, Kedes L, et al. (1984) Evolutionary conservation is in the untranslated regions of actin mRNAs: DNA sequence of a human III-actin cDNA Nuc Acid Res12: 1687-1696 Poulsen K and Jacobson J. (1993) Enzymatic reactions of the renin-angiotensin system In: Robertson JIS, Nicholls MG, eds. The renin- angiotensin system: Volume one: Biochemistry and Physiology. New York: Gower Medical Publishing; P.5.1-5.12 Proudn D (1991) Production and metabolism of kinnins. In: Crystal, RG. and West, J .B., Editors, The Lung: Scientific Foundations, Raven, New York, 61—68 Raghu G, Cain K, et al. (1991) Azathioprine combined with prednisone in the treatment of idiopathic pulmonary fibrosis: a prospective double-blind, randomized, placebo- controlled clinical trial. Am Rev Respir Dis 144: 291 -296 Raghu G, Safrin S, et al. (2004) Association of statin and angiotensin-converting enzyme inhibitor use with survival and disease progression in patients with idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 169: A706 (abstract) Robertson JIS, Nicholls MG, eds. (1993) The renin- angiotensin system: Volume one: Biochemistry and Physiology. Gower Medical Publishing Re RN. (2004) Tissue renin angiotensin systems Med Clin North Am 88(1): 19-38 Sasaki K, Yamano Y, et al. (1991) Cloning and expression of a complementary DNA encoding a bovine adrenal angiotensin II type-1 receptor. Nature 351(6323): 230-233 Sato E, Koyama S, et al. (1999) Bleomycin stimulates lung epithelial cells to release neutrophil and monocyte chemotactic factors. Am J Physiol 276:L941-L950 Saraste A and Pulkki K. (2000) Morphological and biochemical hallmarks of apoptosis Cardiovasc Res 45: 528—537 Sausville EA, Stein RW, et al. (1978) Properties and products of the degradation of DNA by bleomycin and iron (II) Biochemistry 17(14): 2746-2754 Schaeffer HJ and Weber MJ. (1999) Mitogen-activated protein kinases: specific messages from ubiquitous messengers. Mol Cell Biol 19: 2435-2444 194 Schwartz DA, Dayton CS, et a1. (1994) Determinants of survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 149: 450-454 Selman M. (1990) Pulmonary fibrosis: human and experimental disease. Rojkind M Eds Connective tissue in health and disease, CRC Press Boca Raton, FL 123-188 Selman M, Pardo A, et al. (2001) American Thoracic Society; European Respiratory Society; American College of Chest Physicians. Idiopathic PF: prevailing and evolving hypotheses about its pathogenesis and implications for therapy. Ann Intern Med 134:136- 15] Shahar I, Topilsky M, et al. (1999) Effect of endothelin-1 on alpha-smooth muscle actin expression and on alveolar fibroblast proliferation in interstitial lung diseases Int J Immunopharmacol 21: 759-775 Sherman T and Brasier AR (2001) Role of signal transducers and activators of transcription 1 and 3 in inducible regulation of the human angiotensinogen gene by interleukin-6 Mol Endocrin 1 5:441 -457 Simon R. (1995) Alveolar epithelial cells in pulmonary fibrosis. In: Pulmonary F ibrosis, P. S., Thrall RS (eds). Marcel Dekker: New York, 511-540 Simon RH, Sitrin RG, et al. (1992) Fibrin degradation by rat pulmonary alveolar epithelial cells. Am J Physiol 262: L482-L488 Singh SN, Fisher SG, et al. (1997) Pulmonary effect of amiodarone in patients with heart failure. The Congestive Heart Failure-Survival Trial of Antiarrhythmic Therapy (CHF- STAT) Investigators (Veterans Affairs Cooperative Study No. 320). J Am Coll Cardiol 30(2): 514-517 Sitrin RG, Brubaker PG, et al. (1987) Tissue fibrin deposition during acute lung injury in rabbits and its relationship to local expression of procoagulant and fibrinolytic activities. Am Rev Respir Dis 135(4): 930-936 Specks U, Rohrbach MS, et al. (1990) Bronchoalveolar lavage fluid angiotensin- converting enzyme in interstitial lung diseases Am Rev Respir Dis 141: 117-123 Stern JB, Crestani B, et al. (2003) Keratinocyte growth factor and Hepatocyte growth factor: their roles in alveolar epithelial repair Rev Mal Respir 202896-903 Stewart JP, Ross AJ, et al. (1999) The detection of Epstein-Barr virus DNA in lung tissue from patients with idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 159:1336- 1341 195 Sugahara K, Iyama K, et al. (1998) Double intratracheal instillation of keratinocyte growth factor prevents bleomycin-induced lung fibrosis in rats. J Pathol 186:90-98 Sun Y and Weber TK. (1998) Cardiac remodelling by fibrous tissue: role of local factors and circulating hormones. Ann Med 1:83—88 Sutherland LM, Murray AW, et al. (2001) Alveolar type 11 cell apoptosis. Comp Biochem Physiol A Mol Integr Physiol 129: 267-285 Swaisgood CM, Noga C, et al. (2000) The development of bleomycin-induced pulmonary fibrosis in mice deficient for components of the fibrinolytic system. Am J Pathol 157: 177-187 Swiderski RE, Floerchinger CS, et al. (1998) Differential expression of extracellular matrix remodeling genes in a murine model of bleomycin-induced pulmonary fibrosis. Am JPathol 152: 821-828 Tanimoto K, Sugiyama F, et al. (1994) Angiotensinogen-deficient mice with hypotension. J Biol Chem 269(50): 31334-31337 Taniyama Y, Nakagarni H, et al. (2000) Potential contribution of a novel antifibrotic factor, hepatocyte growth factor, to prevention of myocardial fibrosis by angiotensin II blockade in cardiomyopathic hamsters. Circulation 102:246-252 Taylor BK, Stoops TD, et al. (2000) Protein phosphatase inhibitors arrest cell cycle and reduce branching morphogenesis in fetal rat lung cultures. Am J Physiol Lung Cell Mol Physiol 278: L1062-1070 Templin MV, Levin AA, et al. (2000) Pharmacokinetic and toxicity profile of a phosphorothioate oligonucleotide following inhalation delivery to lung in mice. Antisense Nucleic Acid Drug Dev, 10:359-368 Terzaghi M, Nettesheim P, et al. (1978) Repopulation of denuded tracheal grafts with normal, preneoplastic, and neoplastic epithelial cell populations. Cancer Res 38(12): 4546-4553 Thomas AQ, Phillips J 3rd LK, et a1. (2002) Heterozygosity for a surfactant protein C gene mutation associated with usual interstitial pneumonitis and cellular nonspecific interstitial pneumonitis in one kindred. Am J Respir Crit Care Med 165: 1322-1328 Thrall R, McCormick J, et al. (1979) Bleomycin-induced PF in the rat. Am J Pathol, 95: 117-130 Thrall RS and Scalise PJ. (1995). Bleomycin In: Phan SH and Thrall RS, ed. Pulmonary fibrosis. New York: Marcel Dekker, Inc., 231-264 196 Timmerrnans PB, Wong PC, et al. (1993). Angiotensin 11 receptors and angiotensin II receptor antagonists. Pharmacol Rev 45(2): 205-251 Uchiyama Y. (2001) Autophagic cell death and its execution by lysosomal cathepsins. Arch Histol Cytol. 64: 233-246 Umezawa H. (1967) Bleomycin Gan No Rinsho 13(10): 735 Umezawa H. (1973) Studies on bleomycin: chemistry and the biological action. Biomedicine 18(6): 459-475 Umezawa H. (1974) Chemistry and mechanism of action of bleomycin. Fed Proc 33(11): 2296-2302 Umezawa H, Maeda K, et al. (1966) New antibiotics, bleomycin A and B. J Antibiot (Tokyo) 19(5): 200-209 Umezawa H, Takeuchi T, et a1. (1972) Studies on the mechanism of antitumor effect of bleomycin of squamous cell carcinoma. J Antibiot (Tokyo) 25(7): 409-420 Uhal BD, Filippatos G, et al. (2003) Inhibition of Amiodarone-induced lung fibrosis but not alveolitis by angiotensin system antagonists Pharmacol Toxicol 92:81-87 Uhal BD, Filippatos G, et al. (1998) Captopril inhibits apoptosis in human lung epithelial cells: a potential antifibrotic mechanism. Am J Physiol 275:L1013-L1017 Uhal BD, Flowers KM, et al. (1991) Type II pneumocyte proliferation in vitro: problems and future directions. Am J Physiol Suppl, 261 :1 10-1 17 Uhal BD and Rannels DE. (1991) DNA distribution analysis of type II pneumocytes by laser flow cytometry: technical considerations. Am J Physiol 261:L296-L306 Uhal BD, Selman M, et al. (1998) AEC death adjacent to underlying myofibroblasts in advanced fibrotic human lung Am J Physiol 275: L1 192-1 199 Uhal BD, (1997) Cell cycle kinetics in the alveolar epithelium. Am J Physiol 272(6 Pt 1): L1031-1045 Uhal BD. (2001) Fas and apoptosis in the alveolar epithelium: holes in the dike? (Editorial) Am J Physiol 281:L326-L327 Uhal BD (2002) Apoptosis in lung fibrosis and repair Chest 122:293S-2988 Uhal BD. (2002) Apoptosis in lung fibrosis and repair Chest 122(6 Suppl): 293 S-298S. 197 Urata H, Husain A, et al. (1990) Identification of a highly specific chymase as the major angiotensin II-forming enzyme in human heart J Biol Chem 266: 22348-22357 Van Kats JP, Danser AH, et al. (1998) Angiotensin production by the heart: a quantitative study in pigs with the use of radiolabeled ANG Infusions. Circulation 98: 73-81 Van Kats JP, de Lannoy LM, et al. (1997) Angiotensin 11 type 1 (AT1) receptor-mediated accumulation of angiotensin II in tissues and its intracellular half-life in vivo. Hypertension 30(1 Pt 1): 42-49 Van Kats JP, Schalekamp MA, et al. (2001) Intrarenal angiotensin II: interstitial and cellular levels and site of production. Kidney Int 60(6): 2311-2317 Venkatesan N, Punithavathi V, et al. (1997) Curcumin protects bleomycin-induced lung injury in rats. Life Sci 61: 51-58 Ververeli K and Chipps B. (2004) Oral corticosteroid-sparing effects of inhaled corticosteroids in the treatment of persistent and acute asthma. Ann Allergy Asthma Immunol 92(5): 512-522 Wang R, Uhal BD, et al. (2000) Apoptosis of lung epithelial cells in response to TNF- alpha requires ANG II generation de novo. J Cell Physiol 185:253-259 Wang R, Uhal BD, et al. (2000) Abrogation of bleomycin-induced epithelial apoptosis and lung fibrosis by captopril or by a caspase inhibitor. Am J Physiol Lung Cell Mol Physiol 279: Ll43-151 Wang R, Uhal BD, et al. (1999) Human lung myofibroblast-derived inducers of alveolar epithelial apoptosis identified as angiotensin peptides. Am J Physiol 277: L1158-ll64 Wang R, Uhal BD, et al. (1999) Fas-induced apoptosis of AECS requires ANG II generation and receptor interaction. Am J Physiol 277: L1245-1250 Wang R, Uhal BD, et al. (1999) ANG II induces apoptosis in human and rat AECS. Am J Physiol 276: L885-889 Ward WF, Molteni A, et al. (1990) Captopril reduces collagen and mast cell accumulation in irradiated rat lung. Int J Radiat Oncol Biol Phys 19: 1405-1409 Weber KT, Campbell SE, et a1 (1995) Structural remodeling of the heart by fibrous tissue: role of circulating hormones and locally produced peptides. Eur Heart J 16(suppl N): 12-18 Wells A, DuBois R, et al. (1993) The predictive value of appearances on thin-section computed tomography in fibrosing alveolitis Am Rev Respir Dis 148: 1076-1082 198 White ES, Lazar MH, et a1. (2003) Pathogenetic mechanisms in usual interstitial pneumonia/idiopathic pulmonary fibrosis J Pathol 201(3): 343-354 Whyte M, Meliconi R, et al. (2000) Increased risk of fibrosing alveolitis associated with interleukin-1 receptor antagonist and tumor necrosis factor- gene polymorphisms. Am J Respir Crit Care Med 162:755-758 Wielbo D, Semia C, et al. (1995) Antisense inhibition of hypertension in the spontaneously hypertensive rat Hypertension 25(3): 314-9 Wielbo D, Simon A, et al. (1996) Inhibition of hypertension by peripheral administration of antisense oligodeoxynucleotides Hypertension 28(1): 147-151 Witschi H. (1990) Responses of the lung to toxic injury Environ Health Perspect 8525-13 Wright JR, Wager RB, et al. (1987) Surfactant apoprotein Mr = 26,000-36,000 enhances uptake of liposomes by type 11 cells J Biol Chem 262(6): 2888-2894 Woessner J. (1961) The determination of hydroxyproline in tissue and protein samples containing small proportions of this imino acid. Arch Biochem Biophys 93: 440-447 Wu G, El-Deiry W, et al. (1998) Potential role for Cathepsin D in p53-dependent tumor suppression and chemosensitivity. Oncogene 16:2177-2183 Yaekashiwa M, Ohnuma K, et al. (1997) Simultaneous or delayed administration of hepatocyte growth factor equally represses the fibrotic changes in murine lung injury induced by bleomycin. A morphologic study Am J Respir Crit Care Med 1562193 7-1944 Yagoda A, Mukherji B, et al. (1972) Bleomycin, an antitumor antibiotic. Clinical experience in 274 patients Ann Intern Med 77(6): 861-870 Yasuda Y, Yamarnoto K, et al. (1999) Characterization of new fluorogenic substrates for the rapid and sensitive assay of cathepsin E and cathepsin D. J Biochem 125:1137-1143 Yi ES, Ulich TR, et al. (1998) Keratinocyte growth factor decreases pulmonary edema, transforming growth factor-beta and platelet-derived growth factor-BB expression, and alveolar type II cell loss in bleomycin-induced lung injury. Inflammation'22z315-325 Yi ES, Ulich TR, et al. (1996) Keratinocyte growth factor ameliorates radiation- and bleomycin-induced lung injury and mortality. Am J Pathol 149:1963-1970 Yonemaru M, Kusumoto H, et al. (1997) Elevation of antibodies to cytomegalovirus and other herpes viruses in pulmonary fibrosis Eur Respir J 10:2040-2045 199 Yoshida K, Hara N, et al. (2002) MAP kinase activation and apoptosis in lung tissues from patients with idiopathic pulmonary fibrosis J Pathol 1982388-396 Yoshiji H, Kuriyama S, et al. (2001) Angiotensin-II type I receptor interaction is a major regulator for liver fibrosis development in rats. Hepatology 342745—750 Young L and Adamson I. (1993) Epithelial-fibroblast interactions in bleomycin-induced lung injury and repair Environ Health Perspect 101: 56-61 Zou LX, Hymel A, et al. (1996) Renal accumulation of circulating angiotensin II in angiotensin II-infused rats Hypertension 27(3 Pt 2): 658-662 Zou LX, Imig JD, et al. (1996) Receptor-mediated intrarenal angiotensin II augmentation in angiotensin II-infused rats Hypertension 28(4): 669-677 Zisman DA, Martinez FJ, et al. (2000) Cyclophospharnide in the treatment of idiopathic pulmonary fibrosis: a prospective study in patients who failed to respond to corticosteroids. Chest 1 17: 1619-1626 200 11131111111111111111111111111