2.5 .1 i . ix .5: I do...“ I. ‘ v 3 ul . as; 2. H... t:\L\v I“ I 31\>\3...vllkl I A." l I. G :9: in I... .81 . . .. . v . . 1.’ i: r $ J‘ 1; 3 | ‘£ 99.2“ § z i? : *3? 6‘33?! E? ' a 1-93... 1.1.! _ .128. 1 A it: 1h‘lp’43 [33.1.2 til. «i ‘ Michigan State University This is to certify that the dissertation entitled THE ROLE OF HYPOXIA INDUCIBLE FACTORS IN LUNG DEVELOPMENT AND COBALT-INDUCED LUNG INJURY presented by YOGESH SAINI has been accepted towards fulfillment of the requirements for the PhD. degree in Genetics (ll (lead U M o Professor’s Signature $183200? Date MSU is an Alfinnative Action/Equal Opportunity Employer PLACE IN RETURN BOX to remove this checkout from your record. To AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 5/08 K'IProj/Acc8PrelelRCIDateDue indd THE ROLE OF HYPOXIA INDUCIBLE FACTORS IN LUNG DEVELOPMENT AND COBALT-INDUCED LUNG INJURY By Yogesh Saini A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Genetics 2009 ABSTRACT THE ROLE OF HYPOXIA INDUCIBLE FACTORS IN LUNG DEVELOPMENT AND COBALT-INDUCED LUNG INJURY By Yogesh Saini The increasing use of disposable electronics and growing industrialization of nation’s economies have drastically raised the risk of exposure to toxic metals. One of these metals, cobalt, is used widely in several industries involved in the production of coloring agent for ceramics, hard metal alloys, sintered carbides, drilling and grinding tools. Cobalt (or hard metal) asthma caused by the inhalation of cobalt containing dust is characterized by ainNay constriction, alveolitis, fibrosis and associated giant cell interstitial pneumonitis. The characterization of cobalt-induced toxicities demands comprehensive understanding of the effects of toxicants on cell signaling and the downstream changes in gene expression. It is well established that cobalt is a hypoxia mimic due to its ability to induce hypoxia-like gene expression responses. To enhance our understanding of cobalt-induced toxicity and determine the role of its ability to mimic hypoxia signaling in the process, we generated a doxycycline inducible lung-specific Hypoxia inducible factor 1a (HIF1ot) deficient system in mice. In utero, lung specific deletion of H|F1o resulted in altered surfactant metabolism and defects in alveolar epithelial differentiation that led to premature death due to respiratory distress. In contrast, the in utero deletion of HIF2a (another prominent form of HlFoc) from the lungs did not affect the viability of neonates. Interestingly, the removal of both H|F1a and HlF2a from the lungs during development led to the birth of phenotypically normal pups, suggesting that the loss of HIFZa can rescue the lethal phenotype associated with the loss of H|F1a from the lungs. Microarray analysis of the lungs from HlF1aA/A, HlF2 OLA/A and HlF1/2 ozA/A identified sets of genes, involved in various cellular pathways such as surfactant metabolism and vesicular trafficking that were specifically affected in the HlF10tA/A neonates and these results suggest possible mechanistic information for the role of HlFs in lung development. In order to elucidate the role of epithelial derived-HlF1a signaling in cobalt-induced lung injury, we deleted the transcription factor postnatally. These mice were exposed to cobalt chloride via oropharyngeal aspiration. Compared to control mice, mice that were H|F1oc deficient in their lungs exhibited ainNay infiltration of eosinophils associated with airway epithelial changes, including mucus cell metaplasia and increased levels of the chitinase-like proteins YM1 and YM2. These results suggested that airway epithelial-derived HlF1a plays a critical role in modulating the inflammatory response of the lung. Moreover, its disruption leads to a tissue that is biased towards a Th2-like response and exhibits an asthma-like phenotype following cobalt challenge. Taken together, the striking differences observed following cobalt exposure in the two mice suggests that they will be a powerful tool to understand the relationship between allergy-induced asthma, hypoxia, and inflammation. ACKNOWLEDGMENTS I would like to take this opportunity to express my profound gratitude to my advisor Dr. John J. LaPres who gave me the opportunity to work in his lab under his quality mentorship. I want to thank him for his sincere guidance, generous time, patience, encouragement and continuous support throughout my research work. His precious scientific advice and great attitude towards science compelled me to adopt those elite qualities in my developing scientific career which I will take with me through my entire career. I feel extremely fortunate to have Dr. LaPres as my major advisor. I greatly appreciate my guidance committee members; Dr. Jack R Harkema, Dr. A. Daniel Jones, Dr. Robert A Roth, and Dr. Susan L Ewart for their great help, insightful comments and constructive discussions. I am indebted to them for their valuable suggestions and continued inputs to improve my research progress as well as my scientific career. I am thankful to the Genetics Program and Dr. Barbara Sears (Director, Genetics Program) for accepting me in to the program which gave me great opportunity to enhance my scientific career. I sincerely thank Jeannine Lee for all her help from the day I submitted my application for admission to the program. I would also like to thank Environmental and Integrative Toxicology Program executive committee for providing me opportunity to gain expertise in Toxicology as my second PhD major. I am obliged to Kyung Y. Kim, Christian Merrill and Krista Greenwood for their help in my research work. I would like to thank all the lab members from Dr. Harkema's laboratory, especially Lori Bramble, Ryan Lewandowski, Dr. Daher l Aibo, Dr. Neil Birmingham, Dr. James Wagner and Dr. Daven Jackson for their endless help during my research work. My sincere thanks are extended to all current and former colleagues in Dr. LaPres's laboratory, especially Dr. Ajith Vengellur, Dr. Scott G Lynn, Dorothy M Tappenden, Heyjin Hwang, Kyunghee Burkitt, Steven Proper, Amy Bays and Dr. Kang Ae Lee for all their kind help, cooperation, and friendly atmosphere. I also want to thank all my friends and colleagues in Genetics Program, Integrative Toxicology Program and Biochemistry and Molecular Biology Department. I also want to thank my friends Vishal, Shipra, Ram and Anita for their kind help and providing me a very friendly atmosphere. I greatly appreciate ULAR and Human Histopathology Division for their help in my research work. I thank with all my heart to my parents for their encouragements, love and support which motivated me to accomplish all my educational aims. Finally, my special thanks go to my dear wife, Sonika Patial who has been a source of motivation during all those tough moments. lt is a very special year of my life as we were blessed with our baby boy, Krish on 15th November. TABLE OF CONTENTS LIST OF TABLES ............................................................................... ix LIST OF FIGURES ............................................................................... x KEY TO ABBREVIATIONS .................................................................. xii CHAPTER 1: INTRODUCTION ................................................................................. 1 1. Oxygen ..................................................................... 1 2. Anatomy and Development of the Respiratory System ........ 2 2.A. Development of lung ............................................. 2 2.8. Anatomy of the respiratory system ........................... 6 3. Mammalian gas exchange ........................................... 1O 4. Hypoxia ................................................................... 12 4.A. Biological Significance .......................................... 12 4.3. Cellular responses to hypoxia ................................ 13 4.0. Hypoxia-responsive transcription factors .................. 18 4D. HIF signaling and regulation of gene expression........25 4.E. HlF signaling and development...............................26 4.E.1. Placentation and Hypoxia signaling............26 4.E.2. Hypoxic regulation of organogenesis .......... 30 4.E.2.a. Vascular System ............................. 31 4.E.2.b. Cardiac System .............................. 31 4.E.2.c. Lung ............................................. 33 5. Hypoxia and its effects on the lung ................................ 35 6. Hypoxia Mimics and HIF signaling .................................. 36 7. Metals ...................................................................... 37 7A. History of metals .......................................... 37 7.8. Biological significance of metals ...................... 37 7.0. Toxicity of Metals ......................................... 38 8. Cobalt ...................................................................... 41 BA. Occurrence and uses.... ............................... 41 8.8. Cobalt metabolism and Exposure ................... 42 vi 8.C. Cobalt toxicity .............................................. 43 8.0. The role of HlFs in cobalt-mediated toxicity ....... 44 Hypothesis and Specific Aims ................................................ 45 Importance ......................................................................... 47 References for Introduction ................................................... 48 CHAPTER 2: HIF1a is essential for normal intrauterine differentiation of alveolar epith- -eIIum and surfactant production in the newborn lung of mice .............. 60 Abstract .............................................................................. 61 Introduction ......................................................................... 62 Material and Methods ............................................................ 64 Results ............................................................................... 69 Discussion .......................................................................... 95 References .......................................................................... 99 CHAPTER 3: Loss of HIFZot rescues the HIF1a deletion phenotype of neonatal respira- tory distress in mice ....................................................................... 103 Abstract ............................................................................. 104 Introduction ........................................................................ 1 06 Material and Methods ........................................................... 110 Results .............................................................................. 1 19 Discussion ......................................................................... 145 References ......................................................................... 1 51 CHAPTER 4: The role of Hypoxia Inducible Factor 1a in modulating Cobalt-Induced Lung Inflammation: A Subchronic Study ........................................... 155 Abstract ............................................................................. 1 56 Introduction ......................................................................... 1 57 Material and Methods ........................................................... 159 Results .............................................................................. 165 Discussion ......................................................................... 192 References ......................................................................... 200 vii CHAPTER 5: The Role of Hypoxia Inducible Factor 10: (HlF1ot) in Modulating Cobalt - Induced Lung Inflammation: An Acute Study ..................................... 203 Abstract ............................................................................. 204 Introduction ........................................................................ 206 Material and Methods ........................................................... 209 Results .............................................................................. 21 7 Discussion ......................................................................... 234 References ......................................................................... 240 CHAPTER 6: Conclusions .................................................................................. 244 APPENDIX TABLE A.1. List of differentially expressed genes ................... 251 viii Table 2.1 Table 2.2 Table 3.1 Table 3.2 Table 3.3 Table 3.4 Table 4.1 Table 4.2 Table 4.2 Table A.1 LIST OF TABLES Primers for PCR-genotyping ............................................. 94 Primers for real-time RT-PCR ........................................... 94 Primers for real-time RT-PCR .......................................... 120 Cellular pathways affected by deletions of HlF1a, HIF20 and HIF1/2c1 ....................................................... 136 Biological processes affected by deletions of HlF1a, HIF20 and HIF1/2r1 ....................................................... 137 Genes involved in surfactant metabolism ........................... 139 List of genes analyzed qRT-PCR ..................................... 172 Gene Expression Changes ............................................. 191 Cytokine Profiles ......................................................... 193 List of differentially expressed genes ............................... 251 LIST OF FIGURES Images in this dissertation are presented in color Figure 1.1. Figure 1.2. Figure 1.3. Figure 2.1. Figure 2.2. Figure 2.3. Figure 2.4. Figure 2.5. Figure 2.6. Figure 2.7. Figure 3.1. Figure 3.2. Figure 3.3. Figure 3.4. Figure 3.5. Figure 3.6. Figure 3.7. Figure 3.8. Domain structure of human HIFs ....................................... 16 Structure of human HlF1a protein and oxygen-dependent modification of HIF1a ............................ 19 Hypoxia Signaling in normoxic and hypoxic conditions ........... 22 Lung specific, doxycycline-inducible deletion of HIF1a in triple transgenic mrce70 Effects of lung specific deletion of HlF1or ............................ 76 Pulmonary Histopathology of Lungs ................................... 79 Electron photomicrographs of alveolar epithelium of newborn pups .............................................. 81 Surfactant protein expression ............................................ 85 Expression of developmentally important genes ................... 89 Immunohistochemistry for Cre recombinase, HIF1ot, and HIF292 Lung specific, doxycycline-inducible deletion of HIF1a and HlF2o in triple transgenic mice .................................. 112 Genotyping of transgenes .............................................. 114 Survivability Plot .......................................................... 121 Pulmonary Histopathology .............................................. 123 Immunohistochemistry for HlF1o ...................................... 126 Immunohistochemistry for HIF20 ...................................... 128 Periodic acid Schiff (PAS) staining ................................... 130 Differential gene expression between HIF1oA/A, HlF2ch/A and HIF1/20 A/A ............................................. 133 Figure 3.9. Figure 4.1. Figure 4.2. Figure 4.3. Figure 4.4. Figure 4.5. Figure 4.6. Figure 4.7. Figure 4.8. Figure 5.1. Figure 5.2. Figure 5.3. Figure 5.4. Figure 5.5. Figure 5.6. Figure 5.7. QRTPCR verification of selected microarray gene expression responses ............................................ 140 HIF10t immunohistochemistry of lungs from control and doxycycline treated mice ................................ 166 Weight change and cell counts from cobalt challenged control and HIF1oc A/A mice ........................... 169 Cell counts from genotype and treatment controls ............... 174 Histopathology and picro-sirius staining control and cobalt-treated control mice ....................................... 178 Major Basic Protein Staining in lungs from control and HlF1aA/A mice ............................................. 181 Alcian Blue/Periodic Acid Schiff Stain and YM1/2 IHC .......... 184 H&E staining and YM1/2 IHC of cobalt treated control and HIF1orA/A mice .................................. 186 Gene expression results ............................................... 189 Experimental Design ..................................................... 210 BALF proteins and total cell counts from control andH|F1a A/A mice ............................................. 215 Effect of cobalt treatment on inflammatory cells Recovered in bronchoalveolar lavage fluid ........................ 218 Histopathology staining of control and cobalt-treated control mice ................................................................. 222 Major Basic Protein (MBP) immunohistochemistry in lungs from control and HlFlaA/A mice .............................. 225 Neutrophil (PMN) immunohistochemistry in lungs from control and HlF1aA/A mice ............................................. 227 Cytokine levels in BALF from cobalt-treated control and HIF 10L A/A mice ............................................................ 230 xi ANOVA ARNT ATP C/EBP CBP CCSP COPD CRE CREB DFO DMOG DPPC DTT EDTA EMT EPO ETC Foxa2 H&E KEY TO ABBREVIATIONS Analysis of Variance Aryl hydrocarbon receptor nuclear translocator Adenosine triphosphate CAAT/enhancer-binging protein CREB binding protein Clara cell secretory protein Chronic obstructive pulmonary disease Cyclization recombination cAMP response element binding Desferrioxamine Dimethyloxaloglutarate Dipalmitoylphosphatidylcholine Dithiothreitol Ethylenediaminetetraacetic acid Epithelial-mesenchymal transition Erythropoietin Electron transport chain Forkhead box A2 Hematoxylin and eosin xii HAPE HGPRT HIF HNF HO-1 HRE Lb LoxP MEF MTF-1 mv NFKB ODD PAS PAS PFK—1 PHD PMSF High-altitude pulmonary edema hypoxanthine guanine phosphoribosyl transferase Hypoxia inducible factor Hepatocyte nuclear nactor Haemoxygenase-1 Hypoxia responsive element lmmunoglobulin isotype E lmmunoglobulin isotype G Interleukin lamellar bodies Locus of crossover in P1 Mouse embryonic fibroblast Metal transcription factor-1 Microvilli Nuclear factor-kappa B Oxygen-dependent degradation domain Per/Arnt/Sim Periodic acid Schiff Phosphofructokinase-1 Prolyl hydroxylase domain protein Phenylmethylsulphonyl fluoride xiii PN p02 pVHL RDS RIPA ROS RT-PCR rtTA SDS-PAGE SEM SP-C TAD TEM TF TGF-B1 TNF-a VEGF pm Postnatal Partial pressure of oxygen von HippeI-Lindau protein Respiratory distress syndrome Radioimmunoprecipitation assay Reactive oxygen species Reverse transcriptase-polymerase chain reaction Reverse tetracycline transactivator Sodium dodecyl sulfate polyacrylamide gel electrophoresis Standard error of mean Surfactant protein C Transactivation domain Transmission electron microscopy Transferrin Transforming growth factor-beta 1 Tumor necrosis factor-alpha Vascular endothelial growth factor Micrometer xiv Introduction 1. Oxygen Oxygen is the third most abundant element, after hydrogen and helium, in the universe and makes up nearly 21% of the earth's atmosphere. Oxygen is the most important factor for the viability of living systems, except for obligate anaerobes. Oxygen acts as the ultimate electron acceptor in aerobic respiration and therefore, is critical for energy production in the form of adenosine triphosphate (ATP). Oxygen also acts as a substrate in various biosynthetic reactions as well as breakdown product in catabolic reactions [1]. The indispensable involvement of oxygen in energy generation and other critical cellular processes made it important to evolve sensing mechanisms for changes in oxygen availability. Evolutionary adaptation has selected various pathways to deal with oxygen tension fluctuations. These pathways regulate short- and long-term responses. Every organism, either unicellular or multicellular, possesses the ability to sense changes in the ambient oxygen and respond through alterations in metabolism and gene expression [2]. For strict aerobic unicellular organisms or cells in culture, oxygen comes from the atmosphere, either directly or dissolved in the surrounding medium. In multicellular organisms, however, oxygen is supplied by tissue bathing fluids containing dissolved or protein-bound oxygen delivered by specialized respiratory organs or tissue systems to allow gas exchange. These respiratory systems consist of conducting zones leading to the gas exchange area where oxygen diffuses into the tissue bathing fluid (blood in mammals). Different organisms have evolved varying anatomical structures to obtain and circulate oxygen, including very simple structures (e.g. gills and trachea) to complex specialized structures (e.g. lungs). The mammalian respiratory system represents one of the most complex respiratory structures. 2. Anatomy and Development of the Respiratory System 2.A. Development of the lung Human lung development initiates at third week of gestation (corresponds to E9.5 of mouse gestation) and proceeds through three chronological periods: embryonic, fetal period proper and postnatal periods. The fetal period proper is further divided in to three stages called pseudoglandular, canalicular and saccular. Since the process of differentiation proceeds from the center to the periphery of the lung asynchronously between lobes, there is significant overlap between these stages [3]. In humans, the embryonic period corresponds to 4-7 weeks after fertilization and represents the beginning of organ development. The lung appears, towards the end of the fourth week, as a bud ventral to the prospective esophagus. As the Iaryngotracheal groove deepens, lung bud grows in to the surrounding mesenchymal matrix by successive dichotomous branching. By the end of the seventh week, the lobar, segmental and subsegmental portion of ainNay tree with high columnar epithelium is already formed [3]. Starting from the eighth week, the lung resembles a small tubuloacinar (epithelial) gland, hence, the stage is called as a pseudoglandular stage which runs from fifth week to seventeenth week (corresponds to E9.5 to E16.5 of mouse gestation). It is in this stage that continuous growth and branching of the peripheral portion of epithelial tube leads to the formation of prospective conductive ainNays and appearance of acinar outline. Epithelial-mesenchymal interactions regulate this growth and branching pattern. Removal of mesenchyme from the tip of a sprouting tube prevents further branching whereas plantation of mesenchyme alongside of a lower order tube results in the appearance of new branch [4, 5]. Moreover, the presence of collagen and mesenchyme is needed for branching and epithelial differentiation [6, 7]. By the twelfth week, mucous glands are present as solid sprouts from the epithelial layer and acquire canal and secretory activities by the fourteenth week [8]. At the end of seventeenth week, acini are composed of stem tubule (prospective terminal bronchiole), 2-4 future respiratory bronchioles and small clusters of short tubules and buds. The canalicular stage of lung development spans from week 17 to week 26 in humans (corresponds to E16.5 to E17.5 of mouse gestation). At this stage compact acinar clusters grow by three processes: peripheral branching, elongation of each tubular branch, and widening of the distal airspaces. Thus, the tubules until about three orders of branching are referred to as canaliculi. It is at this stage that capillarization starts taking place around the airspaces to establish close contact with the overlaying cuboidal epithelium lined with Type II cells. These contact points are the sites where the cuboidal epithelial cells decrease in height and transform into Type I cells. These type I cells have attenuated cytoplasmic processes and form the future air-blood barrier. The two processes, close apposition of capillary epithelium to the epithelial cells and the epithelial flattening are intimately related, but it is unclear which of the two processes induces the other. The air blood barrier transformation starts peripherally as the cuboidal (undifferentiated) epithelial cells (Type II cells) towards the center are needed for further growth and branching [3]. It has been shown that Type II cells can incorporate tritiated thymidine (cell division) and lose their granules to differentiate eventually into Type I cells [9]. A similar pattern of transformation has also been observed in damaged alveolar epithelium suggesting the stem cell role of Type II cells [10]. Similarly, it has been shown that the Clara cells, secretory cell type of the bronchioles, represents the progenitor cells of the bronchiolar epithelium [11]. Saccular stage of lung development represents period from week 24 to week 36 in humans (corresponds to E17.5 to PN6 of mouse gestation). In the terminal phase of the canalicular stage, the ainlvays consist of clusters of relatively thin-walled terminal saccules and is referred to as saccular or terminal sac. These saccules give rise to the last generation of ainrvays, some prospective alveolar ducts, and the alveolar sacs at the outermost periphery. Thus each canalicule, except the last branch, goes through the stages of being a saccule, then a smooth-walled channel, and after birth, when alveoli have formed, a typical alveolar duct. The vast elaboration of the air space coupled with marked decrease in interstitial tissue has an important consequence on the alveolar capillary arrangement. The capillaries network form a mesh round the ainNay channels and within the intersaccular septa as the airspaces approach each other. It is in the saccular stage that the pulmonary surfactant system matures (between week 29 and week 32 in humans). At birth, the lung parenchyma structure is still in an immature state despite the complete branching pattern and number of airway generations. Neonatal human lung contains only 20 million alveoli, which represents approximately 10 % of the full population of 300 million observed in adult lung [12]. At birth, the rat lung consists of smoothly contoured ducts and saccules having thick primary septa and relatively thick layer of connective tissues. It takes two postnatal weeks to transform these immature structures into alveolar ducts and alveoli lined with thin inter-alveolar septa [13, 14]. 2.3. Anatomy of the Respiratory System The mammalian respiratory system is composed of two major components: the conducting portion and the respiratory portion. The conducting portion, situated both outside and within the lungs, conveys air from the external milieu to the lungs whereas the respiratory portion, located strictly within the lungs, functions in the actual exchange of oxygen for carbon dioxide (external respiration). The conducting portion of the respiratory system is composed of the nasopharynx, trachea, primary bronchi, secondary bronchi (lobar bronchi), tertiary bronchi (segmental bronchi), bronchioles, and terminal bronchioles. The bronchial tree, stemmed from trachea, divides 15 to 20 times before reaching the level of the terminal bronchioles. The left and right lobes have 2 and 3 secondary bronchi (branches of the primary bronchi), also known as lobar bronchi, respectively. As these secondary bronchi enter the lobes of the lung, they subdivide into smaller branches, tertiary (segmental) bronchi. Each tertiary bronchus arborizes and leads to the formation of bronchioles. Each bronchiole supplies air to a pulmonary lobule. It is generally accepted that the bronchioles represent the 10th to 15th generation of dichotomous branching of the bronchial tree. Their diameter can vary among authors from 0.3mm to 5mm. The epithelial lining of larger bronchioles is primarily ciliated simple columnar with occasional goblet cells whereas the smaller bronchioles have simple cuboidal (many with cilia) with occasional Clara cells and no goblet cells. Clara cells are columnar cells with dome-shaped apices that have short, blunt microvilli. Clara cell cytoplasm houses numerous secretory granules containing glycoproteins that protect bronchiolar epithelium. Additionally, these cells degrade toxins in the inhaled air via cytochrome P-450 enzymes in their smooth endoplasmic reticulum [15]. Some investigators suggest that Clara cells produce a surfactant-like material that reduces the surface tension of bronchioles and facilitates the maintenance of their patency [16]. It has been shown that Clara cells divide to regenerate the bronchiolar epithelium [17-20]. Thus, Clara cells forms a very important cell population lining the bronchiolar epithelium. The respiratory portion of the respiratory system is composed of respiratory bronchioles, alveolar ducts, alveolar sacs, and alveoli. Terminal bronchiole branch to give rise to respiratory bronchioles and forms the first region of the respiratory system where exchange of gases can occur. Respiratory bronchioles are similar in structure to terminal bronchioles except that their walls are interrupted by the presence of thin-walled, pouch-like structures known as alveoli, where efficient gaseous exchange occurs. As respiratory bronchioles branch, they become narrower in diameter and their population of alveoli increases. Subsequent to several branchings, each respiratory bronchiole terminates in an alveolar duct. Alveolar ducts do not have walls of their own; they are merely linear arrangements of alveoli. An alveolar duct that arises from a respiratory bronchiole branches, and each of the resultant alveolar ducts, usually ends as a blind outpouching composed of two or more small clusters of alveoli, known as an alveolar sac. Each alveolus is a small outpouching, about 200pm in diameter, of respiratory bronchioles, alveolar ducts, and an alveolar sac that is composed of highly attenuated type I pneumocytes (also known as type | alveolar cells or squamous alveolar cells) and larger type II pneumocytes (also known as great alveolar cells, septal cells, and type II alveolar cells). Approximately 95% of the alveolar surface is lined by type I pneumocytes which are highly flattened with cytoplasm thickness of approximately 80nm. The type II pneumocytes are more numerous than type I pneumocytes, however, they occupy only about 5% of the alveolar surface. The most distinguishing feature of these cells is the presence of membrane-bound lamellar bodies that contain pulmonary surfactant, the secretory product of these cells. Pulmonary surfactants, heterogenous phospholipoproteinaceous material consisting of 90% lipids and 10% proteins, that are synthesized on the rough endoplasmic reticulum (RER) of type II pneumocytes. The surfactants are composed primarily of two phospholipids (dipalmitoylphosphatidylcholine (DPPC) and phosphatidylglycerol) a neutral lipid, and four unique proteins (surfactant apoproteins SP-A, SP-B, SP-C, and SP-D). The surfactants are modified in the golgi apparatus and are then released from the trans golgi network into secretory vesicles, known as composite bodies, the immediate precursors of lamellar bodies. The lamellar body-bound surfactants are released via exocytosis into the lumen alveolar lumen where it forms a lattice-like network known as tubular myelin. The surfactants decrease surface tension, thus preventing atelectasis, namely the collapse of the alveolus. The surfactants are continuously manufactured by type II pneumocytes and are phagocytosed and recycled by type II pneumocytes and, less frequently, by alveolar macrophages. In addition to producing and phagocytosing surfactants, type II pneumocytes also act as precursor of type I pneumocytes. Alveolar macrophages, also known as Dust Cells, phagocytize particulate matter in the lumen of the alveolus as well as in the interalveolar spaces. The circulating blood monocytes, after infiltrating into the pulmonary interstitium, become alveolar macrophages and migrate between Type I pneumocytes into the alveolar spaces. These cells phagocytose particulate matter, such as dust and bacteria, and thus maintain a sterile environment within the lungs. Alveolar macrophages also assist Type II pneumocytes in the recycling of surfactants. Approximately 100 million macrophages migrate to the bronchi each day and are transported from there by ciliary action to the pharynx to be eliminated by being swallowed or expectorated. Some alveolar macrophages, however, reenter the pulmonary interstitium and migrate into lymph vessels to exit the lungs. 3. Mammalian Gas exchange Mammals have one of the most advanced respiratory systems. In animal physiology, respiration is defined as the transport of oxygen from the outside air to the cells within tissues, and the simultaneous transport of carbon dioxide in the opposite direction. This is different from cellular respiration, which is a biochemical oxidation pathway by which cells release energy from the chemical bonds of food molecules (e.g. glucose) and provide that energy for the essential processes of life. In humans and other mammals, physiological respiration involves four steps: Ventilation, the process of moving ambient air in to and out of alveolar space. Pulmonary gas exchange or external respiration, the process of gaseous exchange between the alveoli and the pulmonary capillaries. Gas transport, movement of gases throughout the circulatory system to deliver oxygen to peripheral tissues and to transport carbon dioxide back to the pulmonary gas exchange site. Peripheral gas exchange, the transfer of gases between the tissue capillaries and the tissues/cells. The respiratory and cardiovascular systems work in concert to carry gases to and from the tissues. Veins carry deoxygenated blood from 10 peripheral tissues to the right atrium of the heart that transfers the blood to the right ventricle. The right ventricle pumps blood through the pulmonary artery towards the lungs. The pulmonary artery branches out to form capillary beds around the alveoli in close apposition to Type | pneumocytes to form 200-600 nm thick blood-gas barrier (alveolar-capillary barrier) [21]. Blood oxygenated at the blood-gas barrier is carried via the pulmonary vein to the left heart from where oxygenated blood (p02: 100mmHg) is pumped into the aorta and eventually to the whole body. The p02 in capillaries varies among different tissues and within the tissues depending upon the location and physiological state. For example, within the liver Iobules, the p02 varies depending on the proximity to the blood carrying vessels. The p02 of hepatic artery is 95-105 mmHg whereas the p02 of vena cava is approximately 40-45 mmHg [22]. At normal oxygen concentration, oxygen levels within the body vary between tissue and cell types. For instance, oxygen concentration required for normal brain function is uniformly higher than that for the muscles. Thus, a range of p02 is found in different tissues makes some tissues or organs more prone to oxygen deficit, a condition known as hypoxia. 11 4. Hypoxia 4.A. Biological Significance The normal p02 state is known as norrnoxia and when the p02 in the cell or tissue drops below this normal level, a state of hypoxia is said to exist. In the systemic context, hypoxia is categorized into four subcategories: hypoxic, anemic and circulatory hypoxia. Hypoxic hypoxia arises due to decrease in ambient oxygen partial pressure attributed to hypoventilation or altitude. Anemic hypoxia results from decreased hemoglobin concentration that leads to declined oxygen-carrying capacity of the blood. Circulatory hypoxia arises because of impaired tissue perfusion [23]. In systemic or organ system context, the hypoxic conditions arise in normal physiological process as well as in pathological conditions. It is well known that mammalian embryos develop in a state of partial hypoxia (p02 = 20—30 mmHg) and hypoxic conditions within the embryo have been characterized in the neural tubes, heart, and intersomitic mesenchyme at an early stage of organogenesis [24]. This decreased oxygen availability is an important regulator of proper angiogenesis and tubulogenesis [25]. Studies have shown that fluctuation in oxygen tension within the uteroplacental environment is an important determinant of cellular events during trophoblastic invasion and the placental remodeling process [26]. In the pathological settings, hypoxia is a critical feature in several diseases, including cancer, 12 myocardial ischemia, cerebral ischemia, atherosclerosis, rheumatoid arthritis, and chronic obstructive pulmonary disease (COPD). The lung’s integral role in oxygen uptake and supply does not preclude it from being negatively impacted by hypoxia. Conditions like ainNay obstruction, intra-alveolar exudates, septal thickening by edema, inflammation, fibrosis, or damage to alveolar capillaries can induce localized hypoxic conditions within the lung. In airway inflammatory diseases like asthma, lung structure changes due to persistent inflammation pose obstruction to air flow, which leads to a hypoxic condition in the affected pulmonary tissue [27]. There is a correlation between allergic airway inflammatory diseases and the upregulation of hypoxia sensitive proteins such as glycolytic enzymes, prolyI-4- hydroxylase, peroxiredoxin 1, and arginase [28]. In the current discussion on hypoxia, it is clear that hypoxia and hypoxia induced downstream processes have both beneficial as well as detrimental roles in the biological system. 4.8. Cellular Responses to hypoxia 6 An active cell might use as much as 2 x 10 molecules of ATP per second. Many fundamental cellular processes, such as transcription, translation, metabolic reactions, cytoskeletal movements, and ion transport require ATP as a primary source of energy. Maintenance of a cellular homeostatic environment using ATP-dependent ion pumping systems such as + + the Na IK ATPase requires 20 to 80% of the cellular metabolic energy. 13 Reduced ATP generation results in the loss of the cell’s ability to maintain ionic and osmotic balance. Failure of these ion-motive ATPase leads to 2+ membrane depolarization, uncontrolled Ca influx through voltage-gated 2+ Ca channels, and subsequent activation of calcium-dependent phospholipases and proteases. These events result in uncontrolled cell swelling, hydrolysis of biomolecules, leakage of cellular compartments and eventually, to cell necrosis [29]. Since oxygen is the primary requirement in efficient ATP generation in mitochondria, hypoxia exhibit its affect through a compromised electron transport chain and reduced ATP generation. Cells have developed selective bypass processes responsible for alternate but efficient energy conservation or generation. These adaptive and pro-survival processes include anaerobic glycolysis and reallocation of cellular energy between essential and nonessential ATP demand processes. Under hypoxia, cells switch to anaerobic glycolysis, a less efficient ATP generating process to meet their energy demands. This switch is turned on through allosteric regulation of phosphofructokinase-1 (PFK-1) activity and the overexpression of enzymes involved in anaerobic glycolysis [30]. PFK-1 catalyzes the conversion of fnJctose 6-phosphate to fructose 1,6-bisphosphate and is considered a major regulatory enzyme that controls carbon flux through glycolysis. PFK-1 is allosterically regulated by a number of metabolites. ADP, AMP and fructose-2,6-biphosphate are allosteric activators, whereas ATP and 14 citrate are the allosteric inhibitors of PFK—1. Under energy deficit (e.g. hypoxic conditions), the cellular AMP/ATP ratio is elevated which results in the activation of PFK-1 via an AMP activated protein kinase (AMPK) dependent process. AMPK activation has also been reported to recruit Glut-4 (glucose transporter) to the plasma membrane and to increase the expression of hexokinase as well as some mitochondrial enzymes involved in TCA cycle and electron transport chain (ETC) [31]. Another strategy adopted by cells to survive or adapt under hypoxic stress is reallocation of cellular energy between essential and nonessential ATP demand processes. Protein synthesis and ion motive ATPases account for more than 66% and 90% of ATP consumption in rat thymocytes and rat skeletal muscles, respectively [32]. The ATP consumption hierarchy is + + rearranged in energy deficit circumstances whereby Na /K pumping and + Ca2 cycling processes become the higher ATP consumers instead of replication, transcription, or translation [33]. Most of the adaptive hypoxic responses take place with in seconds through the activation or inhibition of already synthesized structural or regulatory proteins. However, some adaptive responses take place or are fine-tuned at the level of gene transcription or translation. As discussed in the 15 Figure 1.1. Domain structure of human HlFs. HIF-oc(HlF-1a, HIF-2a, HIF-3a), HIF-1B (ARNT), and ARNT2 belong to Per-ARNT-Sim (PAS) protein superfamily. HIF-oz consists of four structural domains: basic Helix-Loop-Helix (bHLH) domain (blue), Per- ARNT-Sim (PAS) domain (green), oxygen-dependent degradation domain (ODD) (purple), and transactivation (TAD) domain (yellow). HIF-1B and ARNT2 lacks ODD but shares all the other three domains present in HIF-or. The presence of the ODD that makes HIF-on oxygen- labile. ”Images in this dissertation are presented in color." 16 mowz coma Nmmx Neva 17 next section, hypoxia-responsive transcription factors function at the hub of oxygen sensing machinery. 4.C. Hypoxia-responsive transcription factors The oxygen sensing system consists of several hypoxia—responsive transcription factors that mediate regulation of genes responsible for maintaining or rescuing tissue perfusion and ATP generation, including vascular endothelial growth factor (VEGF), glycolyic enzymes, and glucose transporters. Several transcription factors are also influenced by hypoxia, including nuclear factor-KB (NFKB), cAMP response element binding (CREB), activator protein 1 (AP-1), p53, Stable Protein 1 (SP-1), SP-3, Early growth response factor 1 (EGR-1), CCAAT/enhancer-binding protein beta (C/EBPB), GATA binding protein-2 (GATA-2) and signal transducers and activators of transcription protein (STAT5) [34]. Although these transcription factors are involved in the cellular response to decreases in oxygen availability, the family of hypoxia-inducible factors (HlFs) comprises the principal regulators of the transcriptional response to hypoxia. HlFs are the most widely studied family of hypoxia responsive transcription factors and they directly influence the expression of more than 1 50 genes. HlFs belong to the basic Helix-Loop-Helix (bHLH)«Per/Amt/Sim 18 Figure 1.2. Structure of human HIF1a protein and oxygen- dependent modification of HIF1a. The stability and transcriptional activity of HlF1oc is regulated in oxygen- dependent manner. The oxygen-dependent degradation domains contain conserved prolyl residues (P) 402 and 564 that undergo hydroxylation in the norrnoxia conditions. The hydroxylated prolyl residues, including acetylated lysyl (K-532) residue, form a binding site for von Hippel-Lindau tumor suppressor protein (pVHL). The oxygen- dependent hydroxylation of asparagyl residue (N-803) blocks the binding of CBP and p300 transcriptional regulators and consequently inhibits HIF1 mediated transcription. "Images in this dissertation are presented in color." 19 co=m>=o< .mcozntowcm; comaimo i cozmnfimoo 5:25:33: 1 .mEomwoaoE (PAS) superfamily of transcription factors [35] (Fig. 1.1). HlFs are heterodimeric factors consisting of a (HIF1o, HIF20, and HlF3o) and [3 subunits (HIF1[3 (also known as aryl hydrocarbon nuclear translocator, ARNT) and ARNT2 [36]. These heterodimers bind at A/(G)CGTG consensus sites in the hypoxia response elements (HREs) in the promoter regions of hypoxia responsive genes. HIFs consists of an N-tenninal basic helix-Ioop—helix (bHLH) domain, a central PAS domain (named for the three founding members of the family, Per, ARNT, and Sim) and two C-terminal transactivation domains (TADs) (Fig. 1.1). The bHLH acts to bind DNA and is a primary dimerization surface. The PAS domain is a secondary dimerization surface, dictating specificity of the interactions between various PAS proteins. The TAD closest to the C-terminus (C-TAD) is the primary activator of transcription. The interior TAD (N-TAD) has Iowertransactivation activity. The unique feature that sets HlFas apart from HlFBs is the presence of an oxygen- dependent degradation domain (ODD) overlapping the N-TAD. These ODDs are responsible for rendering the protein oxygen labile. The ODD contains two conserved prolyl residues (Pro402 and Pro564 in human HlF1a; Pro402 and Pro577 in murine HlF1a) which are post-translationally hydroxylated under normoxic conditions [37] (Fig. 1.2). The family of enzymes, prolyl hydroxylase domain containing proteins (PHDs), responsible for this modification are related to the hydroxylase involved in collagen synthesis [38]. PHDs were first identified in C. elegans as egg laying abnormal nine homologs (EGLN) and 21 Figure 1.3. Hypoxia Signaling in normoxic and hypoxic conditions. The hypoxia signaling in the normoxic conditions begins in the cytosol where PHDs continuously respond to the changes in oxygen concentrations. Under normoxic conditions, PHDs hydroxylate HlFa that, in turn, leads to the pVHL mediated proteasomal degradation thus inhibition of HIF1 mediated transcription (A). Under hypoxic conditions, PHDs activity is inhibited leading to the stabilization of HlF1a. Stabilized HlF1oc translocates into the nucleus where it binds to HIF1B to form active HIF1heterodimer. "Images in this dissertation are presented in color." 22 fi Normoxia l 2+ Fe Ascorbate O 2 2-OG l ' i, uccinate ' HIFIa o/"\ ‘ HO I‘ ’ 'r‘, Nucleus I, ‘ '0 {it I i we 3 I: '3 ‘1' g ARNT l Ubiquitination g X I é“ l t 3 RCGTG ] Cytoplasm g A Hypoxia 2+ Fe Ascorbate o2 Z-OG pVHL x Succinate ' 4 ii Hw1a Cytoplasm Nuclear Translocation AR RCGTG Nucleus NT Target Gene . 3 mRNA 23 later shown to have three mammalian homologs (PHD1-3) [39]. PHDs are Fe(ll)-, ascorbate-, and 2-oxoglutarate-dependent dioxygenases that utilize oxygen as a co-substrate. One oxygen atom of the oxygen molecule is used in the hydroxylation of the HlFa prolyl residue (within a consensus LXXLAP amino acid sequence) and the other reacts with 2-oxoglutarate, yielding succinate and C02 as products [40]. Once hydroxylated, the HlFoc is recognized by the tumor suppressor protein, von-Hippel Lindau (pVHL), which acts as the recognition component of E3-ubiquitin ligase complex and targets HIFa for proteolysis by the ubiquitin—proteasome pathway [41] (Fig. 1.3A). Under hypoxic conditions, the PHDs are unable to hydroxylate the cytoplasmic HIFa, which results in its stabilization and accumulation in the cytoplasm (Fig. 1.3B). There are two proposed mechanisms underlying the inhibition of PHDs under hypoxic stress. First, direct loss of substrate (i.e oxygen) decreases the enzymes activity. The second involves hypoxia- induced changes in reactive oxygen species (ROS) levels within the cytoplasm that impacts various aspects of PHD’s function, including iron redox cycling. Regardless of mechanism, it has been established that hypoxia leads to PHD inhibition and subsequent stabilization of HlFa that initiates the adaptation responses through gene expression [37]. Besides hypoxia, PHDs Can also be inhibited by certain compounds referred to as hypoxia mimics. These mimics include cobalt (competes with Fe(||)). desferrioxamine (DFO, an 24 iron chelator), and dimethyloxalylglycine (DMOG, an aKG analog). Exposure to these compounds results in HIF1a stabilization and subsequent transcriptional response [42]. 4-0. HIF signaling and regulation of gene expression More than 150 genes are directly regulated by HIF-mediated transcription following exposure to hypoxia. It has been estimated that more than 2% of human arterial endothelial cell genome is regulated by HIF-1, directly or indirectly [43]. These genes are involved in a wide variety of functions including cellular energy metabolism, cell proliferation, angiogenesis, matrix and barrier functions, hormonal regulation, cytoskeletal integrity, cell migration, vasomotor regulation, growth and apoptosis [44]. Switching to anaerobic glycolysis is probably the first pro-survival step that cells choose under hypoxic stress (Pasteur effect). Several genes involved in glycolysis and glucose uptake have been identified as HIF1 target genes [45, 46]. Genes involved in cell proliferation such as transforming growth factor-oz and [33 (TGF-a and B3), insulin-like growth factor-2 (IGF-2), IGF binding protein 1, 2, and 3 (IGF-BP), cyclin G2 and WAF1 are some of the known HIF target genes. 25 At the systemic and tissue level, exposure to hypoxia leads to changes in HIF-responsive genes that regulate important processes such as erythropoiesis, angiogenesis, and vascular remodeling. HIF regulated genes, including erythropoietin (EPO), transferrin (TF), transferrin receptor, ceruloplasmin (CP), haemoxygenase-1 (HO-1), VEGF, Flt-1 (VEGF receptor 1), PAI-1, adrenomedullin, endothelin 1 (END-1), inducible nitric oxide synthase (NOSZA), endothelial nitric oxide synthase (N083) and ferrochelatase (FECH) are all involved in enhanced oxygen supply [47]. 4.E. HIF signaling and development Perhaps, the most obvious physiological role of HIF signaling is observed in mammalian embryonic and fetal development. Hypoxia is a consistent feature during gestational development, with p02 ranging from 55 mmHg (conception) to 12 mmHg (late gestation). Despite the placental formation, the late gestational p02 values in umbilical artery, umbilical vein and amniotic fluid remains below approximately 23, 30 and 12 mmHg, respectively [48]. Under these hypoxic conditions, embryonic and fetal stages rely on the mechanisms dedicated to maintain oxygen supply and using the p02 as a developmental cue. 4.E.1. Placentation and hypoxia signaling 26 During oocyte release and fertilization the oxygen delivery is diffusion mediated. These stages are the most anaerobic stage of fetal development due to several factors, including increase in the diffusion distance of oxygen to the mitochondria and poor vascularization and low intrauterine p02. In order to meet the increasing oxygen demands of the growing conceptus, the process of placentation is very important. The first phase of placentation is characterized by cytotrophoblastic proliferation into the endometrial stroma followed by cell fusion and formation of syncytium. The mass formed by the fusion of invading cytotrophoblasts is called the syncytiotrophoblasts and it massively increases the surface area available for nutrient exchange between the mother and the fetus. The second phase of placentation includes the differentiation of remaining (non-syncytial) cytotrophoblasts to form primary villi that penetrate the maternal decidua, myometrium and vasculature which ultimately develop to form true placental gas exchange interface. During placentation, the morphogenesis of placental gas exchange interface is regulated by oxygen tension in two different forms of cytotrophoblasts. The proliferative form of cytotrophoblasts is undifferentiated which is characteristically prominent at the low p02 (fetal p02) whereas the other non-proliferative (differentiated, motile and invasive) form appears at arterial p02 [49]. The formation of placental gas exchange interface is thus, 27 precisely controlled by oxygen tension surrounding the tissues through an oxygen sensor mechanism. The oxygen sensor involved in the transduction of change in oxygen tension to the physiological signal (placental gas exchange interface establishment) has been recognized as PHDs, with the effector pathway as HIF signaling [50]. It has been demonstrated that HlF1ot mRNA is expressed maximally between 5 to 8 weeks of gestation and falls abruptly at 12 weeks (corresponds to E95 and E10.5 of mouse fetus) with the establishment of placental gas exchange process [51]. This finding suggested the possible role of HIF responsive genes in the process of placentation. Later it was shown that antisense mediated knockdown of HIF1a in the cultured villous implants at fetal p02 resulted in the adoption of the differentiated, non-proliferative, motile, and invasive form of cytotrophoblasts, which normally appears at arterial p02. It suggested that HIF1a stabilization at low p02 signals the retention of undifferentiated and proliferative form of cytotrophoblasts that is relieved after the p02 is raised to arterial levels leading to HlF1a degradation [52, 53]. It has also been shown that the knockdown of HlF1a results in the suppression of TGFB3 (HIF1 target gene) which is responsible for the switch from the proliferative to the invasive form. This switch was reversed on the addition of recombinant TGFB3 [49, 54, 55]. Further, to explore the HIF 28 signaling underlying the roles of hypoxia in placental development, various murine models with disrupted HIF pathways have been generated [50, 54, 56]. Disruption of all the three PHDs (PHD1, PHDZ and PHD3) in the murine model has shown that mice lacking PHD1 (Phd1 'l') and PHD3 (Phd3 'l') had -/- apparently normal placental development whereas Phd2 exhibited several placental defects such as abnormal distribution of trophoblast giant cells, reduced villous branching in the labyrinth, and widespread penetration of the -/. labyrinth by syncytiotrophoblasts. Due to these defects, Phd2 embryos exhibit lethality on day E 12.5 [50]. The disruption of PHDZ led to the upregulation of HlF1oc as well as HlF2a in the placenta and embryonic tissues ./- - - [50]. Contrary to Phd2 mice, Vhl I mice exhibited more severe defects in the placental development such as failure to develop syncytiotrophoblasts, lack of invasion of maternal blood vessels leading to embryonic lethality at £95 to E10.5 (stage of placental dysgenesis) [57, 58]. The roles of HIF1B (ARNT) in the placental development were reported in three different studies showing embryonic mortality at day E9.5 due to defective vascularization of labyrinth of placenta and yolk sac, an increased number of trophoblast giant cells and defective cell fate determination [54, 59-61]. These differences suggest additional roles of PHD2 and VHL other than mediation of HIF stabilization or additional mechanism of regulation of HIFs activity [57]. 29 A detailed study on the effect of different allelic dosage combinations of . +/- +/+ -/- +/+ +/- +/- -/- eitheror both HlFoc(HIF1a l2a ,HlF1a /20L ,HIF10t /20t ,HIF10t -/— +/+ +/- +/+ -/- /20t , HlF1oc l2a , HIF1oc l2a ) on the placental development have -/- -/- been undertaken. HIF10t /2a mice showed absence of placental vasculature, increase in trophoblastic giant cells, poor invasive efficiency and defective placental structure thus representing phenocopy of ARNT deficient -/- mice [56]. The defective phenotype of HlF1cx were further aggravated by deleting either or both alleles of HIF2a, which suggested that the phenotypes among different dosage combinations showed allele-specificity. HIF10t-l- mice exhibited defective chrioallantoic fusion with maternal lacunae and fewer and -/- defective placental vascularization that were normal in E 9.5 HIF2a embryos [56]. These results suggest that hypoxia and its signaling is also important in the embryonic development of organ systems. 4.E.2. Hypoxic regulation of organogenesis It is clear from above studies that murine germ line inactivation of either HIF1oc, HIF2a, ARNT, PHD2 or the VHL tumor suppressor results in embryonic or perinatal lethality. To overcome embryonic lethality, conditional 30 alleles for HIF1a, HIF20L, ARNT and VHL have been generated that now allow tissue and cell type specific knock down. Several murine knockouts of HIF, pVHL, PHD, and VEGF have demonstrated the essential role of hypoxia- responsive pathway proteins in organogenesis. 4.E.2.a. Vascular System Embryonic vasculature development is a crucial process contributing to the survival of the fetus and development of the other organ systems. Disruption studies for different members of HIF signaling pathways have contributed vastly towards our understanding of embryonic vascularization events. Homozygous deficiency of ARNT leads to defective angiogenesis of the yolk sac [61]. Homozygous deficiency of HlF2a in murine embryo results in vascular disorganization in yolk sac and failure to assemble and maintain vascular tubular structure and leading eventually to death between day E95 and E13.5 [62, 63]. Similarly, the HlF1oc knockout mouse displayed disorganized yolk sac vascularization with disorganized branching pattern. HlF1a deficient embryos exhibited only a few capillaries in the neural fold with complete absence of vascular network and also reduced size of dorsal aorta [64]. Current insights in to the role of different HlFs in the vascularization clearly define the importance of oxygen tension, hypoxia and HIF signaling. 4.E.2.b. Cardiac System 31 The role of HIF signaling in cardiac development has been extensively studied by various groups using systemic, as well as, tissue specific mice models. Systemic HlF1a knockout mice display cardiac defects such as myocardial hyperplasia, pericardial obliteration of ventricular lumen, increased number of myocardial cells and cardia bifida (bilateral heart tubes that develop + - in to two hearts) [64-67]. Inactivation of single allele of HlF1a (HlF1a I ) had no observable developmental phenotype, however, further analysis of these partially deficient adult mice revealed abnormal physiological responses to +/- chronic hypoxic exposures. As compared to control animals, HlF1a mice exhibited impaired physiological responses to hypoxia such as weight loss, polycythemia, right ventricular hypertrophy, pulmonary hypertension and pulmonary vascular remodeling [68]. The cardiac myocytes specific HlF1oc deletion do not exhibit any lethality phenotypes or abnormalities, however, these mice exhibited physiological abnormalities in cardiac function, vascularity, energy availability, and calcium handling even under nonnoxia conditions, suggesting that HIF-1o coordinates gene expression at all oxygen levels [69]. Although systemic HlF2a null progeny from congenic CS76BL/6J strain failed to survive due to embryonic or perinatal lethality, progeny from isogenic 12986/SvaTac strain survived with multi-organ pathologies. The surviving HlF2a./. progenies exhibited cardiac mitochondrial hypertrophy but no 32 hyperplasia. Cardiac myocytes from HlF2a+ mice contained many degenerating mitochondria with intact inner mitochondrial membranes [70]. In conclusion, deletion of hypoxia signaling proteins results in the defective cardiac development and compromised functioning. 4.E.2.c. Lung Lung morphogenesis proceeds in a consistently low p02 environment characterized by shallow gradients of oxygen from the fetal vasculature into the tissue mass. Several studies have demonstrated that the hypoxic condition that a fetus faces is an important determinant in lung development. It has been well documented that low fetal p02 serves to maintain epithelial lumen fluid secretion and lung expansion [71]. Land et al. have shown that rat lung explants display increased ainNay surface complexity (complex branching pattern) at fetal p02 (23 mmHg) as compared to ambient (142 mmHg) steady- state p02. The observed difference in branching pattern was confined towards the periphery suggesting the hypoxia dependency of mesenchyme differentiation and ainlvay bifurcation events [72]. Fisher et al. have demonstrated that the ROS produced under hypoxic conditions interferes with the ainNay branching morphogenesis, and N-acetylcysteine (antioxidant) treatment afforded protection against hyperoxia [73]. Similarly, several other 33 studies have shown the consequences of redox modulation on ainlvay morphogenesis [74, 75]. Hypoxia and HIF signaling pathway activation have been studied through investigation of the spatial and temporal expression of HIFNEGF pathway proteins in the developing human lung. Expression analysis of staged human embryo at third trimester showed consistent HIF1or expression, and there was no observable increase in the expression of proteins of HlF1a degradation pathway [76]. This finding suggests the continually activated hypoxia signaling in the post-placentation lung. HlF1a expression was restricted to branching epithelium, whereas HlF2a appeared to be present in the vascular structures as well as branching epithelium, reflectingtheir different roles in pulmonary development. HIF1B was shown to be present in the mesenchymal as well as epithelial structures, providing a basis for downstream activation of both HlF1a and HIF20L [76]. All of the systemic knockout mice models for genes involved in the HIF signaling die in utero from various defects in organogenesis. However, some of the HlF2a knockout embryos that survived full term exhibited postnatal respiratory distress due to insufficient surfactant production [77]. The midgestational lethalities of the systemic knockout models has made it challenging to study the role of HlFs in fetal lung development. 34 5. Hypoxia and its effects on the lung Alveolar epithelial cells require normal oxygenation to run their high energy demanding processes like surfactant biosynthesis, fluid uptake, and maintenance of alveolocapillary barrier. Several clinical conditions involve hypoxic challenge to the alveolar epithelial cells resulting in functional changes [78]. Alveolar epithelial cells survive hypoxia through several adaptive mechanisms like increased glycolysis, increased vasculogenesis, downregulation of high energy consuming Na,K-ATPase activity and protein synthesis. Alveolar edema clearance requires active Na,K-ATPase. Thus, downregulation of this protein in hypoxia results in the altered fluid uptake and edema clearance. The most well studied hypoxia-affected processes in alveolar epithelium are the activity and maintenance of Na,K-ATPase and amiloride-sensitive epithelial sodium channel (ENaC) pumps. Acute hypoxemic respiratory failure is associated with the flooding of the alveolar spaces with fluid and the resorption of this fluid into the interstitial and vascular spaces depends on these two ion pumps. The amiloride-sensitive ENaC pump actively uptakes sodium ions at the apical surface of alveolar epithelial cells, whereas at the basolateral surface sodium ions are actively transported against a gradient in exchange for potassium ions into the interstitium, predominantly by Na,K—ATPase. The development of ion gradient due to the movement of sodium ions results in the movement of water into the epithelial cells and eventually in to the interstitium via aquaporins [79-82]. High-altitude 35 pulmonary edema (HAPE), a life-threatening condition characterized by alveolar flooding that occurs in predisposed individuals at high altitudes, has been linked to hypoxic pulmonary vasoconstriction [83]. A direct role of hypoxia in alveolar epithelium is not well characterized. Hypoxia (1.5% 02 for 60 min) induced Na,K-ATPase trafficking out of the plasma membrane into the intracellular membranes, which suggests a role for hypoxia in the regulation of topological distribution of these pumps [84]. It has also been shown that hypoxia downregulates plasma membrane specific Na,K-ATPase activity without affecting the whole cell protein expression levels [85]. These effects are observed under short-term severe hypoxic exposure and are reversible. In contrast, prolonged hypoxic conditions results in reduction in the cellular pool of Na,K-ATPase [78]. A549 cells exposed to 1.5% 02 for 2 hours displayed approximately a 50% loss of the plasma + membrane bound ATP-driven Na pumps. These findings suggest that the cells undertake this adaptive strategy to save energy and reduce oxygen consumption through the inhibition and/or degradation of energy consuming metabolically active molecules [86]. 6. Hypoxia Mimics and HIF signaling 36 HlFa stabilizers are also known as hypoxia mimics. Agents such as metals (Ni(ll), Co(ll), Mn(ll), and V(V)), desferroxamine (DFO) and dimethyloxallyl glycine (DMOG) are commonly studied hypoxia mimics. It has been shown that cobalt can stabilize HlFa, and HlFa modulates cobalt induced cell toxicity [87]. Additionally, occupational exposure of metals like cobalt (Co(ll)) are known to cause lung injury. Hence, it is rational to use cobalt-induced lung injury models to understand the role of HIF signaling in various respiratory diseases involving hypoxia such as respiratory distress syndrome, HAPE and COPD. 7. Metals 7.A. History of metals It was in the new Stone Age that ancient man started using metal rich rocks as weapons and tools. The Bronze Age (3000-1000 BC) began with the advent of metallurgical technique of mixing tin ore with copper ore to produce bronze. Bronze proved to be a very useful alloy in the production of new tools for farming and hunting. It was not until the start of Iron Age (1500-800 BC) that bronze was displaced as the most widely used metal and brought the advent of the steel industry. The New Metal Age has also posed several health concerns especially for the workers who are involved in mining and metal industries [88-90]. 37 7.B. Biological significance of metals The metal’s property to donate its electrons to become cations and hence, electrophiles, makes these cations an important component of the active site of enzymes with redox reaction mechanisms. In addition, some metal ions form an integral part of nonenzyme proteins including hemoglobin, ferritin, ceruloplasmin, rubredoxin, cytochromes, and cyanocobalamin. Proteins containing bound metal ions are referred as metalloproteins (metalloenzymes). The metal ion forms coordinated bonds with nitrogen, sulfur or oxygen atoms of amino acids in the polypeptide chain. Approximately 25-33% of proteins require metals for proper function or maintenance of structural integrity. Metalloproteins such as cytochromes play important roles in cellular energy production via their role in the electron transport chain (ETC). In addition, metalloproteins play critical roles in processes such as signal transduction (calmodulin), metabolism (cyanocobalamin), antioxidation (superoxide dismutase), and transport (ceruloplasmin). Not all the metals are biologically beneficial. Metals such as lead and mercury are toxic to the cellular machinery. Another group of metals known as trace metals (Mn, Fe, Co, Ni, Cu, Zn, and Cd), are required in minute concentrations for cellular functions but at higher concentrations they pose health hazards . 7.C. Toxicity of Metals 38 Metals are probably the oldest known toxicants in human medicine. For many, the mechanism of action that leads to toxicity is not well characterized for many of them. Biogeological cycles and industrialization have redistributed metals in the environment and lead to the appearance of new metals in the food chain, soil, air, and water. When injected into the atmosphere, gaseous or particulate metals may be transported to distant places [91]. The toxicological effects of metals depend on the time, dose and route of exposure [92]. Toxicities are further determined by other factors such as half-life in the tissues (varies among tissues), valence state, and ligand binding. Hexavalent chromium is highly toxic whereas its trivalent form functions as an essential trace element. Finally, organometallic forms of some metals differ in their toxic properties. For instance, non ionized alkyl compounds such as methyl mercury Is a neurotoxin, whereas mercuric chloride is a nephrotoxic compound [92]. A majority of transition metals are characterized as nonessential to the cellular machinery and are toxic at minimal doses. The few, such as cobalt, that are required for cellular functions, show toxic phenotypes at higher concentrations. These toxic phenotypes fall under three categories: disturbance of normal metal ion equilibrium, damage to biomolecules, and alteration in gene expression patterns. Increased concentrations of divalent cobalt interfere with iron metabolism and compete for the iron-binding site on metalloproteins and metalloenzymes. Certain metal exposures result in 39 damage to biomolecules such as proteins and nucleic acids. Elements such as As, Hg and Cd form covalent bonds with the —SH group of proteins. For example, arsenic can inactivate the pyruvate dehydrogenase complex through covalently binding to the -SH group of dihydrolipoic acid [93]. Some redox active transition metals including cobalt generate reactive ROS that, in turn, oxidize cellular proteins, nucleic acid and lipids [94, 95]. Metals regulate and alter the activity and/or stability of various metal- responsive transcriptional factors that cause changes in the level of gene expression and activate various signaling pathways. Metals and metal- induced ROS generation have been demonstrated to affect a number of receptors and genes, including growth factor receptors, src kinases, ras signaling, mitogen-activated protein kinases, and nuclear transcription factors such as NFKB, AP-1, p53, MTF-1, NFAT, and HIF-1 [96]. NFKB is an important redox sensitive transcription factor that is affected by metal exposure resulting in oxidative stress. It has been shown that divalent nickel and cobalt exposure causes NFKB induced upregulation of adhesion molecules ICAM-1, VCAM-1, and E-selectin in endothelial cells [97]. AP-1 is another nuclear transcription factor that is stimulated by metal induced oxidative stress. Arsenic(|l), vanadium(V), chromium(Vl), nickel(ll), cadmium(ll), lead, cobalt, and iron are known to activate AP-1 in a variety of cells [96]. Metallothioneins (MTs) are a class of cysteine-rich proteins that play a critical roles in the heavy metal metabolism and detoxification. Metal- 40 induced expression of MTs depends on the zinc-finger transcription factor, metal transcription factor-1 (MTF-1) which acts as a cellular stress-sensor protein [98]. MTF-1 binding to the metal responsive element on the MTF-1 targeted genes is allosterically regulated by zinc leading to elevated MTs expression [99, 100]. Certain metals such as Co(ll), Ni(ll), V(ll), Mn(ll) are referred as “hypoxia mimics” due to their ability to stabilize HlFoc. Cobalt has been known to induce the expression of erythropoietin and thus this metal was used to treat anemia. [101, 102]. Cobalt also activates angiogenic responses through VEGF expression [103]. These responses are known to be mediated through HlFa stabilization. Several different mechanisms have been proposed for the cobalt-mediated stabilization of HlF1a however; the exact mechanism is still not well understood. It has been shown that cobalt inhibits pVHL binding to the ODD domain of HIFot [104]. In addition, cobalt-induced ROS generation leading to ferrous oxidation, chelating of ascorbic acid and replacing non-covalently held iron atom from the PHDs have also been suggested as mechanisms for the metal’s ability to stabilize HIFa [105]. 8. Cobalt 8.A. Occurrence and uses Cobalt is a rare magnetic transition metal and falls in period four of the periodic table with nickel, iron and manganese. It primarily occurs in nature as 41 oxides, arsenides and sulfides, and its cobaltous (Co(ll)) state is predominantly used in the chemical industry [106, 107]. Historically, it has been used as a dye in pottery, oil paints, high speed drills and cutting machines as a component of hard metal (tungsten carbide (BO-95%) combined with cobalt (5-20%) and nickel (0-5%)). In present industrial settings, it is primarily used in the production of superalloys, corrosion resistant alloys, high speed steels, batteries, magnets, wear resistant coatings and various metallurgical applications [95]. Other industrial uses include diamond polishing with cobalt containing disks, radioactive isotopes in medicine and the production of drying agents, pigments, and catalysts [107]. SB. Cobalt metabolism and Exposure Cobalt is an essential trace element for humans and is consumed as cobalamin (essential component of vitamin B12). Cobalamin is required for the production of red blood cells and the prevention of pernicious anemia. In addition, cobalamin is also required in the methionine and nucleic acid biosynthesis pathways. For the general population, diet is the main source of exposure and the average daily dietary intake ranges from 5 to 45 pg. Major dietary sources of cobalt are seafoods, bran, cocoa, and certain meats [108]. Generally, the concentration of cobalt in the drinking water is low (less than 5 rig/L) I107]. 42 Cobalt salts are well absorbed in the jejunum and without significant accumulations, 80% of ingested cobalt is excreted in the urine while 15 % is excreted in feces via enterohepathic pathway. In the human body, the total amount of cobalt is estimated to be approximately 1.1 mg. It has been shown that muscles contain the largest fraction whereas its highest concentration is found in fat tissues. The normal levels in human urine and blood are about 1.0 and 0.18 ug/L, respectively. Cobalt is found to be present in urbanized air in the range between 0.5 to 60 ng/m3 [109]. Tobacco smoke contains 0.3-2.3 mg Co/kg dry weight, which puts smokers at higher risk of cobalt-induced lung injury [110]. In industrial settings, metal refinery workers and those involved in the production of certain alloys, such as tungsten carbide, are at greatest risk for cobalt- induced toxicity. 8.C. Cobalt toxicity Despite its use for treatment of anemic patients, associated toxic effects were reported to be goitrogenic due to hypothyroidism and thyroid hyperplasia [111]. Epidemiological studies have shown an association between goiter prevalence and the elevated cobalt levels in water and soil. Cobalt induced cardiomyopathy was observed in brewery workers who consumed cobalt- contaminated beer. Postmortem findings included pericardial effusion, 43 myofibrillar loss and congestive heart failure [112]. Excessive erythrocytosis (polycythemia) has been reported in cobalt miners [113]. In humans, gastrointestinal effects such as vomiting, diarrhea, liver injury and allergic dermatitis were also reported in case of oral exposure to cobalt [114]. Acute inhalation exposure of cobalt in humans results in respiratory problems such as congestion, edema, and hemorrhage of the lung. Chronic exposure to cobalt through inhalation leads to respiratory irritation, coughing, shortness of breath, wheezing, asthma, pneumonia, and fibrosis [107]. Inhalation exposure of hard metal workers has been shown to cause hard metal lung disease that is interchangeably used in literature with cobalt lung, hard metal asthma, and giant cell interstitial pneumonitis. Ambient 3 concentrations of 0.002 to 0.01 mg/m have been shown to cause respiratory 3 irritation, and higher concentrations (0.1 mg Co/m or higher) can cause hard metal lung disease [92]. 8D. The role of HIFs in cobalt-mediated toxicity The role of hypoxia signaling in cobalt-induced cellular toxicities has been studied in various in vitro models. Cobalt chloride induced toxicity has been tested in mouse embryonic fibroblasts (MEFs) and the in vitro role of HlF1a has already been established in cobalt-induced toxicity [87]. The cobaltous chloride-induced oxidative stress mechanism has been investigated 44 in skeletal muscle cell lines, where exposure to CoCl2 resulted in elevated intracellular oxidants, the accumulation of HlF1a protein, and the expression of VEGF [115]. In addition, CoCl2-induced astrocyte toxicity is mediated through HIF1a regulated apoptosis [116]. Hypothesis and Specific Aims Cobalt exposure leads to pathological conditions such as airway constriction, alveolitis, fibrosis and associated giant cell interstitial pneumonitis. There is little known, however, about the role of hypoxia signaling in these conditions. Several in vitro studies have shown that HlF1a is an important mediator of the toxic responses observed upon cobalt exposure. Previous studies in our lab, using an immortalized mouse embryonic fibroblast (MEFs) cell line lacking HlF1o, established the correlation between HIF1o removal and protection against CoCI2 toxicity. Validation of our in vitro understanding of cobalt-induced HlFs signaling necessitates the in vivo studies of cobalt toxicity in HlFs null animals. Based on these previous findings, I propose the following hypothesis. Hypothesis: Cobalt-induced lung toxicity is mediated by HIF stabilization and transcription and the subsequent changes in inflammatory mediators and cellular response. 45 Each of the chapters in this dissertation is dedicated to address following specific aims. Aim 1: Create and characterize a lung specific inducible HIF1q deficient mouse model. To validate in vitro data, a lung-specific inducible HIF1a deficient mouse model was generated. The generated mice were characterized using techniques such as histopathology, western blotting, real-time PCR and electron microscopy. Aim 2: Determine the role of HIF1ot in cobalt-induced lung injury in mice. A postnatal strategy to delete HlF1oc from the lung epithelial cells was adopted. HIF1a deficient mice were used to study the time-dependent progression of cobalt-induced lung inflammation. Aim 3: Create and characterize combined HIF1a and HIFZa lung specific inducible knockout model (quadruple transgenic model). To investigate the role of HIF20L in the lung development, another prominent isoforrn of HlFa in lungs, lung-specific inducible HlF2a deficient mice were generated. To further enhance our understanding of the collective role of these two HIFs in lung development, mice with a lung-specific deletion 46 of HIF20 (termed HIFZGNA) and HIF1/20 (termed HIF1/20AM) were generated. Importance: The findings from these studies will have a great impact on our current understanding on the roles of HlF1o and HIF2a in the lung development. The studies on cobalt exposures in HIF-deficient mice will provide a mechanistic understanding on the roles of these transcription factors in airway inflammatory responses. 47 REFERENCES 1 10 11 Wagner, P. D. (2008) The biology of oxygen. Eur Respir J. 31, 887-890 Bunn, H. F. and Poyton, R. O. (1996) Oxygen sensing and molecular adaptation to hypoxia. Physiological reviews. 76, 839-885 Burri, P. H. (1984) Fetal and postnatal development of the lung. Annual review of physiology. 46, 617-628 Alescio, T. and Cassini, A. (1962) Induction in vitro of tracheal buds by pulmonary mesenchyme grafted on tracheal epithelium. The Journal of experimental zoology. 150, 83-94 Wessells, N. K. (1970) Mammalian lung development: interactions in formation and morphogenesis of tracheal buds. The Journal of experimental zoology. 175, 455-466 Spooner, B. S. and Faubion, J. M. (1980) Collagen involvement in branching morphogenesis of embryonic lung and salivary gland. Developmental biology. 77, 84-102 Masters, J. R. (1976) Epithelial-mesenchymal interaction during lung development: the effect of mesenchymal mass. Developmental biology. 51, 98-108 Bucher, U. and Reid, L. (1961) Development of the mucus-secreting elements in human lung. Thorax. 16, 219-225 Kauffman, S. L. (1980) Cell proliferation in the mammalian lung. lntemational review of experimental pathology. 22, 131 -1 91 Crapo, J. 0., Barry, B. E., Foscue, H. A. and Shelbume, J. (1980) Structural and biochemical changes in rat lungs occurring during exposures to lethal and adaptive doses of oxygen. The American review of respiratory disease. 122, 123-143 Evans, M. J., Cabral-Anderson, L. J. and Freeman, G. (1978) Role of the Clara cell in renewal of the bronchiolar epithelium. Laboratory 48 12 13 14 15 16 17 18 19 20 21 investigation; a journal of technical methods and pathology. 38, 648- 653 Davies, G. and Reid, L. (1970) Growth of the alveoli and pulmonary arteries in childhood. Thorax. 25, 669-681 Burri, P. H. (1974) The postnatal growth of the rat lung. 3. Morphology. The Anatomical record. 180, 77-98 Burri, P. H., Dbaly, J. and Weibel, E. R. (1974) The postnatal growth of the rat lung. l. Morphometry. The Anatomical record. 178, 711-730 Ji, C. M., et al. (1995) Pulmonary cytochrome P-450 monooxygenase system and Clara cell differentiation in rats. Am J Physiol. 269, L394- 402 Mason, R. J. (1987) Surfactant synthesis, secretion, and function in alveoli and small ainrvays. Review of the physiologic basis for pharrnacologic intervention. Respiration; international review of thoracic diseases. 51 Suppl 1, 3-9 Hong, K. U., et al. (2004) Basal cells are a multipotent progenitor capable of renewing the bronchial epithelium. The American journal of pathology. 164, 577-588 Nettesheim, P., et al. (1990) Pathways of differentiation of ainlvay epithelial cells. Environmental health perspectives. 85, 317-329 Park, K. S., et al. (2006) Transdifferentiation of ciliated cells during repair of the respiratory epithelium. American journal of respiratory cell and molecular biology. 34, 151-157 Stripp, B. R. and Reynolds, S. D. (2008) Maintenance and repair of the bronchiolar epithelium. Proceedings of the American Thoracic Society. 5, 328-333 Richard Harding, K. P., Charles Plopper. (2004) The Lung: Development, Aging and The Environment. Academic Press 49 22 23 24 25 26 27 28 29 30 31 32 Jiang, J., et al. (1996) Measurement of P02 in liver using EPR oximetry. J Appl Physiol. 80, 552-558 Scheufler, K. M. (2004) Tissue oxygenation and capacity to deliver 02 do the two go together? Transfus Apher Sci. 31, 45-54 Lee, Y. M., et al. (2001) Determination of hypoxic region by hypoxia marker in developing mouse embryos in vivo: a possible signal for vessel development. Dev Dyn. 220, 175-186 Land, 8. C. (2003) Oxygen-sensing pathways and the development of mammalian gas exchange. Redox Rep. 8, 325-340 Cartwright, J. E., Keogh, R. J. and Tissot van Patot, M. C. (2007) Hypoxia and placental remodelling. Advances in experimental medicine and biology. 618, 113-126 Guzman, G. J., Cueto, A. H. and Yepez, S. H. (2008) Contribution of hypoxia in pulmonary tissue remodeling in asthmatic processes. Rev Alerg Mex. 55, 18-32 Fajardo, I., Svensson, L., Bucht, A. and Pejler, G. (2004) Increased levels of hypoxia-sensitive proteins in allergic ainrvay inflammation. American journal of respiratory and critical care medicine. 170, 477-484 Michiels, C. (2004) Physiological and pathological responses to hypoxia. The American journal of pathology. 164, 1875-1882 Bartrons, R. and Caro, J. (2007) l-lypoxia, glucose metabolism and the Warburg's effect. Journal of bioenergetics and biomembranes. 39, 223- 229 Hardie, D. G. and Hawley, S. A. (2001) AMP-activated protein kinase: the energy charge hypothesis revisited. Bioessays. 23, 1112-1119 Rolfe, D. F. and Brown, G. C. (1997) Cellular energy utilization and molecular origin of standard metabolic rate in mammals. Physiological reviews. 77, 731-758 50 33 34 35 36 37 38 39 40 41 42 Buttgereit, F. and Brand, M. D. (1995) A hierarchy of ATP-consuming processes in mammalian cells. The Biochemical journal. 312 ( Pt 1), 163-167 Cummins, E. P. and Taylor, C. T. (2005) Hypoxia-responsive transcription factors. Pflugers Arch. 450, 363-371 Wang, G. L., Jiang, B. H., Rue, E. A. and Semenza, G. L. (1995) Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proceedings of the National Academy of Sciences of the United States of America. 92, 5510-5514 Semenza, G. L., et al. (1997) Structural and functional analysis of hypoxia-inducible factor 1. Kidney lntemational. 51, 553-555 Weidemann, A. and Johnson, R. S. (2008) Biology of HIF-1alpha. Cell death and differentiation. 15, 621-627 Kaelin, W. 6., Jr. and Ratcliffe, P. J. (2008) Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway. Molecular cell. 30, 393-402 del Peso, L., et al. (2003) The von Hippel Lindau/hypoxia-inducible factor (HIF) pathway regulates the transcription of the HIF-proline hydroxylase genes in response to low oxygen. The Journal of biological chemistry. 278, 48690-48695 Epstein, A. C., et al. (2001) C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell. 107, 43-54 Cockman, M. E., et al. (2000) Hypoxia inducible factor-alpha binding and ubiquitylation by the von Hippel-Lindau tumor suppressor protein. The Journal of biological chemistry. 275, 25733-25741 Groenman, F. A., et al. (2007) Effect of chemical stabilizers of hypoxia- inducible factors on early lung development. Am J Physiol Lung Cell Mol Physiol. 293, L557-567 51 43 44 45 46 47 48 49 50 51 52 Manalo, D. J., et al. (2005) Transcriptional regulation of vascular endothelial cell responses to hypoxia by HIF-1. Blood. 105, 659-669 Chowdhury, R., Hardy, A. and Schofield, C. J. (2008) The human oxygen sensing machinery and its manipulation. Chemical Society reviews. 37, 1308-1319 Wenger, R. H. (2002) Cellular adaptation to hypoxia: OZ-sensing protein hydroxylases, hypoxia-inducible transcription factors, and 02- regulated gene expression. Faseb J. 16, 1151-1162 Wenger, R. H. (2000) Mammalian oxygen sensing, signalling and gene regulation. The Journal of experimental biology. 203, 1253-1263 Rocha, S. (2007) Gene regulation under low oxygen: holding your breath for transcription. Trends in biochemical sciences. 32, 389-397 Land, S. C. (2004) Hochachka's "Hypoxia Defense Strategies” and the develOpment of the pathway for oxygen. Comparative biochemistry and physiology. 139, 41 5-433 Caniggia, I., et al. (2000) Hypoxia-inducible factor-1 mediates the biological effects of oxygen on human trophoblast differentiation through TGFbeta(3). The Journal of clinical investigation. 105, 577-587 Takeda, K., et al. (2006) Placental but not heart defects are associated with elevated hypoxia-inducible factor alpha levels in mice lacking prolyl hydroxylase domain protein 2. Molecular and cellular biology. 26, 8336- 8346 Rodesch, F., Simon, P., Donner, C. and Jauniaux, E. (1992) Oxygen measurements in endometrial and trophoblastic tissues during early pregnancy. Obstetrics and gynecology. 80, 283-285 Cowden Dahl, K. D., et al. (2005) Hypoxia-inducible factors 1alpha and 2alpha regulate trophoblast differentiation. Molecular and cellular biology. 25, 1 0479-1 0491 52 53 54 55 56 57 58 59 60 61 62 Fryer, B. H. and Simon, M. C. (2006) Hypoxia, HIF and the placenta. Cell cycle (Georgetown, Tex. 5, 495-498 Adelman, D. M., et al. (2000) Placental cell fates are regulated in vivo by HIF-mediated hypoxia responses. Genes & development. 14, 3191- 3203 Schaffer, L., et al. (2003) Oxygen-regulated expression of TGF-beta 3, a growth factor involved in trophoblast differentiation. Placenta. 24, 941- 950 Cowden Dahl, K. D., et al. (2005) Hypoxia-inducible factors 1 alpha and 2 alpha regulate trophoblast differentiation. Molecular and cellular biology. 25, 10479-10491 Gnarra, J. R., et al. (1997) Defective placental vasculogenesis causes embryonic lethality in VHL-deficient mice. Proceedings of the Natibnal Academy of Sciences of the United States of America. 94, 9102-9107 Tang, N., et al. (2006) pVHL function is essential for endothelial extracellular matrix deposition. Molecular and cellular biology. 26, 2519- 2530 Kozak, K. R., Abbott, B. and Hankinson, O. (1997) ARNT-deficient mice and placental differentiation. Developmental biology. 191, 297-305 Abbott, B. D. and Buckalew, A. R. (2000) Placental defects in ARNT- knockout conceptus correlate with localized decreases in VEGF-R2, Ang-1, and Tie-2. Dev Dyn. 219, 526-538 Maltepe, E., et al. (1997) Abnormal angiogenesis and responses to glucose and oxygen deprivation in mice lacking the protein ARNT. Nature. 386, 403-407 Peng, J., Zhang, L., Drysdale, L. and Fong, G. H. (2000) The transcription factor EPAS-1/hypoxia-inducible factor 2alpha plays an important role in vascular remodeling. Proceedings of the National Academy of Sciences of the United States of America. 97, 8386-8391 53 63 64 65 66 67 68 69 70 71 72 Patel, S. A. and Simon, M. C. (2008) Biology of hypoxia-inducible factor-2alpha in development and disease. Cell death and differentiation. 15, 628-634 Ryan, H. E., Lo, J. and Johnson, R. S. (1998) HIF-1 alpha is required for solid tumor formation and embryonic vascularization. The EMBO journal. 17, 3005-3015 Semenza, G. L. (2006) Regulation of physiological responses to continuous and intermittent hypoxia by hypoxia-inducible factor 1. Experimental physiology. 91, 803-806 lyer, N. V., et al. (1998) Cellular and developmental control of 02 homeostasis by hypoxia-inducible factor 1 alpha. Genes & development. 12, 149-162 Compemolle, V., et al. (2003) Cardia bifida, defective heart development and abnormal neural crest migration in embryos lacking hypoxia-inducible factor-1 alpha. Cardiovascular research. 60, 569-579 Yu, A. Y., et al. (1999) Impaired physiological responses to chronic hypoxia in mice partially deficient for hypoxia-inducible factor 1alpha. The Journal of clinical investigation. 103, 691-696 Huang, Y., et al. (2004) Cardiac myocyte-specific HIF-1 alpha deletion alters vascularization, energy availability, calcium flux, and contractility in the normoxic heart. Faseb J. 18, 1138-1140 Scortegagna, M., et al. (2003) Multiple organ pathology. metabolic abnormalities and impaired homeostasis of reactive oxygen species in Epas1-l- mice. Nature genetics. 35, 331-340 Baines, D. L., et al. (2001) Oxygen-evoked Na-I- transport in rat fetal distal lung epithelial cells. The Journal of physiology. 532, 105—1 13 Land, S. C. and Darakhshan, F. (2004) Thymulin evokes lL-6- C/EBPbeta regenerative repair and TNF-alpha silencing during endotoxin exposure in fetal lung explants. Am J Physiol Lung Cell Mol Physiol. 286, L473-487 54 73 74 75 76 77 78 79 80 81 82 Fisher, J. C., Kling, D. E., Kinane, T. B. and Schnitzer, J. J. (2002) Oxidation-reduction (redox) controls fetal hypoplastic lung growth. The Journal of surgical research. 106, 287-291 Kramer, B. W., Kramer, S., lkegami, M. and Jobe, A. H. (2002) Injury, inflammation, and remodeling in fetal sheep lung after intra-amniotic endotoxin. Am J Physiol Lung Cell Mol Physiol. 283, L452-459 Young, S. L., Evans, K. and Eu, J. P. (2002) Nitric oxide modulates branching morphogenesis in fetal rat lung explants. Am J Physiol Lung Cell Mol Physiol. 282, L379-385 Groenman, F., et al. (2007) Hypoxia-inducible factors in the first trimester human lung. J Histochem Cytochem. 55, 355-363 Compemolle, V., et al. (2002) Loss of HIF-2alpha and inhibition of VEGF impair fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice. Nature medicine. 8, 702-710 Jain, M. and Sznajder, J. l. (2005) Effects of hypoxia on the alveolar epithelium. Proceedings of the American Thoracic Society. 2, 202-205 Matthay, M. A., Robriquet, L. and Fang, X. (2005) Alveolar epithelium: role in lung fluid balance and acute lung injury. Proceedings of the American Thoracic Society. 2, 206-213 Matthay, M. A., Folkesson, H. G. and Verkman, A. S. (1996) Salt and water transport across alveolar and distal ainrvay epithelia in the adult lung. Am J Physiol. 270, L487-503 Canessa, C. M., et al. (1994) Amiloride-sensitive epithelial Na+ channel is made of three homologous subunits. Nature. 367, 463-467 McCann, J. D. and Welsh, M. J. (1990) Basolateral K+ channels in airway epithelia. ll. Role in Cl- secretion and evidence for two types of K+ channel. Am J Physiol. 258, L343-348 55 83 84 85 86 87 88 89 90 91 92 Scherrer, U., et al. (1999) High-altitude pulmonary edema: from exaggerated pulmonary hypertension to a defect in transepithelial sodium transport. Advances in experimental medicine and biology. 474, 93-107 Dada, L. A., et al. (2003) Hypoxia-induced endocytosis of Na,K-ATPase in alveolar epithelial cells is mediated by mitochondrial reactive oxygen species and PKC-zeta. The Journal of clinical investigation. 111, 1057- 1064 Carpenter, T. C., et al. (2003) Hypoxia reversibly inhibits epithelial sodium transport but does not inhibit lung ENaC or Na-K-ATPase expression. Am J Physiol Lung Cell Mol Physiol. 284, L77-83 Comellas, A. P., et al. (2006) Hypoxia-mediated degradation of Na,K- ATPase via mitochondrial reactive oxygen species and the ubiquitin- conjugating system. Circulation research. 98, 1314-1322 Vengellur, A. and LaPres, J. J. (2004) The role of hypoxia inducible factor 1alpha in cobalt chloride induced cell death in mouse embryonic fibroblasts. Toxicol Sci. 82, 638-646 Chandler, H. (1998) Metallurgy for the Non-Metallurgist. ASM lntemational Gunnar F. Nordberg, B. A. F., Monica Nordberg, Lars Friberg (2007) Handbook of the Toxicology of Metals Academic Press Nordberg, G. (2007) Handbook on the toxicology of metals. Academic Press, Amsterdam ; Boston Morel, F. M. and Price, N. M. (2003) The biogeochemical cycles of trace metals in the oceans. Science (New York, NY. 300, 944-947 Casarett, L. J., Doull, J. and Klaassen, C. D. (2008) Casarett and Doull's toxicology : the basic science of poisons. McGraw-Hill, New York 56 93 94 95 96 97 98 99 100 101 102 Hu, Y., Su, L. and Snow, E. T. (1998) Arsenic toxicity is enzyme specific and its affects on ligation are not caused by the direct inhibition of DNA repair enzymes. Mutation research. 408, 203-218 Stohs, S. J. and Bagchi, D. (1995) Oxidative mechanisms in the toxicity of metal ions. Free radical biology & medicine. 18, 321-336 Vengellur, A. (2007) The role of HlF1[alpha] signalling in metal-induced toxicity. ed.)"eds.). pp. xiii, 227 leaves, Michigan State University. Dept. of Genetics, 2007. Leonard, S. 8., Harris, G. K. and Shi, X. (2004) Metal-induced oxidative stress and signal transduction. Free radical biology & medicine. 37, 1921-1942 Goebeler, M., et al. (1995) Activation of nuclear factor-kappa B and gene expression in human endothelial cells by the common haptens nickel and cobalt. J Immunol. 155, 2459-2467 Lichtlen, P. and Schaffner, W. (2001) The "metal transcription factor” MTF-1: biological facts and medical implications. Swiss Med Wkly. 131, 647-652 Westin, G. and Schaffner, W. (1988) A zinc-responsive factor interacts with a metal-regulated enhancer element (MRE) of the mouse metallothionein-l gene. The EMBO journal. 7, 3763-3770 Heuchel, R., et al. (1994) The transcription factor MTF-1 is essential for basal and heavy metal-induced metallothionein gene expression. The EMBO journal. 13, 2870-2875 Goldwasser, E., Jacobson, L. 0., Fried, W. and Plzak, L. (1957) Mechanism of the erythropoietic effect of cobalt. Science (New York, NY 125, 1085-1086 Goldberg, M. A., Dunning, S. P. and Bunn, H. F. (1988) Regulation of the erythropoietin gene: evidence that the oxygen sensor is a heme protein. Science (New York, NY. 242, 1412-1415 57 103 104 105 106 107 108 109 110 111 112 Ladoux, A. and Frelin, C. (1994) Cobalt stimulates the expression of vascular endothelial growth factor mRNA in rat cardiac cells. Biochemical and biophysical research communications. 204, 794-798 Yuan, Y., Hilliard, G., Ferguson, T. and Millhom, D. E. (2003) Cobalt inhibits the interaction between hypoxia-inducible factor-alpha and von Hippel-Lindau protein by direct binding to hypoxia-inducible factor- alpha. The Journal of biological chemistry. 278, 15911-15916 Salnikow, K., et al. (2004) Depletion of intracellular ascorbate by the carcinogenic metals nickel and cobalt results in the induction of hypoxic stress. The Journal of biological chemistry. 279, 40337-40344 Malard, V., et al. (2007) Global gene expression profiling in human lung cells exposed to cobalt. BMC genomics. 8, 147 Barceloux, D. G. (1999) Cobalt. Journal of toxicology. 37, 201 -206 Cobb, A. G. and Schmalzreid, T. P. (2006) The clinical significance of metal ion release from cobalt-chromium metal-on-metal hip joint arthroplasty. Proceedings of the Institution of Mechanical Engineers. 220, 385-398 Morgan, G. B., Ozolins, G. and Tabor, E. C. (1970) Air pollution surveillance systems. Science (New York, NY. 170, 289-296 Wehner, A. P., Busch, R. H., Olson, R. J. and Craig, D. K. (1977) Chronic inhalation of cobalt oxide and cigarette smoke by hamsters. American Industrial Hygiene Association journal. 38, 338-346 Kriss, J. P., Carnes, W. H. and Gross, R. T. (1955) Hypothyroidism and thyroid hyperplasia in patients treated with cobalt. Journal of the American Medical Association. 157, 117-121 Centeno, J. A., Pestaner, J. P., Mullick, F. G. and Virmani, R. (1996) An analytical comparison of cobalt cardiomyopathy and idiopathic dilated cardiomyopathy. Biological trace element research. 55, 21-30 58 113 114 115 116 Jefferson, J. A., et al. (2002) Excessive erythrocytosis, chronic mountain sickness, and serum cobalt levels. Lancet. 359, 407-408 Lauwerys, R. and Lison, D. (1994) Health risks associated with cobalt exposure--an overview. The Science of the total environment. 150, 1-6 Ciafre, S. A., et al. (2007) CoCI(2)-simulated hypoxia in skeletal muscle cell lines: Role of free radicals in gene up-regulation and induction of apoptosis. Free radical research. 41, 391-401 Karovic, 0., et al. (2007) Toxic effects of cobalt in primary cultures of mouse astrocytes. Similarities with hypoxia and role of HIF-1alpha. Biochemical pharmacology. 73, 694-708 59 Chapter 2 HlF1a ls essential for normal intrauterine differentiation of alveolar epithelium and surfactant production in the newborn lung of mice This chapter is the edited version of a research article that was published in The Journal of Biological Chemistry, Volume 283, No. 48 (33650-33657), November 28, 2008. Authors: Yogesh Saini, Jack R. Harkema and John J. LaPres. 60 Abstract Neonatal respiratory distress syndrome (RDS) is mainly the result of perturbation in surfactant production and is a common complication seen in premature infants. Normal fetal lung development and alveolar cell differentiation is regulated by a network of transcription factors. Functional loss of any of these factors will alter the developmental program and impact surfactant production and normal gas exchange. During development, the fetus is exposed to varying oxygen concentrations and must be able to quickly adapt to these changes in order to survive. Hypoxia-inducible factor 1a (HlF10t) is the primary transcription factor that is responsible for regulating the cellular response to changes in oxygen tension and is essential for normal development. Its role in lung maturation is not well defined and to address this knowledge gap, a lung-specific HlF1oc knockout model has been developed. Loss of HIF1oz early in lung development elads to pups that die within hours of parturition, exhibiting symptoms similar to RDS. Lungs from these pups display impaired alveolar epithelial differentiation and an almost complete loss of surfactant protein expression. Ultrastructural analysis of lungs from HIF1a deleted pups had high levels of glycogen, aberrant septal development, and changes in several factors necessary for proper lung development, including HlF20t, B—catenin and vascular endothelial growth factor. These results suggest that HIF1oc is essential for proper lung maturation and alteration in its normal signaling during premature delivery might explain the pathophysiology of neonatal RDS. 61 Introduction During development an embryo is exposed to varying levels of oxygen as a balance is created between vascularization and tissue growth. Localized hypoxia, a decrease in available oxygen, is a normal part of this process. The programmed responses to these decreases in available oxygen are essential for viability [1, 2]. In utero, the embryo is supplied with oxygen and nutrients through the placental barrier. Following parturition, oxygen is supplied by the neonatal lungs and therefore, proper intrauterine development of the alveolar gas exchange regions of the lung is essential for the newbom’s first breath and for sustaining life outside the womb [3]. Lung morphogenesis is a complex process that is orchestrated by several transcription factors, growth factors, and extracellular cues [4]. For example, thyroid transcription factor-1 regulates the expression of the genes for Clara cell secretory protein (CCSP) produced in Clara cells in the tracheobronchial ainrvays and various surfactants produced by alveolar type II cells in the lung parenchyma [5, 6]. CCAAT-enhancer binding protein a (CEBPa) is essential for proper regulation of alveolar Type ll cell differentiation, and Forkhead box A2 (Foxa2) controls various cellular programs involved in lung development (eg. surfactant expression) (6-8). Ablation of any of these factors results in major structural and functional abnormalities ranging from undeveloped alveolar structure and/or improper ainNay branching to the faulty processing of various secretory components [7, 8]. One of the extracellular cues that is 62 important for fetal vascular growth and lung morphogenesis is the physiologically low 02 environment of the fetus. The ability to cope with this developmental “hypoxia” is not only important for lung maturation but essential for the viability of the fetus [1 , 2]. Cellular responses to decreased oxygen availability are regulated by a family of proteins called hypoxia-inducible factors (HIFs). The ability of HIFs to respond to hypoxia is controlled by oxygen-dependent post-translational hydroxylation. The prolyl hydroxylases, PHDs, modify HIFs on essential residues in an oxygen-, iron- and a—ketoglutarate-dependent manner. Once hydroxylated, the HIF is quickly degraded in a proteasomal-dependent process that involves the Von Hippel Lindau (VHL) tumor suppressor. HIF1a, the most ubiquitously expressed HIF, has been shown to play a critical role in normal development. HIF1oc regulates the expression of genes important for cellular adaptation to hypoxia, including glycolytic enzymes and angiogenic factors [9]. Gene deletion studies have established that HIF1a is indispensable during fetal development as HlF1oc_/- mice die at midgestation, in association with defects in VEGF + _ expression and vascularization [10-12]. In addition, HIF1 I mice developed pulmonary hypertension and pulmonary vascular remodeling under hypoxic conditions [13]. PHD inhibitor-induced HIF stabilization has been shown to improve lung growth in prematurely born baboon neonates, predominantly through enhanced expression of VEGF [14]. Finally, loss of HIF2a has been 63 shown to perturb normal lung development through loss of VEGF expression and to subsequently decrease alveolar type II cell production of pulmonary surfactants [15]. Previous studies using systemic HlF1a deletions have demonstrated its central role in development and partial HlF1a deletions resulted in disrupted normal functioning of tissues such as heart and lung [13, 16, 17]. The present study was aimed at elucidating the role(s) of HIF1a in the in utero differentiation of the alveolar epithelium. To this aim, mice with a lung—specific A/A deletion of HlF1oc (termed l-llF1c1 ) were generated. Lung specific embryonic deletion of HIF1o earlier than 3 days prior to parturition led to HIF1dA/Aneonates exhibiting cyanosis and respiratory failure soon after birth. NA The expression of surfactant proteins was significantly lower in HIF1o pups as compared to litterrnate control pups. Examination of the lungs from A/A . . . . . HlF1o pups vra light and transmussron electron microscopy confirmed defects in both alveolar epithelial differentiation and septal development that resulted in the observed respiratory distress. Material and methods fl - Transgenic mice and genotyping: HlF1oc ox/flox and SP-C-rtTA ltg/(tetO)7- tg/tg CMV-Cre transgenic mice were generous gifts of Randall Johnson (UCSD) and Jefferey Whitsett (Cincinnati Children's Hospital Medical Center), 64 respectively [10, 17-19]. HlFmflox/flox mice were mated to SP-C-rtTA-ltg/ (tetO)7-CMV-Cretglt9 transgenic mice to generate SP-C-rtTA-ltg/(tetOh-CMV- fl x/fl Cretg/tg/HIFm o ox mice line capable of respiratory epithelium specific conditional recombination in the floxed HlF1oc gene. In this model, depending on the day doxycycline is administered to the darn, various cell populations fl fl within the lung undergo recombination in HIF10t ox/ ox alleles (20). Genotyping screening of the mice progeny was performed by PCR for all the three loci using previously published primer sequences (Table 1). Genomic DNA extraction from tail clipping was performed using Direct PCR extraction system (Viagen Biotech, CA) via manufacturer's instructions. PCR conditions were standardized for all the three alleles: denaturation at 94°C for 3 min; 38 cycles of denaturation at 940 for 45 sec, annealing at 60'C for 45 sec and polymerization at 72°C for 60 sec. followed by a 7 min extension at 720. Size of the amplified products obtained were approx 210 bp for HlF1a (wild type); fl x/fl 244bp for HIF1oc O 0X; 370 for Cre transgene; 350 for rtTA transgene (Fig. 1B). Doxycycline treatment and animal husbandry: Dams bearing triple (SP-C- rtTA'ltgltteto)7-CMV-Cret9/t9/HIF1JIM/fl”) and double ((tetO)7-CMV- fl x/fl Cretg/tg/HlFm 0 OX) transgenic embryos were maintained on doxycycline 65 feed (625 mg doxycycline/Kg; Harlan Teklad, Madison, WI) and drinking water (0.8 mg/ml: Sigma chemicals Co.). The day of parturition was taken as reference point to calculate the duration of doxycycline exposure. Control and NA HlF1o pups exposed to doxycycline treatment for 0 to14 Days in utero were sacked within one hour of parturition. All pups were sacrificed by decapitation. Mice used in this study were kept at the animal housing facility under the strict hygienic and pathogen free conditions approved by the university laboratory animal resource (ULAR) regulatory unit. Lungs harvesting and processing: 3-8 pups were analyzed from each genotype and doxycycline treatment. Pups were assessed for total body and lung weight. Lung tissues were isolated and left lung lobe was fixed in 10% neutral buffered formalin (NBF) or 4% glutaraldehyde for morphological analysis. The remaining lung tissue was divided; half was snap frozen for Western blotting, half was stored in RNA/ater RNA Stabilization Reagent (Qiagen) for RNA isolation and qRT—PCR analysis. Western blotting: Snap frozen lung tissue (10 mg) was lysed and homogenized in RIPA buffer (50 mM Tris HCI, 1% NP-40, 0.25% sodium deoxycholate, 150 mM NaCI, 1 mM each of EDTA, PMSF, sodium vandate, sodium fluoride, 1 M DT‘I') containing protease inhibitors (1ug/ml each of aprotinin, leupeptin, pepstatin) using a bead beater system (45 seconds at 30 Hz frequency). Insoluble material was removed by centrifugation (10,000 x g 66 for 10 min.) and protein concentration was measured in supernatant using Bradford protein assay [20]. Protein samples (30 pg) were separated by SDS- PAGE (NuPage 4-12% Bis-Tris gradient gel, lnvitrogen, CA) and transferred to a nitrocellulose membrane. Western blots were performed with rabbit or goat antibodies against surfactant associated proteins (SP-A (sc 13977, Santa Cruz, CA), SP-B (AB 3780, Chemicon), SP-C (so-13979, Santa Cruz, CA) and SP-D (SC 7709, Santa Cruz, CA), and B—actin (loading control; SC-7210, Santa Cruz, CA). Proteins were visualized with HRP-conjugated goat anti- rabbit lgG (SC-2004, Santa Cruz, CA) or rabbit anti-goat (sc-2768, Santa Cruz, CA) and ECL western blot system (Pierce, USA). Histopathology and immunohistochemistry: At least four to six pups of each genotype and doxycycline treatment were analyzed for histopathological changes. Fon'nalin fixed left lung lobe tissues were paraffin embedded and 5- micron thick sections were mounted on glass slides and stained with hematoxylin and eosin (H&E), periodic acid Schiff (PAS), or immunostained with SP-B (1: 500 dilution, A33780, Chemicon, MA), SP-C (1:4000 dilution, antibody kindly provided by Jeffrey Whitsett) and CRE (1:100 dilution, AB24608; Abcam, Cambridge, MA). Briefly, tissue sections were deparaffinized and endogenous peroxidase activity was quenched by incubation with 6% H202 for 30 min. Immunostaining was performed using Rabbit Vector Elite ABC kit (Vector Laboratories, CA), according to the manufacturer's recommendations. 67 Transmission electron microscopy: Neonatal lung sections were fixed in 4% buffered glutaraldehyde overnight at 4°C, stored in the 10% NBF fixative until postfixation with 1% phosphate-buffered osmium tetroxide. Tissues were dehydrated through a graded series of ethanol and propylene oxide, and embedded in Poly/Bed-Araldite resin (Polysciences, lnc., Warrington, PA). One pm-thick sections were cut and stained with toluidine blue for light microscopic identification of specific tissue sites for transmission electron microscopy (TEM). Ultrathin tissue sections for TEM were cut at approximately 75 nm and stained with lead citrate and uranyl acetate. Sectioning was performed with an LKB Ultratome III (LKB Instruments, Inc., Rockville, MD). Ultrastructural tissue examination and photography were performed with a JEOL JEM 1OOCXII electron microscope (JEOL Ltd., Tokyo, Japan) RNA isolation and real-time PCR analysis: 10 mg lung tissue stored in RNA/afar RNA Stabilization Reagent was homogenized in RLT buffer (RNeasy RNA isolation Kit, Qiagen, Maryland) using a Retsch MM200 bead beater system (Retsch, Haan, Germany). Total RNA quantification was performed spectrophotometrically (NanoDrop ND-1000 UV-Vis Spectrophotometer). Total RNA (1 pg) was reverse transcribed using superscript |I reverse transcriptase kit (lnvitrogen, CA). Expression level of selected genes involved in surfactant metabolism and lung development were analyzed by Real-Time PCR using SYBR green (Applied Biosystems, Foster City, CA) as previously 68 described [21]. Gene specific primers are listed Table 2. Copy number was determined by comparison with standard curves of the respective genes. This measurement was controlled for RNA quality, quantity, and RT efficiency by normalizing it to the expression level of the hypoxanthine guanine phosphoribosyl transferase (HGPRT) gene. Statistical significance was determined by use of normalized relative changes and ANOVA. Quantitative analysis: qRT-PCR analysis was performed using unpaired two- tailed Student's t-test on GraphPad Prism. Results were considered significant at the 5% level. Results Conditional inactivation of HIF1a in the lung: The HIF1a gene was inactivated in the lung by mating HIF1a conditional fl fl null mice (HIF1o ox/ ox) [10] to an inducible bitransgenic mouse, SPC-rtTA, that expresses the reverse tetracycline transactivator (rtTA) under the control of the human surfactant protein C (SP-C) promoter and the Cre recombinase gene with a tetracycline operon (Fig. 2.1A) [18]. SPC-rtTA mice have been shown to express Cre recombinase specifically in the epithelial cells of the primordial lung buds as well as in the alveolar and bronchiolar epithelium of postnatal mice in the presence of an inducer (i.e. doxycycline) [19]. In the present study, compound litters from reciprocal matings between SP-C flox/flox tg/tg -rtTA'/t9/ (tetOh-CMV-Cretg/tg/ HIF1a and (tetO)7-CMV-Cre / 69 Figure 2.1. Lung specific, doxycycline-inducible deletion of HIF1a In triple transgenic mice. (A) Triple transgenic mice were generated to induce expression of rtTA protein in the epithelial cells of the lung. rtTA expression is controlled by human SP-C promoter. Another transgene is (tetO)7-CMV-Cre recombinase transgene in which Cre recombinase expression is controlled by (tetO)7-CMV promoter in the presence of doxycycline, and rtTA protein. Cre recombinase recognizes the onP sites flanking exon 2 of HlF1a locus and facilitates homologous recombination and thus inactivation of HlF1oc locus. (B) Genotyping of transgenes. Gel shows the genotyping results for all the combinations of three transgenes. Floxed HlF1a size is 274 bp and wild type HIF1a is 240 bp. C, qRT- PCR results for HlF1a mRNA from triple transgenic mice in the NA absence (Control) and presence (HIF1o ) of doxycycline (14 day in utero exposure). Values are the average of five separate animals per group. D, Western blot analysis for HlF1a from lungs of triple transgenic mice in the absence (Control) and presence (HlF1oA/A) of doxycycline (14 day in utero exposure). Blots were also probed with a tubulin-specific antibody to demonstrate equal loading. Two independent animals from both groups are shown (1—4). "Images in this dissertation are presented in color." 70 _ <5. Ulfi LouoEoE 0mm 52:3: 38. 5E: 22:28 ownEnEooE 29:03.: <._.t-on_w: 71 Figure 2.1. Continued. BE: 25 25> 5E: .3on >080 <._.t wmo X“. X“. X”. Xu— X“. X“. Us ”:1 72 Figure 2.1 Continued. C 0.6- g 0.5- 0.4- u5:03- -.-; 0.2- o: 0.1- 0.0- p-value= 0.0091 Control I-IIF1orAIA D Control HIF1aA/A 1 2 3 4 FHF1a Tubufin 73 fl fl HlF1q ox/ ox mice were composed of pups with mixed genotypes with respect to SPC-rtTA transgene. Their genotypic ratios were in accordance with Mendelian inheritance as demonstrated by genotyping (Fig. 2.1 B). Analysis Of these ratios from litters studied (N=30 litters and 190 pups) confirmed no in utero mortalities (data not shown). In the absence of doxycycline treatment, pups were phenotypically indistinguishable between litters. The functionality of the doxycyline treatment was demonstrated by the significant decrease in HlF1o mRNA as measured by qRT-PCR (Fig. 2.10) and loss of HIF1a protein by Western blot analysis (Fig. 2.1 D) Lung-specific deletion of HIF1a causes lethal phenotype: To determine if in utero loss of HlF1a influenced litter size or the Mendelian distribution of pups, dams carrying triple transgenic embryos were exposed to doxycyline through feed and water for various lengths of time prior to parturition (i.e. 2, 4, 6, 8, 10, 12, and 14 days). Our findings indicate pups exposed to doxycyline in utero for more than 8 days prior to parturition had a 100% mortality rate, while those receiving the drug for 4-6 days prior to parturition had approximately a 15% chance of survival (Fig. 2.2A). Following parturition, A HlF1qA/ pups [doxycyline-induced] developed respiratory distress, severe cyanosis, and died within an hour of birth (range of 10-60 minutes) (Fig. 2.23). NA At necropsy, the lungs from HlF1o pups were dark red and atelectic, in contrast to pink, aerated lungs from control (in the absence of doxycycline treatment) pups. Excised lung lobes from HlF1ch/A pups weighed slightly 74 more than the lungs from control mice and sank to the bottom of a saline-filled vial in contrast to the langs from control litterrnates which floated in saline (Figs. 2.20 and 2.2D). At birth, the average total body weights of control litterrnate and HIF1aA/A were 1.318 :I: 0.018 (SEM) and 1.288 :I: 0.024 grams, respectively. Defective lung morphology: The lungs of HlF1ch/A pups had histologic and ultrastructural features that indicate an impairment of normal in utero differentiation of alveolar epithelium, with a related loss of lung surfactant proteins that are essential for preventing collapse of individual alveoli and ensuring proper gas exchange to maintain life. Lungs of HIF1aA/A pups at birth had abnormally thickened alveolar septa (Fig 2.3C) lined by undifferentiated cuboidal epithelial cells (immature pneumocytes) containing large amounts of intracytoplasmic periodic acid Schiff(PAS)-positive material (Fig 2.3D) that was ultrastructurally identified, via transmission electron microscopy, as glycogen (Figs. 2.40 and 2.4D). There was a marked reduction of alveolar airspaces in the lungs of these mice due in part to the thickened septa and to focal areas of atelectasis. In contrast, the lungs of control mice at birth had uniformly dilated alveolar airspaces with much thinner alveolar septa that were lined by more differentiated surface epithelium containing much less PAS-positive glycogen (Figs. 2.3A and 2.3B). 75 Figure 2.2. Effects of lung specific deletion of HlF1o . Graph of the percent viability of pups at birth versus intrauterine doxycycline treatment days. Doxycycline-induced lethality was more pronounced when the drug was delivered more than 4 days before parturition (circles). No lethality was observed in control group (no doxycyline, squares) or other possible genotype controls (data not _ A/A . . shown). (B) At birth HIF1d pups were cyanotlc (nght) compared to normal, pink well oxygenated controls (left). (C) Mean lung weights of NA control (black bar) and HlF1o pups (white bar). The asterisk indicates a significant difference (P value < 0.05) based on a two-tailed ttest for samples of unequal variance. The P value is indicated. (D) Vial containing excised lungs from newborn pups. Lungs from A HIF10N (A/A) pups sink whereas control (Ctrl) lungs float in saline. "Images in this dissertation are presented in color." 76 Viability (%) 120 100 80— 60- 4o- 20- * 777 #7 i —0— Control l g+ Doxy , e e e e *5_, - l l 1' Hr Treatment Days A 1 Control HIFltlA/A 77 Figure 2.2. Continued. Control HIF1 aA/A Control -> Hll=1oiAIA 9 78 Figure 2.3. Pulmonary Histopathology of Lungs. Light photomicrographs of hematoxylin and eosin stained lung sections from control (A) and HlF1aA/A (C) pups. Lung from HIF1otA/A pup has less alveolar airspace (a) and thicker alveolar septa (S) compared to control. Large cuboidal epithelial cells line the alveolar septa of A HIF1oA/ pup while the alveolar septa of control pup is lined primarily by squamous epithelial cells (type 1) and only a few widely scattered cuboidal cells (type II). Periodic acid Schiff (PAS) stained lung sections from control (B) and HIF1aA/A (D) pups. Cuboidal epithelium lining A/ alveolar septa in HIF1a A pup has greater PAS-stained glycogen (arrows) than that of the control pup (D). "Images in this dissertation are presented in color." 79 .9550 80 Figure 2.4. Electron photomicrographs of alveolar epithelium of newborn pups. Lung from control pup (A, B) has a well differentiated alveolar epithelium containing both type I and II cells (B). Stipled arrow indicates thin, squamoid, type I cells and solid arrows indicate tubular myelin figures (secreted surfactant) within the alveolar airspace of control pup. Type II cells have numerous intracytoplasmic lamellar bodies (lb) and NA apical microvilli (mv). In contrast the alveolar surface in HlF1o pup (C, D) is lined by large undifferentiated cuboidal epithelial cells The cytoplasm of these epithelial cells are primarily filled with glycogen (it). No lamellar bodies are present in these cells. C, Capillary; rbc, red blood cells; ll, type II alveolar epithelial cell; I, type I alveolar epithelial cell; if, interstitial fibroblast; *, glycogen; n, nucleus; 1, mitochondrion; 2, rough endoplasmic reticulum; pl, pleural surface; solid black arrows, tubular myelin; stipled arrow, type I cells. ”Images in this dissertation are presented in color." 81 82 Figure 2.4 Continued. 83 Ultrastructurally the luminal epithelium lining the alveolar airspace of control pups consisted of both alveolar type I and II cells (differentiated pneumocytes). Distinctive lamellar bodies were present in the apical cytoplasm of alveolar type II cells along with tubular myelin and secreted lamellar material (ultrastructural features of secreted surfactant) widely scattered along the alveolar luminal surface (Figs. 2.4A and 2.4B). These distinctive cuboidal epithelial cells also had a round to ovoid nucleus, an apical luminal surface lined by short microvilli, and numerous cytoplasmic organelles consisting of mitochondria, rough endoplasmic reticulum, and lysosomes. In contrast, Type I cells were identified by their thin squamous morphology containing few organelles and a fusifonn nucleus (Figs. 2.4A and 2.48). Interestingly, there were no microscopically detectable differences in the morphology of intrapulmonary conducting ainlvays (preterminal and terminal bronchioles), lined principally by ciliated cells and nonciliated cuboidal (Clara) cells, between control and HIF1dA’A pups at birth. Altered surfactant metabolism: qRT-PCR analysis of the genes for surfactant proteins show a decreased expression of SP-A, SP-B, and SP-C in the HlF1r1A/A mice compared to control pups (Fig. 2.5A). SP-D mRNA expression was not significantly different between the two genotypes. 84 Figure 2.5. Surfactant protein expression. (A) mRNA levels for Surfactant associated proteins (SP-A, SP-B, SP-C and SP-D). Each expression level is expressed relative to HGPRT. (B) lmmunoblot analysis of surfactant associated proteins (SP-A, SP-B, SP-C, and SP-D) in lung homogenates from control and HIF1aA/A pups (n=3). [3- actin (actin) was used as loading control. (C) lmmunohistochemical staining of SP-B (left panels) and SP-C (right panels) in lung sections from control (upper panels) and HIF‘IGA/A (lower panels) pups. HIF1oA/A pups show decreased expression of SP-B and SP-C as compared to control littermates. The asterisk indicates a significant difference (P value < 0.05) based on a two-tailed ttest for samples of unequal variance. The P values are indicated. "Images in this dissertation are presented in color." 85 O '3 2 fl' |.I.l E E at" Relative Expression (HQ 0 40 ‘8'!“ 0°00 Control Control SPA SPB Acti n SPC SPD * HIF1aA A SPC Control Relative Expression HIF1aA’A Relative Expression Control HIF1aA’A SPD T Control HIF1<1AIA Hll=1'o,AIA Acti n 86 Figure 2.5. Continued. 87 A/A Western blot analysis of protein extract from HIF1a and control animals was also performed to determine if surfactant protein levels were similar to gene expression patterns. All four surfactants displayed a substantial A/A reduction in protein levels in the HlF1d mice as compared to control pups (Fig. 2.5B). lmmunohistochemical staining on lung sections confirmed the NA reduced levels of SP-B and SP-C in the HIF1q compared to control pups (Fig. 25C). The reduced levels of surfactant mRNA and protein expression were consistent with the lack of alveolar epithelial differentiation and NA respiratory distress in the HlF1o pups. Altered Gene expression: To begin to understand how loss of HIF10t induced changes in lung morphology, qRT-PCR was performed on several critical genes known to be involved in lung development. These genes include B-catenin, clara cell secretory protein (CCSP), forkhead box A2 (Foxa2), forkhead box protein A1 (Foxa1, also known as HNF3o), thyroid transcription factor 1 (TTF-1), GATA binding protein 6 (Gata6), ATP-binding cassette, sub- family A (ABC1 member 3 (ABCA3), and CCAAT/enhancer binding protein alpha (CEBPa). There were no significant differences in the expression levels of Foxa2, ABCA3, CEBPo, ‘l‘l'F-1, Foxa1, or GATA, suggesting that HIF10t is not required for their normal expression and is most likely involved in later events in lung development (Fig. 2.6). However, lower levels of B-catenin, 88 figure Quanlll various express a slgnll sample Figure 2.6. Expression of developmentally important genes. Quantitative real time PCR was used to measure the mRNA levels of various genes known to play a role in lung development. Each expression level is expressed relative to HGPRT. The asterisk indicates a significant difference (P value < 0.05) based on a two-tailed ttest for samples of unequal variance. The P values are indicated. 89 Relative Expression I“ 9' .“ 0| 0 0| 0. Control pro bio Relative Expression HIMF HlF1aA/A VEGF 99 O Control Control Control HlF1Ex “4 HIFZG * TTF HIF1a A’A Hing,“ _.1_ Control HIF1aA’A B-catenin C .2 In In 0 I- 2 I: 0.10 g 0.05 * O n: 0.00 Control HIF1aA/A CCSP C .2 Z“: 750 h D. ”’5 500 * 2 I E 250 O m I Control HIF1aA’A FoxA2 C '2 0.03 i i- 0.02 2 I I.“ .‘z’ E d) n: 90 Relative Expression 9 .° P O O O O A N CEBPa Control HIF121A’A CCSP, HIMF, VEGF and HlF2q mRNA in neonatal HIF1c1A/A pups compared to control littermates suggest that HIF10t plays a role in controlling their expression during maturation of the lung [15, 22]. Given the established role of these factors in regulating lung development, our data suggest that HIF10t is centrally located in the transcriptional network that is necessary for proper lung morphogenesis. The previously described role of HIF20t in lung development, the similarity in phenotype between the HlF20t knockout survivors and HlF10t lung deletion strains, and the loss of HlF20t expression in the l--llF10lA/A pups led us to determine the level of HlF20t expression at the protein level using immunohistochemistry [15]. In the presence of doxycyline there was a pronounced expression of Cre recombinase in all cell types of the lung (Fig. 2.7D). There was a corresponding loss in HlF10t expression in the alveolar tissue (Fig. 2.7E). Though HIF10t is not expressed ubiquitously throughout the lung, its cell specific expression was abrogated upon doxycycline treatment in the triple transgenic animal (Fig. 2.7). Surprisingly, the increased Cre recombinase and subsequent loss of HlF10t expression also led to a drastic decline in HIF20t levels in the lung (Figs. 2.70 and 2.7F). 91 Figure 2.7. Immunohistochemistry for Cre recombinase, HlF10l, and HIF20t. Lung sections from control (A,B,C) and HlF10tA/A pups (D,E,F) were immunostained for Cre recombinase (A,D), HlF1ot (BE) and HlF20t (C,F). Immunohistochemistry for Cre recombinase shows strong staining of HIF10tA/A mouse alveolar epithelial cells (D, brown color). Positively stained cells for HlFtot and HIF20t are depicted by solid arrows. HIF10tA/A pups show decreased expression of HIF10t and HIF20t as compared to control littermates. a, air space; p, pleural surface; s, septa. "Images in this dissertation are presented in color." 92 Cre Recomblnue 93 Table 2.1: Primers for PCR-genotyping. Allele Forward Primer (5’-3’) Reverse Primer (5’-3’) HIF 1a gca gtt aag agc act agt tg gga gct atc tct cta gac c Cre tgc cac gac caa gtg aca gca atg aga gac gga aat cca tcg etc 9 rtTA gac aca tat aag acc ctg gtc a aaa atc ttg cca gct ttc ccc Table 2.2: Primers for real-time RT-PCR. Gene Forward Primer (5’-3’) Reverse Primer (5’-3’) SPA gct gtc agt ggg gga taa ag ctt tgt aat gct tgc gat gg SPB ctt gtc ctc (Ea tgt tcc ac ggc ctggfigat cac aga ct SPC cagctc cag gaa cct act 90 got tag agg tgg gtg tgg ag SPD gag gtt_gcc ttc tcc cac ta agc ctg ttt gca cat ctc ct HIF1-a t ctc cct ata tcc caa tg ggt ctg ctg lea ccc agt aa HIF2-a gag caa gcc ttc caa gac ac ttc gca ctg atg gtc ttg to B- ggc agc agc agt ctt act tg aag gac tgg gaa aag cct tg catenin VEGF tca cca agg cca gca cat ag aat gct ttc tcc gct cgaa CCSP cat cat gaa gct cac gga ga ggg aca cag ggc agtgac aa FoxA2 gcc ag_c_gag tta aggtat gc tca tgt tgc tca cgg aag ag HNF 3- ccc ttt ctc cct ttc act cc gtg gtg ggc cta aca aca ac a TTF-1 gct ggc ctt tca gaa aat tg gga cta ggg act mad gg CEBPa gca agc cag gac tag gag at c g aaa gtc tct cgg tct ca HGPRT aag cct aag atg agc gca ag tta cta ggc aga tgg cca ca PPI-A agc ata cag gtc ctg gca tc ttc acc ttc cca aag acc ac ABCA3 gagctt tgc cca cct aca ac gaa act ggg agg gag agg ac GATA caa aag ctt gct ccg gta ac ctg agg tgg tcg ctt gtg ta 6 Retnla tat gaa cag atg ggc ctc ct ggt cca gtc aac gag taa gca 94 Discussion Mammalian lung development and successful transition to extrauteline respiration involves a wide battery of biochemical, cellular and ultrastructural changes. This complex process begins at embryonic day E9.5 with most of the alveolar epithelial differentiation events starting at embryonic day E17.5. Several transcription factors control the temporal and positional epithelial differentiation process, as well as, the function of these differentiated cells. When the function of these critical factors is disrupted during development, lung function is compromised, leading to respiratory distress at birth. Our results have added HlF1oc to this list of transcription factors that are critical for lung development. We have also demonstrated that loss of HIF10t during development leads to changes in the expression of other critical factors, such as HlF20t, VEGF and 0t-catenin [23]. HlFs are a class of transcription factors that play a critical role in oxygen sensing and the metabolic adaptations to hypoxia. Previous studies have shown that HIF1a levels are predominant during lung development until day E135, at which time, HlF20t becomes the major HIF in this tissue [24, 25]. Targeted gene disruption of the HIF1a locus results in the embryonic lethality due to cardiovascular defects [12]. Mice partially deficient for HIF1o alleles showed normal development but were physiologically compromised as they exhibited impaired pulmonary vascular remodeling under chronic hypoxic stress. In addition, the heterozygote was used to show that partial loss of 95 HlF1o alleles results in impaired pulmonary arterial myocyte electrophysiological responses under hypoxic stress [13, 25, 26]. The study presented here has shown that the lung-specific loss of HIF10t leads to neonatal respiratory distress syndrome due to impaired alveolar epithelial A/A differentiation. Interestingly, the lungs of HlF1o pups had normal morphogenesis of conducting airways with phenotypic alterations restricted to the alveolar parenchyma. These results suggest that HlF1ot is involved in the late events of lung morphogenesis and alveolar epithelial differentiation but not in the early lung biogenesis and airway branching morphogenesis. Respiratory distress syndrome and associated histopathology are phenotypic hallmarks of targeted deletion of several transcription factors, such as Foxa2, CEBPa, GATA-6, SMAD3, Calcineurin b1, and thyroid transcription factor-1, suggesting an intricate network of transcriptional regulation governing the complex ultrastructural maturation of lungs [8, 27-30]. Expression analysis demonstrated that the levels of Foxa2, Foxa1, CEBPa, and GATA-6 are comparable among pups with and without HIF10t (Fig. 2.6). This suggests that HIF’IG acts either downstream or independent of these transcription factors. In contrast, the loss of HIF10t led to a decreased expression of other critical factors, including CCSP, B-catenin, HIMF, VEGF, and HlF20t (Fig. 2.6). Recently, the loss of HlF20t and subsequent decrease in VEGF has been linked to respiratory distress syndrome [15]. These experiments involved a ./- subset of HIF20t mice that were viable up to parturition and displayed similar 96 pathology to the HIF10tA/A mice described here. Delivery of VEGF, either ./- intrauterine or postnatal intratracheal instillation protected the HlF20t mice from symptoms of respiratory distress syndrome. Given that HIF20t ' expression is abrogated in the HIF1 (IA/A pups suggests that HIF10t regulates HlF20t early in lung development and the subsequent loss of VEGF might explain the pathology of the HIF‘ICXA/A pups. It remains to be seen if the decrease in VEGF expression is due to direct loss of HlF10t or indirectly through the loss of HlF20t expression. More importantly, the pathophysiology of the HlF1aA/A mouse resembles that of the HIFZG-I- mouse in relation to the lack of differentiation of type II pneumocytes and increased septal thickness. A/A In addition, the HlF1o pups display large glycogen stores with concurrent loss of surfactant protein expression. In conclusion, these studies provide evidence that HIF10t plays an important role in lung development. The respiratory failure induced by lung- sDecific deletion of HlF1o, suggests that hypoxia and HIF10t-regulated genes play an essential role in lung maturation, especially proper differentiation of tElbe ll alveolar cells. HlF10t’s potential upstream regulatory function of HlF20t and VEGF expression, directly or indirectly, offers a mechanism of action for the phenotypic observations. Finally, the results suggest that the hypoxia Signaling cascade is important during the later stages of lung development but 97 not essential in the initial patterning of the tissue. HlFs seem to play a later roIe in directing cell differentiation and regulating growth factors necessary for lung remodeling required for sustaining life extrauterine. Understanding the pathways necessary for proper HIF expression and function and the downstream pathways influenced by these factors will impact our ability to treat RDS and other diseases of the lung. 98 RE FERENCES 1 Webster, W. S. and Abela, D. (2007) The effect of hypoxia in development. Birth Defects Res C Embryo Today. 81, 215-228 2 Giaccia, A. J., Simon, M. C. and Johnson, R. (2004) The biology of hypoxia: the role of oxygen sensing in development, normal function, and disease. Genes Dev. 18, 2183-2194 3 James Metcalfe, H. B., Waldemar Moll. (1967) Gas Exchange in the Pregnant Uterus. Physiol. Rev. 47, 782-838. 4 Groenman, F., Unger, S. and Post, M. (2005) The molecular basis for abnormal human lung development. Biology of the Neonate. 87, 164- 177 5 Cassel, T. N., Suske, G. and Nord, M. (2000) C/EBP alpha and TTF-1 synergistically transactivate the Clara cell secretory protein gene. Annals of the New York Academy of Sciences. 923, 300-302 6 deFelice, M., et al. (2003) TTF-1 phosphorylation is required for peripheral lung morphogenesis, perinatal survival, and tissue-specific gene expression. Journal of Biological Chemistry. 278, 35574-35583 7 Mucenski, M. L., et al. (2003) beta-Catenin is required for specification of proximal/distal cell fate during lung morphogenesis. J Biol Chem 278, 40231-40238 8 Wan, H. J., et al. (2004) Foxa2 is required for transition to air breathing at birth. Proceedings of the National Academy of Sciences of the United States of America. 101, 14449-14454 Semenza, G. L. (2000) HIF-1: mediator of physiological and pathophysiological responses to hypoxia. Journal of Applied Physiology. 88, 1474-1480 99 1O 11 12 13 14 15 16 17 18 79 Ryan, H. E., Lo, J. and Johnson, R. S. (1998) HIF-1 alpha is required for solid tumor formation and embryonic vascularization. Embo Journal. 17, 3005-3015 Kotch, L. E., lyer, N. V., Laughner, E. and Semenza, G. L. (1999) Defective vascularization of HIF-1 alpha-null embryos is not associated with VEGF deficiency but with mesenchymal cell death. Developmental Biology. 209, 254-267 lyer, N. V., et al. (1998) Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor 1 alpha. Genes & Development. 12, 149-162 Yu, A. Y., et al. (1999) Impaired physiological responses to chronic hypoxia in mice partially deficient for hypoxia-inducible factor 1alpha. J Clin Invest. 103, 691-696 Asikainen, T. M., et al. (2006) Improved lung growth and function through hypoxia-inducible factor in primate chronic lung disease of prematurity. Faseb Journal. 20, (E986-E994) 1698-1700 Compemolle, V., et al. (2002) Loss of HIF-2alpha and inhibition of VEGF impair fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice. Nat Med. 8, 702- 710 Tomita, S., et al. (2003) Defective brain development in mice lacking the Hif-1 alpha gene in neural cells. Mol Cell Biol. 23, 6739-6749 Ryan, H. E., et al. (2000) Hypoxia-inducible factor-1 alpha is a positive factor in solid tumor growth. Cancer Research. 60, 4010-4015 Perl, A. K., et al. (2002) Early restriction of peripheral and proximal cell lineages during formation of the lung. Proc Natl Acad Sci U S A. 99, 10482-10487 Lobe, C. G., et al. (1999) Z/AP, a double reporter for ore-mediated recombination. Dev Biol. 208, 281-292 100 20 21 22 23 24 25 26 27 28 Bradford, M. M. (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Analytical biochemistry. 72, 248-254 Vengellur, A. and LaPres, J. J. (2004) The role of hypoxia inducible factor 1 alpha in cobalt chloride induced cell death in mouse embryonic fibroblasts. Toxicological Sciences. 82, 638-646 Mucenski, M. L., et al. (2003) beta-catenin is required for specification of proximal/distal cell fate during lung morphogenesis. Journal of Biological Chemistry. 278, 40231-40238 Maeda, Y., Dave, V. and Whitsett, J. A. (2007) Transcriptional control of lung morphogenesis. Physiological reviews. 87, 219-244 Ema, M., et al. (1997) A novel bHLH-PAS factor with close sequence similarity to hypoxia-inducible factor 1alpha regulates the VEGF expression and is potentially involved in lung and vascular development. Proc Natl Acad Sci U S A. 94, 4273-4278 Shimoda, L. A., et al. (2001) Partial HIF-1 alpha deficiency impairs pulmonary arterial myocyte electrophysiological responses to hypoxia. American Journal of Physiology-Lung Cellular and Molecular Physiology. 281, L202-L208 Semenza, G. L. (2005) Pulmonary vascular responses to chronic hypoxia mediated by hypoxia-inducible factor 1. Proceedings of the American Thoracic Society. 2, 68-70 Dave, V., et al. (2006) Calcineurin/Nfat signaling is required for perinatal lung maturation and function. The Journal of clinical investigation. 116, 2597-2609 Devriendt, K., Vanhole, C., Matthijs, G. and de Zegher, F. (1998) Deletion of thyroid transcription factor-1 gene in an infant with neonatal thyroid dysfunction and respiratory failure. New England Journal of Medicine. 338, 1317-1318 101 29 30 Liu, C., Morrisey, E. E. and Whitsett, J. A. (2002) GATA-6 is required for maturation of the lung in late gestation. American Journal of Physiology-Lung Cellular and Molecular Physiology. 283, L468-L475 Martis, P. C., et al. (2006) C/EBP alpha is required for lung maturation at birth. Development. 133, 1 155-1164 102 Chapter 3 Loss of HIF20l rescues the HIF10t deletion phenotype of neonatal respiratory distress in mice 103 Abstract Hypoxia is defined as a state of decreased oxygen reaching the tissues of the body. During normal prenatal development, the fetus is exposed to hypoxic conditions and the fetus’ response to these localized occurrences of oxygen deprivation is essential for proper organogenesis and survival. The mammalian response to these decreases in oxygen availability is regulated by the hypoxia-inducible factors (HIFs). HlFs are transcription factors responsible for regulating the cellular adaptation to hypoxia through direct modulation of key genes involved in glycolysis, angiogenesis, and erythropoiesis. HlF10t and HIF20l, two key isoforms, are important in embryonic development, however; their role in lung maturation is not well defined. We have recently shown that the loss of HIF1oc early in lung development leads to pups that die within an hour of parturition, exhibiting symptoms similar to neonatal respiratory distress syndrome (RDS). To further investigate the independent role of HlF20t and its ability to alter HlF10t-mediated lung maturation, we generated two additional lung-specific HIF0t knockout models (HIF20t and HlF10t+HlF20t). Our current study has shown that the intrauterine loss of HlF20t does not display signs of RDS. More interestingly, survivability observed after the loss HIF10t and HlF20t suggests that the loss of HIF20t is capable of rescuing the neonatal RDS phenotype seen in HIF10t deficient pups. Microarray analyses on the total mRNA from these three genotypes have identified several factors, such as MMP-9, Acct-9, CDS-1, and lL1R-ll, 104 that are differentially regulated by the two isoforms of HlFa. These results suggest that HlFor isoforrns are involved in lung maturation and differentiation and further understanding of mechanisms underlying the observed phenotype might explain the pathophysiology of neonatal RDS. 105 Introduction During gestation, the oxygen needs of the developing fetus are met via placental gas exchange. Developmental defects specific to the lung fail to manifest until the parturition stage, when placental roles of oxygen and nutrient supply are taken over by the neonate. Thus, proper intrauterine development of the alveolar gas exchange regions of the lung is essential for the newbom’s first breath and for sustaining life outside the womb [1]. Lung development, similar to all the other tissues in the fetus, is a highly coordinated series of events involving complex intracellular and extracellular signals that control transcriptional programs leading to proper cellular behavior and morphogenesis. Lung morphogenesis and alveoli formation processes are regulated through the temporo-spatial expression of a plethora of transcription factors, growth regulators, and environmental signals [2]. Proteins, such as TTF-1, TGF-B, CEBPot, GATA-6, CEBP-a, KLF-5, Foxa1, and Foxa2 have been shown to be critical for proper lung development [3-9]. Alveolar epithelium- specific ablation of these and several other genes compromise lung function and perinatal survivability [6, 10]. Most of these factors function to transduce extracellular signals into developmental outcomes. One important extracellular signal is oxygen availability. A decrease in available oxygen, referred to as hypoxia, is a natural occurring condition within a fetus. Sensing 106 hypoxia is essential for proper development and mammals use a family of proteins called, the hypoxia inducible factors (HlFs) to perform this task. There are three cytosolic HlFs, HIF10t,HlF20t, HIF30t, and each is oxygen labile. In the presence of adequate oxygen, these HIFs are modified by prolyl hydroxylases(PHDs), in an oxygen-, iron- and oc-ketoglutarate- dependent manner. Upon hydroxylation, the cytosolic HIF becomes a substrate for ubiquitination by the Von Hippel Lindau (VHL) tumor suppressor and thereafter, HIF0t is proteasomaly degraded. Under the conditions of oxygen deprivation, PHD activity is inhibited leading to the stabilization and translocation of HIch into the nucleus and subsequent dimerization with the aryl hydrocarbon nuclear translocator (ARNT, also known as HIF1B). The active dimer binds to the hypoxia responsive element sequence in the promoter region of hypoxia responsive genes [11]. HIF target genes have been shown to play important roles in processes such as glycolysis, angiogenesis, erythropoiesis and development. Studies have suggested that the low oxygen environment in the developing lung is required for HIF-regulated pathways that are essential for proper organogenesis [12, 13]. For example, mice lacking HlF10t, the most ubiquitously expressed HIF, display disorganized yolk sac vascularization and cardiac defects [14]. HIF10t-deficient embryos also exhibited only a few capillaries in the neural fold with complete absence of vascular network and 107 + _ also reduced size of dorsal aorta [15]. In addition, HIF1 I mice developed pulmonary hypertension and pulmonary vascular remodeling under hypoxic conditions [16]. Similarly, homozygous deficiency of HlF20t in murine embryo results in vascular disorganization in yolk sac and failure to assemble and maintain vascular tubular structure and leading eventually to death between day E95 and E13.5 [17, 18]. The defective phenotype of HIF10t'l. were further aggravated by deleting either or both alleles of HlF20t. HlF10t./-/20t'/' mice showed absence of placental vasculature, increase in trophoblastic giant cells, poor invasive efficiency and defective placental structure thus representing phenocopy of ARNT deficient mice [13]. The mid-gestational lethality of the systemic knockout models has made it challenging to study the role of different HIF proteins in fetal lung development. Current understanding of the role of HIF signaling in the lung development is limited. It has been shown that the PHD inhibitor, FG-4095, can improve lung growth in prematurely born baboon neonates, predominantly through HIF-mediated enhanced expression of VEGF [19]. In addition, some of the progeny from HIFZa-l- mice within the isogenic 129S6/SvaTac strain survive to parturition and die shortly after birth from a respiratory distress syndrome-like pathology. The cause of this was linked to loss of VEGF expression that resulted in pulmonary surfactant deficiency [20]. Finally, lung- 108 specific knockout mice demonstrated that HIF10t is indispensable for lung development and is an important factor in surfactant production and cellular differentiation [21]. Moreover, the lung-specific deletion of HlF10t alters the expression of factors with putative roles in lung development, including HlF20t. The collective role of HIF10t and HIF20t in lung development, their place in the transcriptional network necessary for proper cellular differentiation, and their potential redundant function in lung development are still unclear. To increase our understanding of the role of these two HlFs in lung development, mice with a lung-specific deletion of HlF1oc (termed HlF1otA/A), HIF20 (termed HIFZCNA), and both HIF10t and HIF20 (termed HIF1/2qA/A) were generated. These studies confirmed that HIF10t is essential for proper lung development. Interestingly, the lung specific deletion of HlF20t did not confirm previous published reports and displayed no lethality upon parturition. More importantly, simultaneous deletion of both isoforrns of HIF rescued the respiratory distressed phenotype that was observed after HIF10tdeletion. In addition, a battery of genes has been identified that might be related to the described phenotype in each of the genotypes. These results demonstrate a non-redundant function for HIF10t and HIF20t in lung development and establish hypoxia and HIF-mediated signaling pathways that are altered upon their manipulation. 109 Material and methods Transgenic mice and genotyping: HIF1qflox’flox, HlF2<1floxm0x and SPC- rtTA-ltg/ (tetOh-CMV-Cretg/tg transgenic mice were generous gifts from Randall Johnson (UCSD), M. Celeste Simon (University of Pennsylvania) and Jeffrey Whitsett (Cincinnati Children’s Hospital Medical Center), respectively [22-26]. In the present study, we used these three transgenic mice to generate three genotypically different mice lines, SPC-rtTA'ltg/(tetOh-CMV- floxfflox Cretg/ t9/Hll=1or “mm”, ,sPc-rtTA'ltg/(teto)y-CMV-Cretg/tg/Hli=2o and -/tg tg/tg flox/flox floxlflox SPC-rtTA /(tetO)7-CMV-Cre /HIF1o /HIF20 [21]. All of these three mice lines are capable of respiratory epithelium specific conditional recombination in the floxed HIFCI alleles (Fig. 3.1). In this model, depending upon the day doxycycline is administered to the darn, various cell populations within the lung undergo recombination in the floxed alleles [24]. Genotyping screening of the mice progeny was performed by PCR for all the three loci using previously published primer sequences. Genomic DNA extraction from tail clipping was performed using Direct PCR extraction system (Viagen Biotech, CA) via manufacturer’s instructions. PCR conditions were standardized for all the three alleles: denaturation at 94°C for 3 min; 38 cycles of denaturation at 94°C for 45 sec, annealing at 60°C for 45 sec and polymerization at 72°C for 60 sec. followed by a 7 min extension at 72°C. Size of the amplified products obtained were approx 210 bp for HlF1o (wild type); 110 fl fl 244 bp for HlF1o °XI °x; 410 bp for HlF2o (wild type); 444bp for HlFZGflox/flox; 370 for Cre transgene; 350 for rtTA transgene (Fig. 3.2). Each mouse line was maintained in a mixed C57BL/6zFVB background. Doxycycline treatment and animal husbandry: Triple and quadruple transgenic pups were exposed to doxycycline in utero to generate lung- specific HIF-deficient pups. Dams were maintained on the doxycycline feed (625 mg doxycycline/Kg; Harlan Teklad, Madison, WI) and drinking water (0.8 mg/ml: Sigma chemicals Co.) from day E4.5 until day PN1. All the HIF- deficient pups born from these dams will be referred to as HIF1qA/A, HlF2aA/A or HIF1oA’A/HIF20A/A. Triple and quadruple transgenic mice that were not exposed to doxycycline will be referred to as controls. All the animal handling and necropsy protocols were approved by the ULAR regulatory unit of Michigan State University. Mice used in this study were kept at the animal housing facility under the strict hygienic and pathogen free conditions approved by the university laboratory animal resource (ULAR) regulatory unit. A/ Lungs harvesting and processing: Control and HlF1o A, HIFZCIA/A o I' HlF1c1A/A/HIF20A/A pups were either sacrificed within one hour of parturition 111 Figure 3.1. Lung specific, doxycycline-inducible deletion of HIF10t and HIFZot in triple transgenic mice. Triple transgenic mice were generated to induce expression of rtTA protein in the epithelial cells of the lung. rtTA expression is controlled by the human SP-C promoter. Another transgene is (tetO)7-CMV-Cre recombinase transgene in which Cre recombinase expression is controlled by (tetO)7-CMV promoter. In the presence of doxycycline, the rtTA protein is produced and activates the expression of the Cre-recombinase. Cre recombinase recognizes the loxP sites flanking exon 2 of the HIF10t and/or HlF20t locus and facilitates homologous recombination and thus inactivation of respective HlFa locus. “Images in this dissertation are presented in color". 112 ZOCMAwD 4u>xoo (ht <._.t Luna—hash. Utm: o... C u HIIIIHH—LIIJ $5.63: 113 Figure 3.2. Genotyping of transgenes. Gel shows the PCR genotyping results for all the combinations of four transgenes. Sizes of the amplified products obtained are: 240 bp for . flox/flox . HlF1a (Wild type); 274 bp for HlF1q ; 410 bp for HlF2d (Wild type); 444bp for HIFZGflOXIflOX; 370 for Cre transgene; 350 for rtTA transgene. One representative sample from each of three generated mice (SPC-rtTA- ltg/(tetOh-Cre-Itg/ H | F1 aflm (Lane 1 ), SPC-rtTA-Itg/(tetOh-Cre- ltg/lezolfl/fl mouse (Lane 2), and SPC-rtTA-ltg/(tetOh -Cre' It fl/fl g/HIF10t/20t (Lane 3)) was genotyped for the four transgenes. 114 115 HlF10t HlF20t CRE SPC or at day PN60 (HIFZGNA or HIF1ch/A/HIF20A/A pups only). All neonatal pups were sacrificed by decapitation and lung tissue was harvested as described previously [21]. Briefly, the adult mice were anesthetized with sodium pentobarbital (50 mg/ml), and a midline Iaparotomy was performed to canulate the trachea. The lung and heart were removed en bloc. The left lobe was fixed via perfusion inflation method with 10% neutral buffered formalin for histopathological analysis. The remaining lung lobes were divided: half were stored in RNA/ater RNA stabilizing reagent (Qiagen, Valencia, CA) for RNA isolation and qRT-PCR analysis, and half were snap frozen for protein extraction. Histopathology and immunohistochemistry: At least four to six pups of each genotype and doxycycline treatment were analyzed for histopathological changes. Formalin fixed left lung lobe tissues were paraffin embedded and 5- micron thick sections were mounted on glass slides and stained with hematoxylin and eosin (H&E), periodic acid Schiff (PAS), or immunostained with HIF10t (1:250 dilution, NB100-479, Novus Biologicals, CO) and HlF20t (1:150 dilution, NB100-122, Novus Biologicals, CO). Briefly, tissue sections were deparaffinized and endogenous peroxidase activity was quenched by incubation with 6% H202 for 30 min. Immunostaining was performed using Rabbit Vector Elite ABC kit (Vector Laboratories, CA), according to the manufacturer's recommendations. 116 RNA Isolation Lung tissue (10 mg) stored in RNAIater RNA Stabilization Reagent was homogenized in RLT buffer (RNeasy RNA isolation Kit, Qiagen, Maryland) using a Retsch MM200 bead beater system (Retsch, Haan, Germany). Total RNA quantification was performed spectrophotometrically (NanoDrop ND- 1000 UV-Vis Spectrophotometer). Isolated RNA was resuspended in RNAase free water, quantified (A250), and concentration was calculated by spectrophotometric methods (A250). Purity was assessed by the A260:A280 ratio and by visual inspection of 3 pg on a denaturing gel. Microarray Experimental Design The lung RNA samples extracted from HlF1oA/A, HIFZCNA or HIF1/2(1A/A and control animals were individually hybridized to 4 x 44K whole mouse genome oligo microarrays (Agilent Technologies, Inc., Santa Clara, CA). Hybridizations were performed with four biological replicates per group, using one-color labeling (Cy3), according to the manufacturer’s protocol (Agilent Manual: 64140-90040 v. 5.7). Briefly, 1 pg of total RNA from each sample was reverse-transcribed to cDNA in the presence of RNA One-Color Spike-In mix, T7 Promoter primer, 5X First Strand Buffer, 0.1M DDT, 10 mM dNTP mix, MLV-RT and RNaseOut during 2 h incubation at 40°C. Obtained cDNA was then converted to fluorescently labeled cRNA and amplified using 4X Transcription Buffer, 0.1M DDT, NTP mix, 50% PEG, RNaseOut, Inorganic 117 pyrophosphatase, T7 RNA Polymerase and Cyanine 3-CTP (Cy3) during second 2 h incubation at 40°C. The cRNA was purified using RNeasy Isolation Kit (Qiagen) and eluted with RNase—free water. Purified cRNA was assessed for Cy3 absorbance and concentration using NanoDrop spectrophometry. Fluorescently labeled cRNA was fragmented using 25X Fragmentation Buffer and 10X Blocking Agent for 30 min at 60°C before samples were hybridized. After 17 h incubation at 65°C, microarray slides were washed and scanned at 532 nm (Cy3) on a GenePix 40008 scanner (Molecular Devices, Union City, CA). Images were analyzed for feature and background intensities using GenePix Pro 6.0 (Molecular Devices). All data were managed in TIMS deach data management system [27]. Microarray Analysis and Functional Annotation All microarray data in this study passed quality assurance protocols [28]. Microarray data were normalized using a semiparametric approach [29] and the posterior probabilities were calculated using an empirical Bayes method based on a per gene and group basis using model-based t-values [30]. Gene expression data were ranked and prioritized using a [fold changel2l.5 and P1(t) values 20.95 to identify differentially expressed genes that were used for further investigation and interpretation. Annotation and functional categorization of differentially regulated genes was performed using Database for Annotation, Visualization and Integrated Discovery (DAVID) [31]. 118 RNA isolation and real-time PCR analysis: Lung tissue (10 mg) stored in RNAlater RNA Stabilization Reagent was homogenized in RLT buffer (RNeasy RNA isolation Kit, Qiagen, Maryland) using a Retsch MM200 bead beater system (Retsch, Haan, Germany). Total RNA quantification was performed spectrophotometrically (NanoDrop ND-1000 UV-Vis Spectrophotometer). Total RNA (1 pg) was reverse transcribed using superscript ll reverse transcriptase kit (lnvitrogen, CA). Expression level of selected genes involved in surfactant metabolism and lung development were analyzed by Real-Time PCR using SYBR green (Applied Biosystems, Foster City, CA) as previously described [32]. Gene specific primers are listed in Table 3.1. Copy number was determined by comparison with standard curves of the respective genes. This measurement was controlled for RNA quality, quantity, and RT efficiency by normalizing it to the expression level of the hypoxanthine guanine phosphoribosyl transferase (HGPRT) gene. Statistical significance was determined by use of normalized relative changes and ANOVA. Quantitative analysis: qRT-PCR analysis was performed using unpaired two-tailed Student’s t-test on GraphPad Prism. Results were considered significant at the 5% level. Results: Generation of HIF10t and HlF20t deficient mice and their survivability: Three strains of mice were generated that were capable of inducible lung- specific deletion of HIF10t, HIF20t, or HlF1oz/HIF20t as described in materials 119 Table 3.1. Primers for real-time RT-PCR. Gene ID Gene Name Forward Reverse 20505 Solute carrier family 34 ttgfiactaccactacagcc agtaggatgcccgagatgtt 841 12 Succinate receptor 1 RW tcctcaaattgggcatgata Methenyltetrahydrofolate 234814 synthetase domain cccaccggggtcatcactac atccttcccagcctgtttct 75512 Glutathione peroxidase 6 gcagtatgcaggaaagcaca caaaacggtgacgttgaatg Glutathione S-transferase 68214 omega 2 cgcctggacgtatatggact ttcaagaagcccaggaagac ‘ Single-minded homolog 1 20464 (Drosophila) tcgtcagcgtcaactacgtc ccgagatagtgggagtggaa Arginine vaSOpressin receptor 1 2000 2 ccccttgagtggagactaa ctgttgctgggagagctagg Heparan sulfate 3-0- 54710 sulfotransferase 3B1 att ct a ttcct c c t at t atct tccctt a tt 320376 BCL6 co-repressor-Iike 1 gggc_tggggtgatatcctga aflagagccagatggtttc BRCA1 interacting protein, 237911 helicase 1 tggctactaaaaficcctca ccaaaagggggc_ttg§ga 3-hydroxy-3-methylglutaryl- 15357 Co A reductase aagggactccccatcgag ggatatflggcattgacc Calcitonin/calcitonin-related 1 2310 golypeptide, alpha aagaagaagttcgcctgctg tgcaggatctcttctgagca 16178 Interleukin 1 receptor, type II ctcccctggggacaatacca agccgagataaaggtgctgt BRCA1/BRCA2-containing 210766 complex, subunit 3 agctgccaaaaatcctgtgt tgctgacatctgactgcaca 108030 Lin-7 homolog A (C. Elegans) agcagcaacaacaaccacaa tttttctgtgtggtgggattc 56360 Acyl-coa thioesterase 9 agacaaggtgggggagaiag tcattttcctggtggtagl 18145 Niemann Pick type C1 agggccatttaccatcattc agcagtcctggcagctacat 245195 Resistin like gamma (F IZZ3) ggaacttcggccaatcggg tagcacaagcacaaccaL Stearoyl-Coenzyme A 20249 desaturase 1 ccgggggaatatcctggttt We CDP-diacylglycerol synthase 74596 1 ctccctccctccattcctac gaatccactggcaaagaagc Glutathione S-transferase, 14866 mu 5 acggtacatctgtggggaag at c ttactct a 17395 Matrix metallopeptidase 9 thflct ctcatggtccaccttgttca 75646 retinoic acid induced 14 gtcctccccaggatcaataa cctgcagtactgtcgctcag 120 ii, VII 100 93.33 ) a L0 0 H O O O L_._Lg l (percent 01 \I 00 O O O wmm ~sss I—I O O 2.‘ ,_I_._,I~..I .. O l l 1aAIA 2dAIA 112dAIA Figure 3.3. Survivability Plot. Viability of neonatal HlF1aA/A,HlF2aA/A and HlF1/2aA/A pups. All the HIF1aA/A pups showed signs of respiratory distress whereas all the HlF2aA/A pups were viable with no signs of respiratory distress. Simultaneous removal of both the form of HIFa resulted in approximately 7% deaths. The data plotted is from n>~300 for HIF10tA/A, n>~100 for HIF2otA/A, n= 41 of HlF1/2aA/A pups. “Images in this dissertation are presented in color”. 121 and methods. To achieve recombination of the floxed alleles, dams were exposed to doxycycline as previously described [21]. Dams of genotype controls for the triple or quadruple embryos were maintained on regular feed and water. The litters were genotyped via PCR using established primers. Lung-specific deletion of HIF10t leads to 100% lethality upon parturition from a pathology that resembles respiratory distress syndrome (Fig. 3.3) [21]. In contrast, deletion of HlF20t in the developing lung shows no adverse consequences to the animal. HlF20tA/A mice are born in appropriate Mendelian ratios and display no signs of R08 (Fig. 3.3). These results suggest that HlF20t is not essential for proper lung development as previously reported [20]. Interestingly, the HlF1/20tA/A mice also displayed little pathology. There was a slight decrease in survivability but othenlvise the double deficient pups appeared identical to their Iittermate and genotype controls (Fig. 3.3). These results suggest that loss of HlF20t can rescue the RDS-like phenotype seen in the HlF1orA/A mice. Histopathological analysis of neonatal HIF-deficient mice: Pups were euthanized approximately 1 hour post-parturition and the lungs were examined. HlF10lA/A mice confirmed previous finding and displayed attenuated alveolar space and thickened septa compared to controls (Fig. 3.4A and 3.4B)[21]. In contrast, the lungs from HIFZqA’A mice displayed thinner septa and larger alveolar space than controls (Fig. 3.4A and 3.4C). Finally, the HIF1/20’”A pups appeared similar to control pups. They displayed 122 Figure 3.4. Pulmonary Histopathology. Light photomicrographs of hematoxylin and eosin stained lung sections from control (A) and HIF1oA/A (B), HIF2aA/A (C), HIF1/20AM (o) pups. Lung from HlF1aA/A pup has less alveolar airspace (a) and thicker alveolar septa (S) compared to control. The lungs from HlF2c1A’A mice displayed thinner septa and larger alveolar space than controls. The lungs from HIF1/20“"A mice resembled lungs from control in terms of septal thickness and alveolar space. "Images in this dissertation are presented in color." 123 124 normal septa and alveolar architecture when compared to controls (Fig. 3.4A and 3.4D). Expression of HIF10t and HIFZot In the three genoptypes: To access the efficiency of HIF deletion, immunohistochemistry was performed on lung sections from HIF1GNA, HIF2oA’A, HIF1/20M, and their respective controls. In neonates, the expression of HlF10t is predominantly localized to alveolar and bronchiolar epithelial cells in controls and HlF2ch’A mice (Fig. 3.5A, C, D, and E). The level of HIF10t positive staining was drastically reduced in the HIF10lA/A and HlF1/20tA/A mice (Fig. 3.58 and F). The extent of HlF10t staining increased in HlF2rxA’A pups at the neonatal stage (Fig. 3.5D). In neonates, the level of HlF20t staining was attenuated in each of the three deficient genotypes as compared to their controls (Fig. 3.6). This confirmed the recombination at this HlF20t floxed locus in the HIF20iA/A and HlF1/2orA/A mice. Moreover, it supports the previous observation that loss of HIF10t leads to a decrease in HIF20t expression (Fig 3.6A and B) [21]. The results confirm the functional deletion of the floxed loci and suggest that there is some level of coordination between the expression of these two hypoxia regulated factors and this coordination plays a role in lung development During development, the newly differentiated Type II cells use glycogen Stores to produce surfactants. Lung sections from HIF1<1“’A pups (Fig. 3.78) had larger reserves of glycogen as compared to HIF2aA’A pups (Fig. 3.7D). In contrast, lung sections from HIF1QNA/HIF20NA pups (Fig. 3.7F) were devoid of 125 Figure 3.5. Immunohistochemistry for HIF10t. Lung sections from control (A,C,E) and HlF10tA/A(B), HIFZGA/A (C), HIF1/2mmA (F) pups were immunostained for HIF10tas described in “materials and methods". Representative positively stained cells for HIF10t are depicted by solid arrows. "Images in this dissertation are presented in color." 126 127 Figure 3.6. Immunohistochemistry for HIFZot. Lung sections from control (A,C,E) and HIF10tA/A(B), HIFZaA/A (C), HIF1/20cmA (F) pups were immunostained for HlF20t as described in “materials and methods” Positively stained cells for HlF20l are depicted by solid arrows. "Images in this dissertation are presented in color." 128 129 Figure 3.7. Periodic acid Schiff (PAS) staining Periodic acid Schiff (PAS) stained lung sections from control (B) and HIF1c1A/A (D) pups. Cuboidal epithelium lining alveolar septa in HIF1uA/A pup has greater PAS-stained glycogen (arrows) than that of the control pup (D). "Images in this dissertation are presented in color." 130 131 PAS-positive cells and were undistinguished from control pups (Fig. 3.7A, 3.70, 3.75). Genomic Response to HIF10t and HlF20t Deletions To understand the collective roles of HIF10t and HIF20l in the fetal lung development, gene expression was assessed using the 4 x 44 K Mouse Agilent array containing approximately 44,000 oligonucleotide probes, representing 34,204 annotated genes, including approximately 20,969 unique genes. Model-based t-values that compared treated and vehicle responses followed by Empirical Bayes analysis identified 1,174 differentially expressed genes based on a p1(t) > 0.95 and absolute fold change 21.5-fold (Fig. 3.8A). Overall, 632 genes were induced and 439 genes were repressed in the HIF10tA/A as compared to control pups. The change in gene expression in NA . . . . HIF10t ranged from 37.07-fold induction (Solute carrier family 34) to -16.00- . A/A fold repressmn (StearoyI-Coenzyme A desaturase 1). In HlF2ot 102 genes were induced and 45 genes were repressed whereas in HIF1/2053]A 43 genes were induced and 108 genes were repressed. The changes in gene expression were ranged from 9.00-fold (Fc receptor-like 6) and 5.84-fold (glutathione S-transferase, mu 5) induction to -3.03-fold (olfactory receptor 583) and -6.6-fold (apolipoprotein F) repression in HIF20tA/A and HIF1/20tA/A, 132 Figure 3.8 Differential gene expression between HIF1aNA, HIFZCNA and HIF1/2aNA. Identification of differentially regulated genes (A). Comparative gene expression analysis between HIF1oA’A, HIF2<1"“’A and HIF1/20’1"A pups. The Venn diagram illustrates common and unique differentially expressed genes (8). “Images in this dissertation are presented in color”. 133 A Probes [ m44,000 ] Entrez GenelD filter ‘1, Unique annotated genes [ 20,969. ] | Fold change I 21.5 ‘1’ P1(t) 2 0.95 Differentially regulated [ 11 74 ] unique genes B 10tA/A (1069) ZaA/A (155) 1/2orNA (149) 134 respectively. List of differentially expressed genes is provided in Appendix Table A.1. Database for Annotation, Visualization, and Integrated Discovery (DAVID) Analysis To further investigate the pattern of cellular and physiological impacts of the HIFoc deletions, Analysis through DAVID was conducted to allow detection of biological pathways and processes affected due to the deletion of either or both the isoforrns of HIFoc. Entrez gene ID list of 1174 differentially expressed unique genes were analyzed using all the genes representing mouse genome oligo-microarrays as a background. Given the wide array of biological processes regulated by HlFs and their roles in perinatal development, the effects on biological processes and cellular pathways were determined. Important cellular pathways that were affected include cell cycle and vesicular transport (table 3.2). Gene set enrichment analysis using the differentially expressed genes showed 45 biological processes that were significantly (p s 0.05) affected by deletions of HIF10t, HlF20t and HIF1/2a (table 3.3). Surfactant biosynthesis and secretion pathways affected by HIF1a and HIF2a Deletions An extensive literature search and microarray analysis identified changes in genes known to be involved in surfactants biosynthesis, processing and 135 Table 3.2. Cellular pathways affected by deletions of HlF1ot, HIF2oi and HIF1/2a Cellular pathways affected by deletions of HIF1a, HIF201 and HIF1/2a Genomic Catemry Pathways Response Count P-Value KEGG Pathways Cell cycle Upregulation 8 0.007 KEGG SNARE interactions in vesicular Pathways transport Downregulation 5 0.014 Biocarta The role of FWE-finger proteins Pathways in vesicle transport Downregulation 3 0.021 KEGG Leukocyte transendothelial Pathways migration Downrggulation 8 0.037 KEGG Pathways Prostate cancer Downregulation 7 0.046 KEGG Pathways Butanoate metabolism Downregulation 5 0.046 KEGG Pathways ABC transporters - General [flewtion 4 0.054 KEGG Pathways Ubiquitin mediated proteolysis ggregulation 7 0.054 KEGG Urea cycle and metabolism of Pathways amino goups Downrefllation 4 0.055 KEGG Pathways Cell Communication Downregulation 8 0.058 KEGG Pathways Tifljunction Downregulation 8 0.060 KEGG Glycosylphosphatidylinositol(GPl)- Pathways anchor biosynthesis Upregulation 3 0.073 KEGG Pathwgys Tryptophan metabolism Upregulation 4 0.090 136 Table 3.3. Biological processes affected by deletions of HlF1ot, HIan and HIF1/2a Term Count P-Value 1 Biopolymer metabolic process 285 1.48E-05 2 RNA metabolic process 168 5.99E-04 3 Cell division 25 0.0020 4 Macromolecule metabolic process 343 0.0041 5 Intracellular signaling cascade 84 0.0044 6 Small gtpase mediated signal transduction 34 0.0059 7 Intracellular transport 51 0.0068 Nucleobase, nucleoside, nucleotide and nucleic acid 8 metabolic process 201 0.0069 9 Biopolymer modification 108 0.0071 10 Primary metabolic process 385 0.0091 1 1 Intracellular protein transport 35 0.0103 12 Gene expression 184 0.0128 13 Cellular localization 61 0.0136 14 Protein modification process 102 0.0150 15 Post-translational protein modification 90 0.0160 16 Establishment omrotein localization 52 0.01 73 17 RNA procesfig 31 0.0194 18 Regulation of transcription, DNA-dependent 1 30 0.0197 19 Transcription 140 0.0210 20 Protein localization 55 0.021 7 21 Cellular component organization and biogenesis 153 0.0218 22 Response to X-ray 4 0.0220 23 Metabolic process 418 0.0232 24 mRNA metabolic process 23 0.0243 25 Regiation of liquid surface tension 3 0.0246 26 Hindgut morphogenesis 3 0.0246 27 Regulation of macrophage activation 3 0.0246 28 Response to ionizing radiation 5 0.0256 29 Establishment of cellular localization 58 0.0263 30 Regulation of cellular process 216 0.0268 31 Biopolymer catabolic process 23 0.0272 32 Macromolecule localization 56 0.0276 33 Transcription, DNA-dependent 130 0.0277 34 RNA biosLnthetic process 130 0.0287 35 Protein transport 48 0.0288 36 Mitosis 17 0.0318 37 Rho protein signal transduction 11 0.0320 38 M phase 21 0.0323 39 M phase of mitotic cell cycle 17 0.0332 40 Biological remilation 256 0.0347 41 ReQJIaIion of angi_ogenesis 6 0.0380 42 Regulation of transcription 133 0.0409 137 Table 3.3. Continued. Term Count P-Value 43 Embryonic organ development 6 0.0421 44 Cellular metabolic process 376 0.0475 45 Amino acid transport 7 0.0489 Regulation of nucleobase, nucleoside, nucleotide and 46 nucleic acid metabolic process 134 0.0551 47 Carboxylic acid transport 8 0.0561 48 Phosphcmid biosynthetic process 8 0.0561 49 Membrane lipid biosynthetic process 9 0.0571 50 Oganic acid transport 8 0.0599 51 Regulation of cell size 13 0.0602 52 Cellular zinc ion homeostasis 3 0.0620 53 Zinc ion homeostasis 3 0.0620 54 Vacuolar transport 4 0.0647 55 Protein targeting 19 0.0675 56 Regulation of biological process 231 0.0680 57 Dephosphorylation 1 3 0.0685 58 Cengrowth 1 2 0.0687 59 Regulation of smooth muscle contraction 4 0.0739 60 Negative regulation of angi_ogenesis 4 0.0739 61 Nucleocytoplasmic transport 1 1 0.0749 62 Protein-RNA complex assembly 9 0.0760 63 Macrophage activation 3 0.0770 64 Central nervous system neuron axonogenesis 3 0.0770 65 Nuclear transport 1 1 0.0784 66 Ras protein signal transduction 16 0.0784 67 Rgpilation of gene expression 138 0.0796 68 Phosphate transport 9 0.0844 69 Transition metal ion transport 7 0.0868 70 Ectodermal gut development 3 0.0930 71 Cerebellar Purkinje cell differentiation 3 0.0930 72 Cerebellar Purkinje cell layer formation 3 0.0930 73 Cerebellar Purkinje cell layer morphogenesis 3 0.0930 74 Lysosomal transport 3 0.0930 75 Ectodermal gut morphogenesis 3 0.0930 76 Pyrimidine nucleotide biosynthetic process 4 0.0941 77 Ubiquitin mle 33 0.0962 138 Table 3.4 Genes involved in surfactant metabolism HIF10l HIF20i HIF1I20t Gene Symbol Ratio Ratio Ratio surfactant associated protein 8 Sftpb -2.40 -1.07 -1.81 surfactant associated protein A1 Sftpat -4.70 -1.02 -1.47 synaptosomaI-associated protein 23 Snap23 -1.50 1.27 -1.13 golgi SNAP receptor complex member 1 Gosr1 -1.54 1.44 -1.19 golgi SNAP receptor complex member 2 Goer 202 1.34 -1.25 syntaxin 19 Stx19 -2.14 1.04 -1.10 swtaxin 11 Stx11 -1.98 1.28 -1.06 ATP-binding cassette, sub-family A (ABC1), member 1 Abca1 -1.72 1.16 -1.20 ATP-binding cassette, sub-family A (ABC1), member 6 Abca6 -4.46 1.98 -1.48 acfl-Coenzyme A oxidase 2, branched chain Acox2 -1.69 1.32 -1.14 stearoyl-Coenzyme A desaturase 1 Scd1 16.67 -1.08 -3.58 sterol reguZIatoerelement bindingfactor 2 Srebf2 -1.51 1.05 -1.23 low density lipoprotein receptor Ldlr -1.33 1.04 -1.09 CDP-diacnglycerol synthase 1 Cds1 -2.30 1.07 -1.39 acyl-CoA thioesterase 1 Acot1 -1.18 1.16 -1.28 apyl-CoA thioesterase 9 Acot9 -2.29 1.12 -1.38 lysophosphatidylcholine acyltransferase 1 chat1 -2.93 -1.04 -2.52 CDP-diacylglycerol synthase (phosphatidate cytidylyltransferase) 2 Cdsz -1.09 1.28 -1.14 phosphatidylethanolamine binding protein 2 Pbp2 -2.91 -1.06 -1.13 inositol pohrphostjlate-S-phosphatase A lnpp5a -1 .51 1 .17 -1.09 inositol 1,4,5-trisphosphate 3-kinase A Itpka -2.38 1.10 1.12 phosphatidylinositol glycan anchor biosynthesis, class A Figa -1.27 1.25 1.21 phosphatidylglycerophosphate synthase 1 3951 -1.32 -1.10 -1.28 choline phosphotransferase 1 Chpt1 -1.74 1.27 -1.11 choline kinase alpha Chka -1.03 1.03 1.26 1-acylglycerol-3-phosphate O- acyltransferase 3 Agpat3 -1.30 1.02 -1.10 fatty acid synthase Fasn -1.34 1.17 -1.28 CAMP responsive element bintfipg protein 5 Creb5 -1.66 1.24 -1.40 CAMP responsive element bindigg protein 3 Creb3 -2.94 1.10 1.00 syntaxin 6 Stx6 -1.16 1.33 -1.51 syntaxin 3 Stx3 -2.04 1.38 -1.71 syntaxin 5A Stx5a -1.41 1.30 -1.18 N-ethylmaleimide sensitive fusion protein st -1.47 1.26 -1.23 139 Figure 3.9 QRTPCR verification of selected microarray gene expression responses. The same RNA used for CDNA microarrays was examined by QRTPCR. All fold changes were calculated relative to controls. Iars (left y-axis) and lines (right y-axis) represent QRT-PCR and microarray data, respectively. Expression levels are expressed relative to HGPRT. “a” means statistically different from 2qA/A, “b" means statistically different from 1oA/A,“c” means statistically different from 1/2CIA/A. a, b and c designate statistical significance at p 50.05. 140 CDP-diacylglycerol Synthase 1 [:QRT-PCR 'V' Microarray .A U" .1. '0 easy UOISSOdea museum 9 m P o EIQRT-PCR 'V' Microarray .3. 0" .1 '0 easy uoissedea ABJJBOJOIW I P m P o 2 1.25 b "if a; 1.00« I i .9. A g 0.75- \ a. 1 iii 0.50 if m x o E 0.25 0.00 10LNA Zeb“ 1/20. NA HIF Deletion Strain 3-HMG Coenzyme A Reductase o 1.5 E g t - 1.0- I - 5 c a: 0.5 I ii at o 0 0 . 1a.NA 2(1.AVA 1/20.AVA HIF Deletion Strain 141 Figure 3.9. Continued. |:|QRT-PCR '0' Microarray Matrix Metallopeptidase 9 o 1.0 1 0 g .— O 3'3 5 a a < 3 w a 0.5 -0.5 '2 III a a: 3;. 0 ac o ’ 3 0.0 , ~ 0.0 ° 1a.NA 206MA “20.13,A HIF Deletion Strain :IQRT-PCR 'V' Microarray Interleukin 1 Receptor- II o 3 2 s z 0 -§ 2. Q 3 Q s -1 2 O 3 1- E. u a o 3 t E a 0 0 ° 1(1NA ZQNA 1/20.Am HIF Deletion Strain 142 Figure 3.9. Continued. CJQRT-PCR '0' Microarray Glutathione S-transferase mu 5 2 a 10.0 g 3°: § 5 12‘ ~7.5 a “g a f“ 5 3‘ ~50 13 a 3 m 2. E 4* ~2.5 3 tr :0 0 III .3 0 0.0 ° 1U.NA ZaNA 1,2“ NA HIF Deletion Strain :lQRT-PCR Hs3st3b1 'V' Microarray o 10.0 a.c 4 g f 3 ‘2 7.5-« 1: -35 '15. e m '3 5.0- - 2 13 3 o (r 3. g 2.51 -1 g a: :0 O b 3.. O 0.0 E , 1am. ZaNA 1120. NA HIF Deletion Strain 143 Figure 3.9. Continued. [:QRT-PCR 'V' Microarray Niemann Pick type C1 0 2.0 1.5 g :3 2 0: c o g 1.5 3 '5 -1.0 Q 3 L“ a 1.0- 1.3, a 3 a: I ~0.5 g. g 0.51 a a: '1; 2.: o 0.0 A, A, 0.0 3' 1aA’A 2a. A 1/2or A HIF Deletion Strain [:lQRT-PCR 'V' Microarray Glutathione S-transferase 00-2 2 4 g £- I 3' a: . g. g I .-3 it .- 9 a a 7;, 0 .. t 5 x -1 § it ~ :0 o 3. 0 A! A/ A/ O 0 1a. A 20. A 1I2a A HIF Deletion Strain 144 secretion pathways. The comparison of HlF1oA/A expression patterns and those of HIF1/20mm identified several genes that showed rescued expression upon loss of both HIF isoforrns. These genes are important in lipid biosynthesis (e.g. acyl-CoA thioesterase 9, sterol regulatory element binding factor 2) or surfactant transport and secretions (e.g. golgi SNAP receptor complex member 2, SNAP 23). Interestingly, some of the genes such as stearoyl- Coenzyme A desaturase 1 and surfactant protein B did not exhibit significantly increased expression after accompanied deletion of HlF2a. QRTPCR verification of selected microarray gene expression responses. Quantitative real-time PCR (QRT-PCR) was used to verify the differential expression for a selected subset of differentially expressed genes representing different response profiles and functions (table 3.1). In general, there was a good agreement in the level of differential expression when comparing microarray and QRT-PCR data (Fig. 3.9). Discussion Normal prenatal lung development is essential for the smooth transition of the placental gas transport to an air breathing at the birth. This complex process is characterized by biochemical, cellular and ultrastructural changes that are precisely controlled by the spatiotemporal pattern of expression (or activity) of large number of transcriptional and paracrine factors. Several 145 transcription factors including KLF5, Foxa2, CEBPa, GATA-6, B-catenin and thyroid transcription factor-1 have already been added to the list of indispensables for lung development [3-7, 9]. However, further testing for the other devel0pmentally critical transcriptional factors, individually or collectively, will provide a broader understanding on the intricate networks and cascades involved in lung development. HlFs are a class of transcription factors that play a critical role in oxygen sensing and the metabolic adaptations to hypoxia. HlFla is a most widely expressed isoform of HlFs. Previous studies have shown that targeted gene disruption of the HlF1a locus results in the mid-gestational lethality due to cardiovascular defects [15, 33]. Mice partially deficient for HlF1o alleles showed normal development but were physiologically compromised as they exhibited impaired pulmonary vascular remodeling under chronic hypoxic stress [16, 34]. Similarly, targeted disruption of HlF2a, another pulmonary isoform of HlFs, have shown that reduced VEGF expression leads to respiratory distress at birth [20]. Recently, using a lung-specific HlF1a deficient mouse model, epithelial- derived HlF1a deletion led to compromised lung development and altered surfactant metabolism [21]. The epithelial-specific loss of HlF1oc leads to neonatal respiratory distress syndrome due to impaired alveolar epithelial differentiation. Morphogenesis of conducting ainNays, however, was unaffected, suggesting the involvement of HlF1oc in late events in lung 146 morphogenesis rather than in the early lung biogenesis and airway branching morphogenesis. To further investigate the role of HlF2a in lung development, lung-specific HlF2a deficient and HlF1od2a deficient mice were generated. The results suggest that lung-specific loss of HlF2a does not result in the respiratory distress. The difference between these results and previously published reports most likely stems from genotype and strain differences of the mice used [20]. The previously published report used traditional HlF2a knockout mice generated in a specific genetic background and the conclusions were based on a percentage of mice that survived until parturition. These differences in genetic background, genotype, and the loss of HIF2a from other organs, most notably the cardiac tissue, are potentially confounding factors. The data presented here suggests that loss of HlF2a specifically from the lungs leads to no overt phenotype and actually might lead to more developed lungs when compared to genotype controls (Fig. 3.4). The simultaneous deletion of HlF1a and HlF2a isoforrns rescues the respiratory distress phenotype that was observed in HlF1oc deficient mice. To investigate the mechanisms underlying these interesting, microarray analysis was performed to identify differentially expressed genes. Extensive data mining and literature search revealed that the deficiency of HlF1a protein results in the differential expression of 1069 unique genes whereas the 147 deficiency of HlFZa protein affected the expression of only 155 genes. Interestingly, after HlF1a/2a deficiency the list of differentially expressed genes was reduced to only 149 out of which 113 genes overlapped with HlFla deficient mice (Fig. 3.88). Out of 1069 genes, the expression patterns of 890 genes were specifically disturbed in HlF1or deficient mice. Previous study on the characterization of the respiratory distress phenotype in HlF1a deficient mice highlighted the altered surfactant proteins expression and the expression of developmentally critical genes [21]. However, the screening for the biological processes affected by the deletion of HlF1a returned a list of wide variety processes such as biopolymer metabolic processes, RNA metabolism, cell division, intracellular signaling cascades and intracellular protein sorting (Table 3.3). Further search for the cellular pathways affected revealed the perturbation of vesicular transport pathways and cell cycle pathways (Table 3.2). Proper functioning and regulation of vesicular transportation is an important step in the processing and secretion of surfactants and associated proteins. Pulmonary surfactants are modified in the golgi apparatus and are then released from the trans golgi network into secretory vesicles, known as composite bodies, the immediate precursors of lamellar bodies. The lamellar body-bound surfactants are released via exocytosis into the lumen alveolar lumen where it forms a lattice-like network known as tubular myelin. The processes involving intracellular sorting (golgi transport, vesicular fusion) and 148 surfactant biosynthesis (fatty acid metabolism, cholesterol sythesis, plospholipids biosynthesis) are specifically affected in HlF1a deficient mice (Table 3.4). Genes involved in vesicular transport such as Gosr2, SNAP-23, Syntaxins are downregulated in HlF1or deficient mice but were restored to normal levels in HlF1o/2a deficient mice [35]. Similarly, levels of expressions of genes involved in lipid metabolism such as Acot9, CDS-1, SOD-1, Acox2, CDS-2, Chpt-1, Itpka were increased in HlF1a/2a deficient mice [36, 37]. PAS staining for glycogen deposition also suggested the complete utilization of glycogen in HlF2a and HlF1a/2a deficient mice (Fig.3.7). Taken together, these findings suggest the global alteration of the surfactant metabolism starting from the phospholipids and protein biosynthesis to the final exocytosis of packaged surfactants into the alveolar space upon perturbation of the hypoxia signaling cascade in the lungs. Interestingly, genes involved in cellular processes such as cell cycle and biopolymer metabolism showed differential gene expression pattern. The first category includes genes such as interleukin 1 receptor type II (lL1-Rll) that are minimally expressed in from HIF1aA/A lungs but show higher expression in HIF1/2(1A/A lungs. The second category includes genes such as heparan sulfate (glucosamine) 3-O-sulfotransferase 3A1 (Hs3st331) that are 149 highly expressed in HlF1c1A/A lungs but show lower expression in HIF1/2(1A/A lungs (Fig. 3.9). Interleukin 1 (lL1oc and IL1B) has been shown to promote lung function and surfactant biosynthesis [38]. Similarly, heparan sulphate proteoglycans are required for various signaling pathways critical in the lung development [39]. I In conclusion, the present study provides evidence that HlF1a plays an important role in multiple processes including surfactant metabolism, metabolic pathways, signal transduction cascades and vesicular trafficking. The lung-specific deletion of HlF2a alone, however, is not lethal to the neonatal pups and there was no sign of observable pathology. Interestingly, the rescue of respiratory distress was observed after simultaneous deletion of both the isoforms of HlFs. Microarray analysis of the lungs has identified interesting genes that might be the focus of further investigation. 150 REFERENCES 1 James Metcalfe, H. B., Waldemar Moll. (1967) Gas Exchange in the Pregnant Uterus. Physiol. Rev. 47, 782-838. Groenman, F., Unger, S. and Post, M. (2005) The molecular basis for abnormal human lung development. Biology of the Neonate. 87, 164- 177 DeFelice, M., et al. (2003) TTF-1 phosphorylation is required for peripheral lung morphogenesis, perinatal survival, and tissue-specific gene expression. The Journal of biological chemistry. 278, 35574- 35583 Liu, C., Morrisey, E. E. and Whitsett, J. A. (2002) GATA-6 is required for maturation of the lung in late gestation. Am J Physiol Lung Cell Mol Physiol. 283, L468-475 Martis, P. C., et al. (2006) C/EBPalpha is required for lung maturation at birth. Development (Cambridge, England). 133, 1 155-1 164 Mucenski, M. L., et al. (2003) beta-Catenin is required for specification of proximal/distal cell fate during lung morphogenesis. The Journal of biological chemistry. 278, 40231-40238 Wan, H., et al. (2004) Foxa2 is required for transition to air breathing at birth. Proceedings of the National Academy of Sciences of the United States of America. 101, 14449-14454 Yang, H., et al. (2002) GATA6 regulates differentiation of distal lung epithelium. Development (Cambridge, England). 129, 2233-2246 Wan, H., et al. (2008) Kruppel-like factor 5 is required for perinatal lung morphogenesis and function. Development (Cambridge, England). 135, 2563-2572 151 10 11 12 13 14 15 16 17 18 19 Dave, V., et al. (2006) Calcineurin/Nfat signaling is required for perinatal lung maturation and function. The Journal of clinical investigation. 116, 2597-2609 Semenza, G. L. (2000) HIF-1: mediator of physiological and pathophysiological responses to hypoxia. J Appl Physiol. 88, 1474-1480 Groenman, F., et al. (2007) Hypoxia-inducible factors in the first trimester human lung. J Histochem Cytochem. 55, 355-363 Cowden Dahl, K. D., et al. (2005) Hypoxia-inducible factors 1 alpha and 2 alpha regulate trophoblast differentiation. Molecular and cellular biology. 25, 10479-10491 Compemolle, V., et al. (2003) Cardia bifida, defective heart development and abnormal neural crest migration in embryos lacking hypoxia-inducible factor-1 alpha. Cardiovascular research. 60, 569-579 Ryan, H. E., Lo, J. and Johnson, R. S. (1998) HIF-1 alpha is required for solid tumor formation and embryonic vascularization. The EMBO journal. 17, 3005-3015 Yu, A. Y., et al. (1999) Impaired physiological responses to chronic hypoxia in mice partially deficient for hypoxia-inducible factor 1alpha. The Journal of clinical investigation. 103, 691-696 Peng, J., Zhang, L., Drysdale, L. and Fong, G. H. (2000) The transcription factor EPAS-1/hypoxia-inducible factor 2alpha plays an important role in vascular remodeling. Proceedings of the National Academy of Sciences of the United States of America. 97, 8386-8391 Patel, S. A. and Simon, M. C. (2008) Biology of hypoxia-inducible factor-2alpha in development and disease. Cell death and differentiation. 15, 628-634 Asikainen, T. M., et al. (2006) Improved lung growth and function through hypoxia-inducible factor in primate chronic lung disease of prematurity. Faseb Journal. 20, (E986-E994) 1698-1700 152 2O 21 22 23 24 25 26 27 28 29 Compemolle, V., et al. (2002) Loss of HIF-2alpha and inhibition of VEGF impair fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice. Nature medicine. 8, 702-710 Saini, Y., Harkema, J. R. and LaPres, J. J. (2008) HlF1alpha is essential for normal intrauterine differentiation of alveolar epithelium and surfactant production in the newborn lung of mice. The Journal of biological chemistry. 283, 33650-33657 Ryan, H. E., Lo, J. and Johnson, R. S. (1998) HIF-1 alpha is required for solid tumor formation and embryonic vascularization. Embo Journal. 17, 3005—301 5 Ryan, H. E., et al. (2000) Hypoxia-inducible factor-1 alpha is a positive factor in solid tumor growth. Cancer Research. 60, 4010-4015 Perl, A. K., et al. (2002) Early restriction of peripheral and proximal cell lineages during formation of the lung. Proc Natl Acad Sci U S A. 99, 10482-10487 Lobe, C. G., et al. (1999) Z/AP, a double reporter for ore-mediated recombination. Dev Biol. 208, 281-292 Gruber, M., et al. (2007) Acute postnatal ablation of Hif-2alpha results in anemia. Proceedings of the National Academy of Sciences of the United States of America. 104, 2301-2306 Burgoon, L. D. and Zacharewski, T. R. (2007) deach toxicogenomic information management system. Pharrnacogenomics. 8, 287-291 Burgoon, L. D., et al. (2005) Protocols for the assurance of microarray data quality and process control. Nucleic acids research. 33, e172 Eckel, J. E., et al. (2005) Normalization of two-channel microarray experiments: a semiparametric approach. Bioinforrnatics (Oxford, England). 21, 1078-1083 153 30 31 32 33 34 35 36 37 38 39 Eckel, J. E., et al. (2004) Empirical bayes gene screening tool for time- course or dose-response microarray data. Journal of biopharrnaceutical statistics. 14, 647-670 Dennis, G., Jr., et al. (2003) DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome biology. 4, P3 Vengellur, A. and LaPres, J. J. (2004) The role of hypoxia inducible factor 1 alpha in cobalt chloride induced cell death in mouse embryonic fibroblasts. Toxicological Sciences. 82, 638-646 lyer, N. V., et al. (1998) Cellular and developmental control of 02 homeostasis by hypoxia-inducible factor 1 alpha. Genes & Development. 12, 149-162 Semenza, G. L. (2005) Pulmonary vascular responses to chronic hypoxia mediated by hypoxia-inducible factor 1. Proceedings of the American Thoracic Society. 2, 68-70 Abonyo, B. 0., et al. (2004) Syntaxin 2 and SNAP-23 are required for regulated surfactant secretion. Biochemistry. 43, 3499-3506 Zhang, F., Pan, T., Nielsen, L. D. and Mason, R. J. (2004) Lipogenesis in fetal rat lung: importance of C/EBPalpha, SREBP—1c, and stearoyl- CoA desaturase. American journal of respiratory cell and molecular biology. 30, 174-183 Batenburg, J. J. and Haagsman, H. P. (1998) The lipids of pulmonary surfactant: dynamics and interactions with proteins. Progress in lipid research. 37, 235-276 Willet, K. E., et al. (2002) lntra-amniotic injection of lL-1 induces inflammation and maturation in fetal sheep lung. Am J Physiol Lung Cell Mol Physiol. 282, L411-420 Hu, 2., et al. (2009) NDST1-dependent heparan sulfate regulates BMP signaling and internalization in lung development. Journal of cell science. 122, 1145-1154 154 Chapter 4 The Role of Hypoxia Inducible Factor 10: (HlF1a) in Modulating Cobalt- lnduced Lung Inflammation This chapter is the edited version of a research article that was published in American Journal of Physiology, Lung Cellular and Molecular Physiology. 2009 Nov 13. Authors: Yogesh Saini, Kyung Y. Kim, Ryan Lewandowski, Lori A. Bramble, Jack R Harkema, John J. LaPres 155 ABSTRACT Hypoxia plays an important role in development, cellular homeostasis, and pathological conditions, such as cancer and stroke. There is also growing evidence that hypoxia is an important modulator of the inflammatory process. Hypoxia inducible factors (HlFs) are a family of proteins that regulate the cellular response to oxygen deficit and loss of HlFs impairs inflammatory cell function. There is little known, however, about the role of epithelial-derived HIF signaling in modulating inflammation. Cobalt is capable of eliciting an allergic response and promoting HlF signaling. To characterize the inflammatory function of epithelial derived HIF in response to inhaled cobalt, a conditional lung specific HlF1or deletion mouse was created. Wild type mice showed classic signs of metal-induced injury following cobalt exposure, including fibrosis and neutrophil infiltration. In contrast, HlF1or deficient mice displayed a Th2 response that resembled asthma, including increased eosinophilic infiltration, mucus cell metaplasia, and chitinase-like protein expression. The results suggest that epithelial derived HIF signaling has a critical role in establishing a tissue’s inflammatory response, and compromised HlF1or signaling biases the tissue towards a Th2-mediated reaction. 156 INTRODUCTION Lung diseases, including chronic obstructive pulmonary disease and asthma, involve a large inflammatory component. The lung's response to allergens involves a complex interplay between resident inflammatory and epithelial cells, cytokine signaling, and the environmental conditions within the tissue. One of the critical environmental features that can impact the inflammatory process is hypoxia. Hypoxia, a decrease in available oxygen reaching the tissues of the body, has profound cellular and metabolic consequences. The cellular response to hypoxia is regulated by a family of transcription factors called the hypoxia inducible factors (HlFs) [1]. HlFs are primarily regulated at the level of protein stability by a family of prolyl hydroxylases. These prolyl hydroxylase domain (PHDs) proteins are members of a broader family of non-heme, iron- and 2- oxoglutarate dependent dioxygenases [2]. Cobalt has been shown to inhibit PHDs and this inhibition causes very similar transcriptional outputs to that of hypoxia [3, 4]. Recent research using human peripheral blood mononuclear cells has shown that this transcriptional overlap applies to tungsten carbide- cobalt particles, linking hard metal lung disease to hypoxia signaling [5]. HlF1or is the most ubiquitously expressed and widely studied HIF isoform. HlF1or heterodimerizes with the aryl hydrocarbon receptor nuclear translocator (ARNT, also known as HIF1B) forming the functional transcription factor HIF1. 157 HIF1 regulates the expression of over one hundred genes, including genes for glycolytic enzymes, sugar transporters, and pro-angiogenic and inflammatory factors [6-9]. Moreover, HlF1or has also been shown to modulate inflammation indirectly by influencing the NFKB signaling pathway [8, 10]. Given the relationship between cobalt, HlF1a, and inflammation, it seems likely that HlF1or will impact cobalt-induced injury in vivo. More specifically, it is hypothesized that cobalt-induced HIF1-mediated transcription will impact cobalt-related asthma and/or hard metal lung disease [5]. Cobalt (or hard metal) asthma is one of three occupational respiratory diseases associated with exposure to the transition metal. The other two are hypersensitivity pneumonitis and interstitial lung disease with fibrosis. These diseases are caused by the inhalation of hard metal particles and are characterized by airway constriction, alveolitis, fibrosis and associated giant cell interstitial pneumonitis [11]. Asthma associated with cobalt exposure most likely involves an allergic response and has variable latency periods following initial sensitization [12-14]. Cobalt-specific lmmunoglobulin isotype E (lgE), has been characterized in workers with signs of cobalt asthma, and their symptoms can be relieved upon removal from the contaminated environment [12]. Besides acting as a pro-oxidant and sensitizer in the lung and skin, cobalt has also been characterized as a hypoxia mimic [4]. 158 To characterize the role of HlF1or in cobalt-induced lung injury, a lung specific HlF1or knock out mouse model was created. In utero deletion of HIF1or led to lethality due to respiratory distress upon parturition [15]. In the present study, post-natal deletion of HlF1oc from Type II and Clara cells had no observable pathology. In order to elucidate the role of epithelial derived- HlF1a signaling in cobalt-induced lung injury, these mice were exposed to cobalt chloride via oropharyngeal aspiration. Compared to control mice, mice that were HlF1or deficient in their lungs (HIF10rA/A) exhibited airway infiltration of eosinophils associated with airway epithelial changes, including mucus cell metaplasia and increased levels of the chitinase-like proteins YM1 and YM2. Mice deficient in HlF1a also showed a drastic change in cytokine profiles in their lavage fluid when compared to their control. These results suggest that loss of HlF1or from alveolar Type II epithelial and Clara cells of the lungs leads to cellular and molecular processes that are associated with asthma following cobalt exposure and that airway epithelial-derived HlF1or plays a critical role in modulating the inflammatory response of the lung. MATERIALS AND METHODS Description of mice: Triple transgenic mice were created by mating HlF1orfl°wIOX (a generous gift of Randall Johnson, Univ. Califomia-San Diego) and SP-C-rtTA'ltg/(tetO)-/-CMV-Cretg/t9 transgenic mice (a generous gifts of 159 Jeffrey A. Whitsett Cincinnati Children’s Hospital Medical Center) [16-19]. The generated mice, SP-C-rtTA-ltg/(tetO)7-CMV-Cretg/tg/H|F1afloxmox, are capable of respiratory epithelium specific conditional recombination in the floxed HIF1o gene upon exposure to doxycycline [18]. In addition to the triple transgenic controls, four additional genotypes were employed to rule out effects of any one locus in the presence and absence of doxycycline. These include, SP-C- nTA'ltg/(tetO)7-CMV-Cre'l'lHlF1a+/+ (sTH), SP-C-rtTA'l'AtetOh-CMV- Cretg/tg/HIF1(1+/+ (StH), SP-C-rtTA'ltg/(tetOh-CMV-Cretg/tg/HlF10+” (stH) and SP-C-rtTA'l'/(tetO)7-CMV-Cre'l'lHlF1afloxmox (STh). The HlF1ofl°xm°x were originally maintained in a CS7BL/6 background, whereas the SP—C-rtTA'ltg/ (tetO)7-CMV-Cretg/tg were generated in an FVB/N genetic background. These parental strains were carefully mated to acquire the necessary genotypes for the described experiments and all of the mice used in this study have been maintained in this mixed CS7BL/6 and FVB/N background. Genotyping of the mice was performed by PCR for all the three loci as previously described [15]. Doxycycline treatment and animal husbandry: In utero exposure to doxycycline in the triple transgenic mice led to lethality upon parturition [15]. Postnatal recombination was carried out by exposing lactating dams to doxycycline containing feed (625 mg doxycycline/Kg; Harlan Teklad, Madison, 160 WI) and drinking water (0.8 mg/ml: Sigma chemicals Co.) until weaning. Triple transgenic mice were then maintained on the same doxycycline containing food and water until they were approximately 7 weeks of age. Doxycycline treatment was terminated 7-10 days prior to first exposure to metals. These mice will be referred to as HlF1adeficient or HlF1aA/A throughout the chapter. Control animals used in the study were triple transgenic (SP-C-rtTA' Itg/(tetO)7-CMV-Cretg/tnglF1afloxmox) mice that were maintained on normal food and water ad Iibitum. All of the remaining genotype control lines described above were exposed to doxycycline for the same 7 week paradigm when appropriate. Mice used in this study were kept at the animal housing facility under the strict hygienic and pathogen free conditions approved by the university laboratory animal resource (ULAR) regulatory unit. All the animal handling and necropsy protocols were approved by the ULAR regulatory unit of Michigan State University. Cobalt exposure, tissue harvesting and processing: 18 mice from each genotype were randomly assigned to one of three groups. Mice were treated with saline, 5 mM, or 10 mM cobalt chloride in 25 [LL volume by oropharyngeal aspiration. These cobalt concentrations correspond to 30 and 60 pg daily exposure, respectively. Mice were treated for 5 days on, 2 days off, 5 days on and then euthanized 72 hours after the last exposure. Mice were assessed for total body weight prior to first treatment and assessed again prior to sacrifice. Following exposure, mice were anesthetized with sodium pentobarbital (50 161 mg/ml), and a midline Iaparotomy was performed. The trachea was exposed and cannulated. The lung and heart were removed en bloc and the lungs were lavaged with two successive 1 ml volumes of sterile saline. These fractions were combined and total cell counts were performed using a hemocytometer. Differential cell counts were performed in cytospin samples using Diff-Quik reagent (Baxter, FL). The remaining BALF was frozen for cytokine profiling. The right lung lobe was removed and stored in RNA/afar RNA stabilizing reagent (Qiagen, Valencia, CA) for RNA isolation. The left lobe was perfusion inflated and fixed in 10% neutral buffered formalin for histopathological analysis. Histopathology and immunohistochemistry: At least four to six mice from each genotype and treatment group were analyzed for histopathological changes. Formalin fixed left lung lobe tissues were paraffin embedded and 5- micron thick sections were mounted on glass slides and stained with hematoxylin and eosin (H&E) or immunostained with HlF1a (1: 500 dilution, NB100-479, Novus Biologicals, Littleton, CO), major basic protein (1:500 dilution, Mayo Clinic, AZ), or YM1 (1:100 dilution, ABZ4608; Abcam, Cambridge, MA) as previously described [15]. Other lung sections were histochemically treated with picro-sirius red solution for interstitial collagen staining to identify areas of pulmonary fibrosis or with Alcian Blue (pH 2.5)lPeriodic Acid Schiff (AB-PAS) stain to identify mucosubstances in mucous cells. 162 RNA isolation and quantitative real-time PCR (qRT-PCR) analysis: Lung tissue (10 mg) stored in RNA/ater RNA Stabilization Reagent was homogenized in RLT buffer (RNeasy RNA isolation Kit, Qiagen, Maryland) using a Retsch MM200 bead beater system (Retsch, Haan, Germany). Total RNA quantification was performed spectrophotometrically (NanoDrop ND- 1000 UV-Vis Spectrophotometer). Total RNA (1 ug) was reverse transcribed using superscript ll reverse transcriptase kit (lnvitrogen, CA). cDNAs from each experimental group were pooled and quantitative real-time PCR array analysis reactions were carried out in duplicate on an ABI Prism 7900HT 384- well block using TaqMan assays (ABI, Applied Biosytems, Foster City, CA). The complete table of the genes analyzed is listed in table 4.1. Changes in gene expression were calculated using the 21““:t method. An average of the number of cycles of three housekeeping genes, GAPDH, Actin-B, and 188, was used to normalize the expression between samples. Genes that had greater than a 2 fold change in expression level when compared to the control group were further analyzed by qRT-PCR using individual samples run in duplicate to confirm the PCR array results. Samples exhibiting Ct values greater than 40 were deemed undetermined and removed from subsequent analysis. Outliers were removed by Grubbs test and averages compared. The fold changes for these genes are listed in Table 4.2. Determination of cytokine levels by Bead array: Bronchoalveolar lavage fluid was analyzed for lL-1B, Eotaxin, KC, lL-6, lL-10, lL-12, lL-4,|L-5, lL-13, IL- 163 2, Rantes, GM—CSF, INF-y, TNF-a, MOP-1, VEGF, MlP-2, MlP-1or using a Bio- Plex 200 system and reagents (Bio-Rad Laboratories Inc, Hercules, CA, USA) according to manufacturer's instructions. Briefly, cytokine specific antibody coupled color-coded beads are allowed to react with the cytokines present in the sample. Following extensive washing, a biotinylated detection antibody is added to the cytokine bound beads. The sandwich complex formation is detected by the addition of streptavidin-phycoerythrin. Cytokines are identified and quantitated based on bead color and fluorescence. Cytokine levels are calculated by system-specific software (Bio-Plex Manager”) using a standard curve derived from a recombinant cytokine standard. A multiplex assay was performed by mixing beads specific to each of above listed cytokines and incubating them with 50 pl of undiluted BALF in the provided 96 well filter plate. Following washing, all the respective detection antibodies were added to wells followed by addition of streptavidin-phycoerythrin. Values from blank wells for each cytokine were subtracted from the corresponding cytokine values in each sample. Negative values were set to zero. Outliers were removed by Grubbs’ Test, averages were calculated, and significance determined by ANOVA followed by Tukey’s HSD test. Quantitative analysis: All cell counts and cytokine and gene expression data were analyzed by ANOVA followed by a Tukey’s HSD test. All the data from the study was presented as standard error of the mean (SEM). Statistical difference of P value less than 0.05 was considered as significant. 164 RESULTS: Post-natal deletion of HlF1a: The initial description of the lung specific cre recombinase model had very little data regarding post-natal deletion, however, it can be assumed that doxy-induced cre expression will be restricted to Clara and Type II cells since SPC expression is restricted to these cells types in the adult animal [18]. To determine if prolonged exposure to doxycyline (Dox) could induce recombination within the conditional HlF1or locus, mice were exposed to Dox for approximately 7 weeks starting on post-natal day 4 (PN4). Initially, they received the drug through their mother’s milk, and then through their food and water. These mice will be referred to as HlFlor deficient or HlF1orA/A. The mice referred to as controls throughout the studies are triple transgenic (SP-C-rtTA-ltg/(tetO)7-CMV-Cretg/tg/HlF1dfloxmox) mice that were maintained on regular feed and water. The lungs of control and Dox treated mice were then removed and analyzed for HIF1or expression via immunohistochemistry (IHC). Control mice showed pronounced HlF1cr expression in the Clara cells lining the bronchiolar airway and Type II cells of the alveoli (Figs. 4.1A and 4.1B). In contrast, the HlF1dA/A mice showed a marked decrease in expression in these cells, with only minor staining visible in the bronchiolar ainlvay lining cells and little or no staining in the alveoli (Figs. 4.1C and 4.1 D). 165 Figure 4.1. HIF1a immunohistochemistry of lungs from control and doxycycline treated mice Lung tissue sections from control (A and B) and HlF1aA/A (C and D) were analyzed by immunohistochemistry using a HlF1a—specific antibody. Control (A and B) and doxycycline (C and D) treated mice were compared. HlF1astaining is prominent in the epithelial cell (e) lining the bronchiolar airway (BA) and type II cells (Solid arrow) in the alveolar duct (AD) and alveolus (a). Staining is greatly reduced in the postnatally doxycycline treated animals (C and I). "Images in this dissertation are presented in color." 166 167 The results suggest that postnatal exposure to Dox can induce significant recombination of the HlF1alocus and that the triple transgenic mouse is a viable model to test the role of HlF1a in cobalt-induced lung injury. Body weight change and cell counts in lavage fluid: To characterize the role of HlF1or in cobalt-induced lung injury, control and HlF1orA/A mice were randomly assigned to three groups. Individual groups received, sterile saline, 30, or 60 [lg cobalt chloride by oropharyngeal aspiration per day using a two week protocol, 5 days on, 2 days off, 5 days on, and 2 days off. Total body weight of some of the control mice rose slightly during the course of the exposure however the body weight gain remained largely unchanged. In contrast, the HlF1orA/A mice lost weight in a dose dependent fashion (Fig. 4.2A). Though this decrease was not significant, it was the first indication that the HlF1or deficient mice responded differently to cobalt challenge. After the final 2-day incubation, mice were euthanized, BALF was collected, and lung tissue was processed for light microscopic examination, IHC, and RNA isolation. HlF1a—deficient mice treated with 60 ug cobalt chloride showed significant increase in total BALF cells as compared to 60 pg cobalt chloride treated control mice suggesting that loss of HlF1or in the Clara and Type II cells made the mice more susceptible to injury (Fig. 4.23). Specific cellularity within the BALF was also different between the two genotypes. 168 Figure 4.2. Weight change and cell counts from cobalt challenged control and HlF1aA/A mice. Mice were exposed to saline, 30 pg (5 mM) or 60 pg (10 mM) CoClz for 2 week, 5 days/week paradigm. Total animal weight change (A) in control (white bars) and HlF1aA/A (black bars) were calculated by subtracting animal weight on the day of sacrifice from weight at the start of exposure. Total cell counts from BALF (B) were assessed from all of the mice and averaged. Cell differential counts were performed for macrophages (C), lymphocytes (D), neutrophils (E), and eosinophils (F). N>5 mice/group. * = Significance (p values noted). 169 . , an TI; .1 ,,, 8 31: g 1001) > d . f0 .5 '5. 3 2 g redo > d e g :3 '8 LL] 5 .‘s’ IOO'O 3" d t 75 m :1. ' In ' In ' Z-t ' (you) rw Jed srrad LI- 2 8 .2 '5 in 3 U = o '5 “g. ‘“ r“? a g .5 ".5: (son) rw Jad 51183 N 7' (,(nx) nu 13d snag I.“ a: “:2 o 21'; I ‘° ° $9 3 ‘6 on o g. 3' I m g 8 .c .‘s’ g- 7; U - I. - f” 3 =- e-q O H Vt + 0 sun afiue Bra 1 a) "3 W ° M (you) rw Jad Suez) o < 170 Cobalt-treated control and HlF1ch/A mice showed dose-dependent increases in lymphocytes and macrophages, however, only HlF1aA/A mice treated with 60 pg cobalt was significant when compared to saline (macrophages and lymphocytes) and 30 pg (macrophages only) treated HlF1aA/A mice (Figs. 4.2C and 4.2D). Moreover, the 60 pg treated HlF1aA/A mice displayed a significant increase in macrophages as compared to control mice treated with 60 pg cobalt. Lymphocytes and neutrophils displayed no significant changes when control and HlF1aA/A mice were compared within treatment groups. Control mice displayed a non-significant increase in neutrophils compared to HlF1or deficient mice (Fig. 4.2E). In contrast, the HlF1aA/A mice following cobalt (60 pg) exposure displayed a significant increase in eosinophils when compared to all other groups, suggesting that HlF1a plays a role in modulating the lung’s inflammatory response to metals (Fig. 4.2F). These changes were specific to the loss of HlF1or and not due to Dox treatment or the SPC-rtTA or tet-Cre transgenes as different doxycycline treated monotransgenic and bitransgenic (SP-C-rtTA'ltg/(tetOh-CMV-Cre"'/HIF10+” (sTH), SP-C-rtTA'l' lltetOh—CMV-Cretg/tg/HlF1q+/+ (StH), SP-C-rtTA'ItQ/(tetOh-CMV- Cretg/tg/HIF1a+/+ (stH) and SP-C-rtTA'I'AtetOh-CMV- Cre"'/Hrr=1a“°"’"°" (STh)) mice behaved similar to control mice (SP-C-rtTA'ltg/(tetOh-CMV— Cret9“9/HIF1a"°"’"°" (sth) (Fig. 4.3). 171 Table 4.1. List of genes analyzed qRT-PCR Gene Symbol Gene Description TaqMan Assay ID 188 188 4352930E Actb Beta Actin 4352933E Arbp acidic ribosomal phosphoprotein P0 Mm01974474 gH Gapd Glyceraldehyde 3phosphate dehfliggenase 4352932E Gusb Muronidase, beta Mm00446953_m1 Alf4 activating transcription factor 4 Mm00515324_m1 Cat catalase Mm00437992__m1 CcI2 Monocyte chemotactic protein-1 Mm00441242_m1 Ccl3 Macrophage inflammatory protein 1 alpha Mm00441258_m1 Ccl4 Macrophageinflammatory protein 1 beta Mm00443111_m1 Regulated upon activation, normal T cell expressed and 00/5 secreted Mm01 30242 7_m1 Ccl1 1 Eotaxin Mm00441238_m1 Chi3/3 chitinase 3-like 3 (YM1) Mm00657889_mH Chi3l4 chitinase 3-like 4 (YM2) Mm00840870_m1 Clca3 chloride channel calcium activated 3 (Gob-5) Mm00489959_m1 Crp C-reactive protein, pentraxin-related Mm00432680 g1 Cxcl1 Cxcl1 keratinogtte chemoattractant Mm00433859_m1 Cxcl2 Cxcl2 macrophage-inflammatory protein-2 Mm00436450_m1 Cyp291 cytochrome P450, family2, subfamily e, polypeptide1 Mm00491127_m1 Efl epidermal flwth factor receptor Mm00433023_m1 F2r coggulation factor II (thrombin) receptor Mm00438851_m1 (ng fibroblast growth factor 9 Mm00442795_m1 fgf10 fibroblast growth factor 10 Mm00433275_m1 fgf18 fibroblast growth factor 18 Mm00433286_m1 Foxa1 forkhead box A1 Mm00484713_m1 Foxa2 forkhead box A2 Mm00839704_mH Foxp3 forkhead box P3 Mm00475165_m1 Gata3 GATA binding protein 3 Mm00484683;m1 Gclc flamate-cysteine ligase, catalytic subunit Mm00802655_m1 Gclm glutamate-cysteine ligase, modifer subunit Mm00514996_m1 Gstk1 glutathione S-transferase kappa 1 Mm00504022_m1 Gstm1 glutathione S-transferase, mu 1 Mm00833915 g1 Gstp1 glutathione S-transferase, pl 1 Mm00496606_m1 Hmox heme oxygenase (decycling) 1 Mm00516004_m1 [er9 interferon gamma Mm00801778_m1 IL 1b interleukin 1 beta Mm00434228_m1 Il1rn interleukin 1 receptor antagonist Mm01337566_m1 IL2 interleukin 2 Mm00434256_m1 IL4 interleukin 4 Mm00445259_fl1 IL5 interleukin 5 Mm00439646_m1 IL6 interleukin 6 Mm00446190_m1 IL10 interleukin 10 Mm00439616_m1 lL-13 interleukin 13 Mm00434204_m1 172 Table 4.1 . Continued. Gene Symbol Gene Description TaqMan Assay ID IL23a interleukin 23, alpha subunit p19 Mm00518984_m1 Mt1 metallothionein 1 Mm00496660 g1 Muc5ac mucin 5, subtypes A & C, tracheobronchial/gastric Mm01276725 g1 Nfe2l2 nuclear factor, erythroid derived 2, like 2 Mm00477784_ m1 Nos2 nitric oxide synthase 2, inducible Mm00440485_m1 Nqo1 NAD(P)H dehydrcgenase, quinone 1 Mm00500821_m1 Pcna proliferating cell nuclear antigen Mm00448100 g1 Pdgfrb platelet derived growth factor receptor, beta polypeptide Mm01262489_m1 Ppari peroxisome proliferator activated receptor gamma Mm00440945_m1 Pth prostaglandin-endoperoxide synthase 2 Mm00478374_m1 Saa3 serum amyloid A3 Mm00441203_gi1 chflm secretglobin, family 1A, member 1 (uteroglobin) Mm00442046_m1 Serpine1 serine (or cysteine) peptidase inhibitor, clade E, member 1 Mm01204469_m1 Sftpc surfactant associated protein C Mm00488144_m1 Sfipd surfactant associated protein D Mm00486060_m1 Socs3 suppressor of cytokine signaling3 Mm00545913_s1 Sod1 superoxide dismutase 1, soluble Mm01344233 g1 Sod2 superoxide dismutase 2, mitochondrial Mm00449726_m1 Stat6 signal transducer and activator of transcription 6 Mm01160477_m1 Tmiz tissue factor pathway inhibitor 2 Mm00436948_m1 Tgfb1 transforminggrowth factor, beta 1 Mm01178820_m1 Tnfa tumor necrosis factor(TNF superfamileember 2) Mm00443258_m1 Vegfa vascular endothelial growth factor A Mm01281447_m1 173 Figure 4.3. Cell counts from genotype and treatment controls. Genotypic control mice were exposed to saline or 60 pg (10 mM) CoCIz for 2 week, 5 days/week paradigm. Total cell counts from BALF (A) were assessed from all of the mice and averaged. Cell differential counts were performed for macrophages (B), neutrophils (C), lymphocytes (D), and eosinophils (E). N>5 mice/group. "Images in this dissertation are presented In color.” 174 Figure 4.3. Cell counts from genotype and treatment controls. U) _ d.) ; U 0 S D. E ~ 3 (h 0 on (B .C Q N O L U .— (D 2 QWNQ'Q"! cmomcm HHHOOO MNNHH m (orxlsrraa Q lorxlsuaa s v 1’ a a ‘—’ E g e '- ‘5 0) Z mcmom omom «soil-is" ”NH” (vorx) sued 175 sth StH STH Genotype sTH stH stH sTH sth StH STH Genotype Figure 4.3. Continued. I11 Cells(X104) H N W h In 01 \l Doxy Cobalt (60pg) EI- .- u- + l:l+ - r:1+ + S-no hSPC-rtTA transgene (WT) s- hSPC-rtTA transgene/+ T-notet-cre transgene (WT) t- tet-cre transgene present H-WT HlFlot allele h- floxed HlFlot allele stH sTH sth StH STH Genotype 176 Histopathology of Cobalt-induced Injury: Histologically, no pulmonary lesions were found in control or HIF10tA/A mice that were instilled with saline alone. In contrast, control mice repeatedly instilled with cobalt had a mild- moderate chronic bronchopneumonia that was histologically characterized by a mononuclear cell infiltrate (mainly small and large lymphocytes, monocytes, and occasional plasma cells) admixed with lesser numbers of neutrophils and eosinophils in the interstitial tissues surrounding large- and small-diameter conducting airways (i.e., axial, preterminal and terminal bronchioles) and extending into the centriacinar regions of the lung (alveolar ducts and adjacent alveoli) (Figs. 4.4A and 4.48). Interstitial fibrosis was a prominent remodeling feature of the alveolar septa in affected parenchymal regions along with minimal to mild hyperplasia of alveolar type II cells and accumulation of mildly hypertrophic macrophages and varying numbers of inflammatory cells (lymphocytes and neutrophils) in alveolar airspaces (Figs. 4.4C and 4.40). These cobalt-induced airway and alveolar changes were dose-dependent and were more consistently found in the hilar rather than the distal aspects of the lunglobe. A similar mild-moderate chronic bronchopneumonia with peribronchiolar lymphoplasmacytic inflammation and variable amounts of interstitial alveolar fibrosis airway was present in the lung lobes of cobalt-exposed HlF1aA/A mice (data not shown). There were, however, marked differences in the character 177 Figure 4.4. Histopathology and picro-sirius staining control and cobalt-treated control mice Light photomicrographs of the lungs of control mice instilled with saline (A and C) or cobalt (B and D). Lung sections in A and B were stained with hematoxylin and eosin, while sections in C and D were histochemically treated with picro-sirius red solution that stains interstitial collagen (red chromagen in interstitial tissues in the alveolar septa and around bronchiolar airways). ln cobalt-treated lungs (B and D), there is marked inflammation and interstitial fibrosis (asterisk in B) around pre-terrninal bronchioles (ptb) and terminal bronchioles (tb) and extending distally into alveolar ducts (ad) and adjacent alveoli (a). There is increased picro-sirius red stained collagen (solid arrows) in the alveolar septa of the cobalt-treated mouse (D) compared to that in the saline-treated control mouse (C). p, pulmonary pleura; e, bronchiolar epithelium; stippled arrow, mixed inflammatory cell infiltrate. "Images in this dissertation are presented in color.” 178 179 of the inflammatory, airway epithelial, and alveolar macrophage changes in the lungs of these transgenic mice compared to their control counterparts. Along with the lymphocytes and plasma cells, there were markedly more eosinophils in the peribronchiolar and alveolar mixed inflammatory cell infiuxes in HIF10rA/A mice compared to those of control mice (Fig. 4.5D). Another distinctive change found only in HlF1orA/A mice was mucous cell metaplasia in the airway epithelium lining axial and preterminal bronchioles. This airway epithelial lesion in large-diameter bronchioles consisted of numerous Alcian Blue/Periodic Acid Schiff stained mucous goblet cells in bronchiolar epithelium that is normally devoid of these mucus-secreting cells in mice (Figs. 4.6A and 4.6B). These areas of epithelial mucous cell metaplasia in cobalt-treated HlF1orA/A mice were also lmmunohistochemically positive for YM1/2 chitinase- like proteins (Figs. 4.60 and 4.60). Characterization of eosinophils and YM1/2 Expression: In addition to these unique eosinophil and epithelial responses, alveolar macrophages accumulating in alveolar airspaces of cobalt-exposed mice were larger and more eosinophilic in the HIF10rA/A mice compared to those in similarly exposed control mice (Figs. 4.7A and 4.78). lmmunohistochemically, the cytoplasm of these phenotypically distinctive macrophages stained positive for YM1/2 protein similar to the mucous goblet cells in the metaplastic bronchiolar epithelium of HIF1dA/A mice (Figs. 4.60 and 4.60). 180 Figure 4.5. Major Basic Protein Staining in lungs from control and HlF1aA/A mice. Light photomicrographs of the lungs of saline (A and C) or cobalt- instilled (B and D) control (A and B) and HlF1aA/A (C and D) mice. All lung sections were immunohistochemically stained for major basic protein to identify infiltrating eosinophils (red chromagen; arrows) and counterstained with hematoxylin. A mixed inflammatory infiltrate consisting of mononuclear leukocytes, eosinophils and lesser numbers of neutrophils are restricted to the lungs in cobalt-treated mice (B and D). Markedly more eosinophils are present in the peribronchiolar and alveolar regions of the cobalt-treated HIF1or A/A mouse (D) compared to that of the cobalt-treated control mouse (B). ptb, pre-tenninal bronchiole; pa, pulmonary arteriole, a, alveolar airspace; e, bronchiolar epithelium. "Images in this dissertation are presented in color.” 181 reef W5“: nexfk tW9 w... .o‘ 182 Variably sized needle-shaped or rectangular refractile eosinophic crystals were also present within these alveolar macrophages or free in the alveolar airspaces (Fig. 4.73). These crystals also stained positive for YM1/2 (Fig. 4.7D). Multinucleated giant cells were also more frequently observed in the cobalt-induced alveolitis of HlF1aA/A mice compared to those of control mice. Cobalt-induced gene expression changes: The differences in cobalt- induced pulmonary pathology between the control and HlF1aA/A suggested an alteration in the stress response upon loss of HlF1a. To begin characterizing this difference, the expression of 63 key genes involved in immunity, inflammation, oxidative stress, and other stress pathways, was assessed by quantitative real time PCR (qRT-PCR) (table 4.1). Initially, samples from each treatment group and genotype were pooled and screened. Those genes that showed a difference when compared to untreated controls within a genotype or between genotypes were characterized as individual samples (table 4.2). Six of these genes showed significant changes in expression when compared between genotype and within treatment or within genotype and between treatments (Fig. 4.8). Two other genes, Ym1 and IL6, were near significance (0.069 and 0.055 respectively). The expression of Ym2 in the HlF1or deficient mice was significantly increased following challenge with 60 pg cobalt compared to the saline treated HlF1aA/A mice and the 60 pg cobalt treated control animal. 183 Figure 4.6. Alcian Blue/Periodic Acid Schiff Stain and YM1/2 IHC Light photomicrographs of pre-terrninal bronchioles (ptb) of cobalt- lnstilled control (A and C) and HlF1aA/A (B and D) mice. Tissues were stained with Alcian Blue (pH 2.5)lPeriodic Acid Schiff (AB/PAS; A and B) to identify acidic and neutral mucosubstances (magenta stain; arrows) in mucous cells within the bronchiolar epithelium (e). Numerous AB/PAS-stained mucous cells are present only in the airway epithelium lining the pre-terrninal bronchiole in the cobalt-treated HlF1aA/A mouse (B). Tissues in C and D were immunohistochemically stained for YM1/2 protein (brown chromagen; arrows) and counterstained with hematoxylin. YM1/2 proteins were present only in the bronchiolar epithelium of the cobalt-treated HlF1aA/A mouse (D). Arrow in C identifies a few alveolar macrophages that were positive for YM1/2 proteins. "Images in this dissertation are presented ' in color." 184 \ A A. r . 332.94... ,: a ...I. 7...? . til . . ...v. 185 Figure 4.7. H&E staining and YM1/2 IHC of cobalt treated control and HIF1orA/A mice Light photomicrographs of alveolar macrophages in cobalt-instilled control (A and C) and HlF1aA/A (B and D) mice. Lung tissues were histochemically stained with hematoxylin and eosin (H&E; A and B) or immunohistochemically for YM1/2 proteins (C and D). H&E-stained alveolar macrophages in the cobalt-instilled HlF1aA/A mouse (B) are larger (hypertrophic) and more eosinophilic than the similarly stained lung section from the cobalt-instilled control mouse (A). In B, a group of macrophages are surrounding an extracellular aggregate of eosinophilic crystals (arrow). In addition, more alveolar macrophages with immunohistochemically staining for YM1/2 proteins (solid arrows) are present in lung section from the cobalt-instilled HlFlor A/A mouse (D) as compared to that of the cobalt-instilled control mouse (C). Stippled arrow in C, alveolar macrophage with no detectable YM1/2 proteins. "Images in this dissertation are presented in color." 186 187 Gob5 expression followed a similar pattern of expression as that of Ym2; however, its expression was only significantly changed when the 60 pg cobalt treated HlF1orA/A animals were compared to the 30 pg treated ones. Muc5ac and IL5 showed dose dependent changes in expression within genotypes and in the case of lL5, expression was significantly elevated in saline treated HlF1crA/A mice compared to saline treated controls. Eotaxin was also significantly elevated in the HIF1orA/A mice when the saline treated mice were compared. Moreover, eotaxin was also significantly different between the genotypes in the 60 pg cobalt treated mice. Cytokine profiling: The pathology of the lung and the gene expression patterns suggested a change in the inflammatory response upon loss of HlF1afrom Type II and Clara cells. To determine if the changes in cytokine gene expression led to changes in the chemoattractants found in the BALF, profiling of 18 cytokines was performed using a bead array (BioRad). Of the 18 characterized, 10 showed significant difference when compared across genotype or within treatment groups (Table 4.3). Interestingly, several of these were different when the saline treated groups of the control and HlF1aA/A mice were compared (i.e. lL-1B, lL-5, lL-12, GM-CSF, RANTES, TNFa and VEGF [Table 4.3]). This suggests that loss of HIF1a from Type II and Clara cells alters the tissue's native cytokine profile. Moreover, cobalt exposure caused a more pronounced phenotype in the HIF1aA/A animals with respect to cytokine changes. 188 Figure 4.8. Gene expression results The expression of 63 genes was analyzed by qRT-PCR. Those genes that showed a difference as pooled samples were further analyzed as independent replicates. White bars represent control groups and black bars represent HIF1or deficient groups. Expression levels were normalized to the saline treated control animal and expressed as fold change. a = p < 0.07, * = p < 0.05, ** =p<001 189 5 Ym2 I——l=_.lj 15 at: at: :1: F j 0 124 -5 9- -10 6. -15 3. -20 o. 10 Gob5 * l—'—‘| 1o. IL5 0 8- -1o 6. -20 4. -30 -4o 2' -50 0- Ym1 F E 5' HmOx 4. a 4 3- 3 ar: 2- 2 1- 1 o. 0 5- MuCSac 4 Eotaxin 4- 3 3‘ ** 2 * ** 2 1 0 o. Saline 30 pg 60 pg Saline 30 pg 60 pg COCIZ COCIZ COCIZ COCIZ Table 4.2 Gene Expression Changes Control HIF1 aA/A Gene Saline 30 L 60 gg Saline 30 pl 60 pg__ 1.00 +I- 1.04 +I- 1.06 +I- 1.15 +I- 1.07 +I- 1.02 +I- 185 0.04 0.03 0.04 0.05 0.02 0.03 1.00 +l- -1.14 +I- -1.13 +I- -1.05 +I- -1.08 +l- -1.06 +I- B-Actin 0.05 0.05 0.03 0.02 0.04 0.02 1.00 +I- 1.08 +I- 1.05 +I- -1.10 +I- -1.01 +I- 1.02 +I- GAPDH 0.05 0.07 0.03 0.05 0.04 0.02 1.00 +I- 1.70 +I- 1.18 +I- 2.38 +I- 1.10 +I- -1.62 +I- MCP 0.35 0.96 0.48 1.03 0.27 0.40 1.00 +I- 1.79 +I- 2.40 +I- 2.39 +I- 2.19 +I- 2.43 +I- MIP1a 0.21 0.46 0.62 0.56 1.83 0.34 1.00 +l- 1.42 +I- 1.15 +I- 1.88 +I- 1.49 +I- 2.52 +I- Eotaxin 0.08 0.23 0.15 0.23 0.10 0.36 1.00 +l- 1.21 +I- 1.88 +I- 1.56 +I- 3.00 +I- 3.38 +I- Ym1 0.25 0.36 0.51 0.62 0.42 0.41 1 .00 +l- -5.88 +l- -3.36 +I- -4.46 +l- 2.04 +I- 2.82 +I- Ym2 0.80 3.99 1.82 2.58 1.63 0.63 1.00 +I- -34.02 +l- -6.91 +I- -2.30 +I- 2.19 +I- 2.03 +I- GOBS 0.60 16.99 2.93 1.54 1.83 0.75 1.00 +I- 2.82 +I- -1.11 +I- 2.92 +I- 2.21 +I- 1.40 +I- KC 0.45 1.73 0.17 1.03 1.02 0.26 1.00 +I- 1.88 +I- 2.72 +I- 1.02 +I- 2.87 +I- 2.08 +I- HmOx 0.19 0.48 0.34 0.09 0.54 0.16 1.00 +I- 1.55 +I- 3.37 +I- -1.90 +I- -1.51 +l- -2.34 +I- IFN'y 0.48 0.82 2.32 0.41 0.40 0.29 1.00 +I- 1.30 +I- -2.50 +I- 1.24 +I- -1.16 +I- -1.69 +I- IL1B 0.43 0.32 0.41 0.30 0.22 0.62 1.00 +I- -1.03 +I- 1.31 +I- 1.05 +I- 1.34 +I- 1.27 +I- lL4 0.22 0.14 0.17 0.28 0.40 0.24 1.00 +I- 1.60 +I- 1.09 +I- 8.44 +I- 3.71 +I- 2.02 +I- lL5 0.1 9 0.67 0.46 3.02 1.36 0.23 1.00 +I- 2.43 +I- 1.29 +I- 1.93 +I- 1.86 +I- 2.13 +I- lL6 0.48 1.22 0.19 0.37 0.31 0.50 1.00 +I- 1.11 +I- 1.89 +l- -2.65 +I- -1.46 +I- -1.26 +I- IL10 0.67 0.51 1.23 0.47 0.50 0.32 1.00 +I- -1.73 +I- -1.18 +I- 1.34 +I- 1.40 +l- 2.46 +I- lL13 0.41 0.22 0.37 0.34 0.44 0.51 1.00 +I- 1.87 +I- 3.08 +I- 1.56 +I- 1.98 +I- 3.36 +I- MUC5ac 0.24 0.39 0.50 0.27 0.23 0.41 1.00 +I- 1.91 +I- 2.59 +I- -1.23 +I- -1.01 +I- 1.15 +I- iNOS 0.14 0.72 1.49 0.22 0.18 0.12 1.00 +I- 1.46 +I- 3.47 +I- -1.18 +I- -1.17 +I- 1.09 +I- SAA3 0.73 0.69 2.13 0.17 0.41 0.58 1.00 +I- -1.10 +I- 1.40 +I- -1.01 +I- 1.72 +l- 1.48 +I- SPC 0.22 0.23 0.29 0.22 0.19 0.05 1.00 +I- 1.48 +I- 2.11 +l- 1.21 +l- 1.07 +I- 1.02 +I- TNFa 0.17 0.28 0.70 0.11 0.17 0.13 1.00 +I- -1.10 +I- -1.47 +l- 1.13 +I- -1.31 +I- -1.51 +I- VEGF 0.08 0.12 0.24 0.13 0.05 0.14 191 DISCUSSION: The research describes a model for the post-natal deletion of HlF1or from Clara and Type II cells of the lung with the intention of determining the role of the transcription factor in cobalt-induced injury. Control mice repeatedly exposed to cobalt via aspiration had classic signs of metal-induced injury, including increased neutrophils, peribronchial inflammation, and fibrotic lesions. In contrast, the mice deficient in HIF1or in their Clara and Type II cells exhibited a Th2-mediated response in response to cobalt challenge. In fact, these HlF1dA/A mice displayed a pathology that can be described as asthma- like, including increased eosinophilic infiltration, increased chitinase-like protein expression, and mucus cell metaplasia. In addition, these HlF1or deficient mice expressed an altered panel of cytokines in the absence and presence of cobalt. These results suggest that alveolar epithelium-derived HlF1a plays a critical role in determining the inflammatory response to cobalt challenge and loss of this critical transcription factor can alter the lung’s ability to cope with the toxicant insults. Attempts to study cobalt-induced injury in animal models have successfully mimicked some facets of hard metal lung disease (HMLD). For example, rabbits exposed to low doses of cobalt chloride (0.4 and 2 mg/m3) for 14-16 weeks (6 hrs/day) displayed increased macrophages and lysozyme activity in BALF, interstitial inflammation, and the presence of large vacuolated macrophages [20-22]. 192 Table 4.3. Cytokine Profiles Control and HlF1aA/A mice were exposed to saline, 30 pg, or 60 pg CoCl2 for 2 week, 5 days/week paradigm. The levels of cytokines in BALF were assessed using the Bio-Rad Bio-Plex cytokine suspension array system. N>5 mice/group. Yellow = p<0.05, when HlF1a A/A saline is compared to control saline. Blue = p<0.05 compared to genotype matched control. ** = p<0.05 when similar treatments are compared between genotypes. Black shade = significance at 0.05 when compared to saline control between genotypes # = significance at 0.05 when compared to saline treated mice within genotype ** = significance at 0.05 when compared between _*genotypes and within treatment group 193 .nj 00.8 -1 $.00 00.0 -1 00.8 $.00 -1 8.8 3R -1 ta... 8.0. -1 $.00 50 -1 0......- 2.: 080.0 -1 00.0 0000.0 -1 8.0 8.0 .1 00.0 8.0 .1 00,0 00.0 -1 00.0 35 8.0 -1 8.0. .00 -1 B... . . 0.80.0 -1 02.. 0.0.0.. -1 8.8 8.. -1 8.0 0.0-.25. 00.. .1 8...... .00 .1 8.0 :0 -1 00.0. 0.... -1 8.0 00.0 -1 00.. 00.0 -1 00.0 .32 4.38.00 -1 0.0.. 00.. -1 00.0 «0.0 .1 00.00 8.0 -1 8.8 00.0. .1 0.0.. 04.0 -1 0.. .0 9. 00.0 -1 00.. 8.0 -1 I. .0. -1 8.0 8.0 -1 0.0 8.0 -1 00.. 04.0 -1 00.0 .2... 00.0 .1 A... $0 -1 00.0 . 00.. -1 8.0 00.0 .1 8.. 00.0 -1 00.0 emu-Eu 8.0 -1 8.0 8.. .1 00.0 00.0 -1 00.0 8.0 -1 0. .0 8.0 -1 0.0 8.0 -1 0.0 00.38 00.0 .1 00.. 0.0 .1 2.0 80 .1 8.. .00 -1 1.. a0. .0 -1 00.0 «0.0 -1 00.. 3.... 0.0 -1 00.. 8.0 -1 8.. 00.0 -1 00.0 00.0 -1 8.0 8.0 -1 00.0 NS. 00.0 -1 0.0 «m0 -1 00.0 00.0 .1 00.0 8.0 -1 0.0 8.0 -1 0.0 00.0 -1 00.0 03. 0.0 -1 03 8.0 -1 8.0 00.0 -1 .00 00.0 -1 00.0 5.0 -1 00.. .00 -1 8.. 0.0. .080 -1 00.0 4.3 -1 00.0 .0 0.11 00 0. 00.0 -1 8.0 00.. .1 00.0 00.0 -1 00.0 3. .Jv 8.0 -1 00.0 11...... -1 :0 8.0 .1 00.01 8.0 -1 00.0 00.0 -1 00.0 00.0 -1 8.0 4.... 00.0 -1 8.0 00.0 .1 00.0 00.0 .1 8.0 8.0 -1 00.0 00.0 -1 00.0 00.0 -1 00.0 3. 0.8.0 -1 8.0 8.0 -1 8.0 .0 m -1 8 . 00.0 -1 8.0 00.0 -1 00.0 3% a a: 8 u: 8- n: 8 01 on 20.8 $482.: _ .288 194 Guinea pigs exposed to 2.4 mg/m3 of cobalt chloride for 2 weeks (6 hrs/day) had a higher rate of BALF neutrophilic and eosinophilic infiltration when presensitized to the metal [23]. The National Toxicology Program exposed rats and mice to cobalt sulfate heptahydrate at doses ranging from 0.3 to 30 mg/m3 for 13 weeks (6 hrs/day) and described interstitial fibrosis, epithelial hyperplasia, and lesions in the upper airways that were more pronounced in the rats [24, 25]. Finally, hamsters instilled with cobalt chloride (1-1000 pg/kg) displayed signs of oxidative stress, including increased GSSG (oxidized glutathione) :GSH (reduced glutathione) ratio [26]. To the best of our knowledge, none of these models described chronic eosinophilic infiltration, mucous cell metaplasia, or the increase in chitinase-like proteins as seen in the HlF1aA/A mice. It has been suggested that an animal model is needed that combines the toxic properties of cobalt with its allergenic potential [11]. The HlF1aA/A mice might be this model. In the presence of cobalt, HlF1aA/A mice respond with an asthma-like phenotype (i.e. eosinophil infiltration and mucus cell metaplasia) and increased expression of the chitinase-like proteins, Ym1 and Ym2. Chitins are acetylated glucosamine biopolymer not found in mammalian systems and chitinase and chitinase-like proteins are believed to function as protection against chitin containing organisms. Ym1 and Ym2 are chitinase- like proteins that share considerable protein homology to the human protein, 195 YKL-40 (also known as human cartilage glycoprotein 39 and chitinase 3-like 1). YKL-40 has recently been identified in the lungs and circulation of asthmatics [27]. Not only was there a correlation with the presence of YKL-40 and asthma, the levels also correlated with the severity of the disease and the thickening of the subepithelial basement membrane [27]. The biological relationship between these enzymes and the etiology of asthma has not been characterized. What is known, however, is that chitinase and chitinase-like proteins are upregulated in asthma models and in the case of chitinase proteins, this induction is dependent upon IL13. IL13 gene expression is slightly elevated in the cobalt treated HlF1aA/A mice and its lack of significance is most likely due to the timing of analysis (Le 14 days after the start of exposure). Increased chitinase expression is required for the subsequent eosinophilia and lymphocytic infiltration [28]. Ym2 was significantly increased in the 60 pg treated HIF1orA/A mice when compared to the 60 pg treated control mice. Moreover, Ym1 expression was approaching significance (p < 0.07) when a similar comparison was made. In contrast, Ym2 show a cobalt-induced decrease in expression in the control animals, and Ym1 is unchanged by cobalt treatment in these control mice. These results suggest that cobalt exposure in the HlF1aA/A mice induces a series of events within the lungs that resembles the progression of asthma in other asthma models, including increased IL13, subsequent increases in Ym1 and Ym2 expression, and the recruitment of eosinophils. 196 The results suggest that the ability of Type II and Clara cells to respond to hypoxia is necessary for the proper lung inflammatory response to metal- induced stress. It is important to point out that this response is not due to loss of HlF1or in resident macrophages. HlF1or expression is still strong in these cells following post-natal doxycycline exposure and confirms that the SPC promoter confines the expression of cre recombinase to Type II and Clara cells (data not shown). In addition, it implies that the observed inflammatory response is not due to the previously established role of HlF1a in myeloid cell mediated inflammation [29]. Taken together, the results suggest a model for the inflammatory differences between the control and HlF1a deficient mice following metal exposure. Initially, the cobalt challenge leads to damage within the lung, either at Type I or II cells. This damage is communicated to the remaining viable Type II cells of the parenchyma directly or through resident macrophages. In control lungs, HlF1a will regulate the expression of classic hypoxia target genes as well as others involved in the inflammatory response. This HIF1-regulated transcription will dictate the lung’s inflammatory response to the metal challenge. The modest increases in cytokines, such as IFNy, and the resulting pathology suggest a Th1 mediated response. Loss of HlF1cr in the remaining viable Type II cells of the HlF1aA/A mice would lead to an alteration in the cell’s ability to respond to the incoming signals from the cobalt damaged cells. The decreased levels of HIF1a would compromise the lung’s ability to respond to cobalt as a toxicant and putative allergen. Ultimately, this difference in epithelium derived HlF1or alters the 197 pattern of released cytokines and presumably leads to the differential expression of Th2 chemokines, such as IL4, IL5, IL10, and IL13. These changes in cytokines will promote the expression of the chitinase and chitinase-like proteins, such as Ym2 and this would lead to the eosinophilic infiltration seen in the HlF1dA/A mice following cobalt challenge (Fig. 4.7). This proposed model explains the difference in the inflammatory response between the control and HlF1aA/A mice; however, it is based on collected data at the end of cobalt exposure. Validation of the model will require a detailed dose and time course response to cobalt challenge. The striking differences observed following cobalt exposure in these two mice genotypes suggests that they will be a powerful tool to understand the relationship between allergy-induced asthma, hypoxia, and inflammation. More importantly, direct comparison of the responses of the control and HlF1dA/A mice in other asthma models (e.g. ovalbumin challenge) and to other inflammatory inhalants (e.g. ozone) will correlate this relationship to specific cytokines. The research also raises several important questions: Does the loss of HIF1or alter an organism’s susceptibility to asthma using other allergens? What role does HlF1or derived from other cell types (e.g. infiltrating inflammatory cells and Type I cells) play in modulating this inflammatory response? Does post-natal deletion of HlF1a alter HlF2a and if so, what impact does this have on the inflammatory response [15]? Most importantly, does a decrease in HlF1or functionality lead to an increased 198 susceptibility to asthma in humans? More specifically, is it possible that loss or decrease in HlF1or function biases the lung towards a Th2 immune polarization and this increases the susceptibility of individuals towards extrinsic asthma? Even with these unanswered questions, the HlF1aA/A mice offer an important step forward In understanding the role of the HlF1a signaling cascade in allergy-induced asthma. Finally, it established epithelial- derived HIF1or as a major regulator of the lung’s response to allergenic compounds and creates a new tool to help understand the relationship between inflammation and asthma progression. 199 REFERENCES 1 Bunn, H. F. and Poyton, R. O. (1996) Oxygen sensing and molecular adaptation to hypoxia. Phys. Rev. 76, 839-885 2 Epstein, A. C., et al. (2001) C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell. 107, 43-54. 3 Salnikow, K., et al. (2004) Depletion of intracellular ascorbate by the carcinogenic metals nickel and cobalt results in the induction of hypoxic stress. J Biol Chem. 279, 40337-44. 4 Vengellur, A., Phillips, J. M., Hogenesch, J. B. and LaPres, J. J. (2005) Gene expression profiling of hypoxia signaling in human hepatocellular carcinoma cells. Physiol. Genomics. 22, 308-318 5 Lombaert, N., Lison, D., Van Hummelen, P. and Kirsch-Volders, M. (2008) In vitro expression of hard metal dust (WC-Co)-responsive genes in human peripheral blood mononucleated cells. Toxicol Appl Pharrnacol. 227, 299-312 6 Semenza, G. L., Roth, P. H., Fang, H. M. and Wang, G. L. (1994) Transcriptional regulation of genes encoding glycolytic enzymes by hypoxia-inducible factor 1. J. Biol. Chem. 269, 23757-23763 7 Forsythe, J. A., et al. (1996) Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol. Cell. Biol. 16, 4604-461 3 8 Jung, Y., et al. (2003) Hypoxia-inducible factor induction by tumour necrosis factor in normoxic cells requires receptor-interacting protein- dependent nuclear factor kappa B activation. Biochem J. 370, 1011- 1017. 9 Mojsilovic—Petrovic, J., et al. (2007) Hypoxia-inducible factor-1 (HIF-1) is involved in the regulation of hypoxia-stimulated expression of monocyte chemoattractant protein-1 (MCP-1/CCL2) and MCP-5 (Ccl12) in astrocytes. J Neuroinflamm. 4, 12 200 ‘IO 11 12 13 14 15 16 17 18 19 20 Rius, J., et al. (2008) NF-kappaB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1alpha. Nature. 453, 807-81 1 Lison, D., Lauwerys, R., Demedts, M. and Nemery, B. (1996) Experimental research into the pathogenesis of cobalt/hard metal lung disease. Eur Respir J. 9, 1024-1028 Shirakawa, T., et al. (1988) The existence of specific antibodies to cobalt in hard metal asthma. Clin Allergy. 18, 451-460 Gheysens, B., Auwerx, J., Van den Eeckhout, A. and Demedts, M. (1985) Cobalt-induced bronchial asthma in diamond polishers. Chest. 88, 740-744 Shirakawa, T., et al. (1989) Occupational asthma from cobalt sensitivity in workers exposed to hard metal dust. Chest. 95, 29—37 Saini, Y., Harkema, J. R. and LaPres, J. J. (2008) HlF1[alpha] Is Essential for Normal Intrauterine Differentiation of Alveolar Epithelium and Surfactant Production in the Newborn Lung of Mice. J. Biol. Chem. 283, 33650-33657 Ryan, H. E., Lo, J. and Johnson, R. S. (1998) HIF-1 alpha is required for solid tumor formation and embryonic vascularization. EMBO Journal. 17, 3005-3015 Ryan, H. E., et al. (2000) Hypoxia-inducible factor-1 alpha is a positive factor in solid tumor growth. Cancer Res. 60, 4010-4015 Perl, A. K., et al. (2002) Early restriction of peripheral and proximal cell lineages during formation of the lung. Proc Natl Acad Sci U S A. 99, 10482-10487. Lobe, C. G., et al. (1999) Z/AP, a double reporter for cre-mediated recombination. Dev Biol. 208, 281-292. Johansson, A., et al. (1986) Rabbit alveolar macrophages after long- term inhalation of soluble cobalt. Environ Res. 41, 488-496 201 21 22 23 24 25 26 27 28 29 Johansson, A., Robertson, B. and Camner, P. (1987) Nodular accumulation of type II cells and inflammatory lesions caused by inhalation of low cobalt concentrations. Environmental research. 43, 227-243 Johansson, A., Camner, P., Jarstrand, C. and Wiemik, A. (1983) Rabbit alveolar macrophages after inhalation of soluble cadmium, cobalt, and copper: a comparison with the effects of soluble nickel. Environ Res. 31, 340-354 Camner, P., et al. (1993) Inhalation of cobalt by sensitised guinea pigs: effects on the lungs. Br J Ind Med. 50, 753-757 Bucher, J. R., et al. (1990) Inhalation toxicity studies of cobalt sulfate in F344/N rats and BGC3F1 mice. Fundam Appl Toxicol. 15, 357-372 Bucher, J. R., et al. (1999) Inhalation toxicity and carcinogenicity studies of cobalt sulfate. Toxicol Sci. 49, 56-67. Lewis, C. P., Demedts, M. and Nemery, B. (1991) lndices of oxidative stress in hamster lung following exposure to cobalt(ll) ions: in vivo and in vitro studies. Am J Respir Cell Mol Biol. 5, 163-169 ChUpp. G. L., et al. (2007) A Chitinase-Iike Protein in the Lung and Circulation of Patients with Severe Asthma. N Engl J Med. 357, 2016- 2027 Zhu, 2., et al. (2004) Acidic mammalian chitinase in asthmatic Th2 inflammation and IL-13 pathway activation. Science. 304, 1678-1682 Cramer, T., et al. (2003) HIF-11: Is Essential for Myeloid Cell-Mediated Inflammation. Cell. 112, 645-657 202 Chapter 5 The Role of Hypoxia Inducible Factor 1a (HlF1a) in Modulating Cobalt- lnduced Lung Inflammation: An Acute Study 203 ABSTRACT: Air pollution is a critical factor in the development and exacerbation of pulmonary diseases. Ozone, automobile exhaust, cigarette smoke, and metallic dust are among the potentially harmful pollution components that are linked to disease progression. Transition metals, such as cobalt have been identified at significant levels in air pollution. Cobalt exerts numerous biological effects, including mimicking hypoxia. Similar to hypoxia, cobalt exposure results in the stabilization of hypoxia inducible factors (HIFs). HlFs are a family of proteins that regulate the cellular response to oxygen deficit. HlFs also play an important role in innate immunity and inflammatory processes. Moreover, epithelial-derived HIF signaling has been shown to modulate the lung’s inflammatory response to subchronic exposure to cobalt and biased the tissue towards a Th2-mediated response. To further understand the role of HlF1a, the most ubiquitously expressed HIF, in establishing the lung’s inflammatory response to cobalt, a series of acute studies was performed. An inducible lung- specific HlF1a deletion model was employed to characterize the inflammatory function of epithelial derived HIF in response to inhaled cobalt. Control mice showed classical sign of metal-induced injury following cobalt exposure, including neutrophilic infiltration and induction of Th1 cytokines. In contrast, HlF1or deficient mice exhibited pronounced eosinophil counts in BALF and lung tissue complemented with Th2 cytokine induction. These results suggests that the loss of epithelial-derived HlF1or result in an asthma-like phenotype following 204 acute cobalt exposure and that HlF1or is an important mediator in establishing the lung’s inflammatory response. 205 INTRODUCTION Postnatal lungs require pollution free ambient air for proper function and development. Air pollution is also a critical factor in the occurrence and exacerbation of lung pathological conditions, such as ainNay hypersensitivity, asthma, chronic obstructive pulmonary disease (COPD) and lung cancer. A plethora of airborne particles are injected into the environment through anthropogenic activities. The first line of defense against airborne particles is innate immunity. Various lung diseases, such as asthma and COPD, occur due to detrimental effects of these particles on epithelial integrity, resident macrophages activations and recruitment of inflammatory cells. Airborne pollutants, such as smoke, dust particles, ozone, noxious gases, automobile exhausts, and metals can induce lung injury with a strong inflammatory component. Depending upon the degree of exposure and tissue injury, inflammatory reactions facilitate repair and remodeling processes. Hard metal lung disease (HMLD) and cobalt asthma are occupational respiratory diseases affecting workers involved in the manufacture and maintenance of hard metals (material consisting of tungsten carbide cemented in a matrix of cobalt), diamond polishing, and coal mining. These workers are exposed to cobalt dust and manifest airway constriction, alveolitis, fibrosis and associated giant cell interstitial pneumonitis [1]. The mechanism for the 206 cobalt-induced pathology remains largely unknown, however, several possibilities have been proposed. One of these possibilities is that ability of cobalt to promote a hypoxic-like response in cells. Given the link between hypoxia and inflammation, cobalt-induced hypoxia mimickry offers a logical link between metal exposure and the observed pathologies of HMLD and cobalt asthma [2, 3]. Hypoxia, a decrease in available oxygen reaching the tissues of the body, can influence the processes such as normal cellular homeostasis, repair, and inflammation. The cellular response to hypoxia is regulated by a family of transcription factors called the hypoxia inducible factors (HlFs) [4]. HlFs are primarily regulated at the level of protein stability by a family of prolyl hydroxylases. These prolyl hydroxylase domain (PHDs) proteins are members of a broader family of non-heme, ferrous ion- and 2-oxoglutarate dependent dioxygenases [5]. Upon exposure to decreases in oxygen availability PHDs become inhibited and HlF1or the most ubiquitously expressed isoform of HIFs, becomes stabilized. Once stable, HlF1atranslocates to the nucleus and heterodimerizes with the aryl hydrocarbon receptor nuclear translocator (ARNT, also known as HIF1B) forming the functional transcription factor HIF1. HIF1 regulates the expression of over one hundred genes, including ones involved in energy metabolism, matrix/barrier function, angiogenesis, and inflammation [6-9]. Similar to hypoxia, cobalt has been shown to inhibit PHDs and this inhibition causes very 207 similar transcriptional outputs to that of hypoxia [2, 10, 11]. Recent study in human peripheral blood mononuclear cells has shown similar transcriptional overlap upon tungsten carbide-cobalt particles treatment, linking hard metal lung disease to hypoxia signaling [12]. Recently, using a lung-specific HlF1or deficient mouse model, a compromised HIF signaling system was shown to alter the tissue’s response to subchronic cobalt exposure. The loss of HlF10t from type II and Clara cells was shown to bias the lung’s inflammatory response towards a Th2-mediated process. Moreover, these HlF1a-deficient mice displayed an asthma-like phenotype, including pronounced eosinophil infiltration, mucus cell metaplasia of airway epithelium, and increased levels of the chitinase-like proteins YM1 and YM2 following cobalt challenge. These results suggest that airway epithelial-derived HlF1cr plays a critical role in modulating the inflammatory response of the lung. However, to understand the progression of cobalt- induced lung injury, further investigation on the role of HlF1or in cobalt-induced acute inflammation is required. In the present study, control and HlF1or deficient mice were exposed to cobalt daily for 1, 2, or 5 days. Bronchoalveolar lavage fluid (BALF) cellularity from HlF1ct deficient mice displays a progressive eosinophilic infiltration whereas control mice displayed a transient increase in neutrophils. Histological analysis revealed accelerated tissue injury following acute cobalt challenge in HlF1or deficient mice. Finally, BALF cytokine analysis showed lL-5 elevation specific to cobalt-treated HlF1or 208 deficient mice. In contrast, control mice showed specific induction of lL-6 and tumor necrosis factor or (TNFa) following cobalt treatment. These results suggests that epithelial-derived HlF1or is essential for regulating early inflammatory events following cobalt challenge and loss of this regulation biases the lung towards a Th2 mediated process and an asthma-like pathology following metal exposure. MATERIALS AND METHODS: Description of mice: The mice used in these studies were created by mating HIF1 aflox/fiox (a generous gift of Dr. Randall Johnson, Univ. Califomia-San . -/tg tg/tg . . Diego) and SP-C-rtTA /(tetO)7-CMV-Cre transgenic mice (a generous gifts of Dr. Jeffrey A. Whitsett, Cincinnati Children’s Hospital Medical Center) [13-16]. The generated triple transgenic mice, SP-C-rtTA-Itg/(teton-CMV- t/t flx/fl regg o ox C /HlF1or , are capable of respiratory epithelium specific conditional recombination in the floxed HIF1a gene upon exposure to doxycycline (20). All the mice genotypes used in this study have been maintained in a mixed C57/BL6 and FVB/N background. Genotyping of the mice was performed by PCR for the three loci as previously described [17]. 209 Figure 5.1. Experimental Design. HIF1ch/A mice were generated through postnatal doxycycline treatment paradigm (doxycycline given from PN4 to PN42). Control (n=36) and HIF10rA/A (n=36) male mice randomly assigned to three different treatment groups (24, 48, and 120 hrs). For each time point, mice were challenged with saline-(n=6) or cobalt chloride (60 pg, n=6) via oropharyngeal aspiration. Animals were euthanized 24 hours after their first dose (24 hrs treatment group), second dose (48 hrs treatment group) or fifth dose (120 hrs treatment group). 210 Saline or 60pg CoClz PN4 PN42 8 g E ‘ ‘ fl. 0. O. Doxy PN55 PN56 24 hrs—> 48 hrs —> 120 hrs —> I Euthanize Mice: Harvest Lung Tissue and BALF 211 Doxycycline treatment and animal husbandry: Postnatal recombination was carried out by exposing lactating dams to doxycycline feed (625 mg doxycycline/Kg; Harlan Teklad, Madison, WI) and drinking water (0.8 mg/ml: Sigma chemicals Co.) until weaning. Triple transgenic mice were then maintained on the same food and water until they were approximately 7 weeks of age. In order to eliminate the effects of doxycycline, the treatment was terminated 7-10 days prior to first exposure to metals. These mice will be referred to as HlF1adeficient or HlF1aA/A. Genotypic designation of these -/tg A mice is SP-C-rtTA / (tetOh-CMV-Cretgltg/HIF1orN . Control animals used in the study were of the same genotype that were maintained on normal food and water ad Iibitum. Mice used in this study were kept at the animal housing facility under the strict hygienic and pathogen free conditions approved by the university laboratory animal resource (ULAR) regulatory unit. All the animal handling and necropsy protocols were approved by the ULAR regulatory unit of Michigan State University. Cobalt exposure, tissue harvesting and processing: Control and HlF1aA/A male mice were randomly assigned to one of 6 groups. Mice were treated with saline, or 10 mM cobaltous chloride in 25 pL volume by oropharyngeal aspiration daily for 1, 2 or 5 days. The 10 mM cobalt chloride concentrations correspond to daily exposure of 60 pg of CoClz. Animals were sacrificed 24 hours following the final exposure. In the case of the one day 212 treatment group, a single dose of cobalt was administered and mice were sacrificed 24 hours later. For the two day time point, two doses were delivered at 24 hours interval and animals were sacrificed 24 hours after second dose. Finally, for five days time point, five doses were delivered at 24 hours intervals and animals were sacrificed 24 hours after fifth dose. Following exposure, mice were anesthetized with sodium pentobarbital (50 mg/ml), and a midline laparotomy was performed. The trachea was exposed and cannulated. The lung and heart were removed en bloc and the lungs were lavaged with two successive one ml volumes of sterile saline. These fractions were combined and total cell counts were performed using a hemocytometer. Differential cell counts were performed in cytospin samples using Diff-Quik reagent (Baxter, FL). The remaining BALF was frozen for cytokine profiling. The right lung lobe was removed and stored in RNAIater RNA stabilizing reagent (Qiagen, Valencia, CA) for protein and RNA isolation. The left lobe was fixed in 10% neutral buffered formalin for histopathological analysis. Protein Assay: The total amount of protein in the BALF was quantified using the Bradford assay [18]. Briefly, BALF sample were diluted in distilled water and mixed with dye reagent via manufacturer’s instructions (Bio- Rad,Hercules, CA). Absorbance was read at 595nm using spectrophotometer (GeneQuant 100, GE Heatlthcare Piscataway, NJ). Protein concentrations were determined by comparison to a standard curve created from serially diluted bovine serum albumin standards of known concentrations. 213 Histopathology and immunohistochemistry: At least four to six mice from each genotype and treatment group were analyzed for histopathological changes. Formalin fixed left lung lobe tissues were paraffin embedded and 5- micron thick sections were mounted on glass slides and stained with hematoxylin and eosin (H&E) or major basic protein (1:500 dilution, Mayo Clinic, AZ), 40kDa antigen of neutrophils (MCA771GA 1:100 dilution, Serotec, Raleigh, NC) as previously described [17]. Determination of cytokine levels by Bead array: Levels of cytokines from acellular BALF samples were measured using BD CBA Mouse Soluble Protein Flex Sets and FACSCaIibur flow cytometer according to the manufacturer’s instruction (CBA; BD Biosciences, San Diego, CA). Cytokines measured were lL-2, KC, lL-4, lL-5, lL-13, TNFor, lL-6, lL-10, INF-y and Rantes. Briefly, BALF was mixed with capture beads and incubated for 1 hour at room temperature. Subsequently, PE detection reagent was added and incubation for 1 hours at room temperature. Following extensive washing, samples were analyzed on a BD FACSArray bioanalyzer (BD Biosciences) according to the manufacturer’s instruction. Quantitative analysis: All cell counts and cytokine and gene expression data were analyzed by ANOVA followed by a Bonferroni posttest. All the data from the study was presented as standard error of the mean (SEM). Statistical difference of P value less than 0.05 was considered as significant. 214 Figure 5.2. BALF proteins and total cell counts from control and HIF1a A/A mice. Total protein concentrations (A) and cell counts (B) were assessed from BALF of saline (white bars),and cobalt (hatched bars) treated control mice and saline (checkered bars) and cobalt (black bars) treated HlF1orA/A mice as described in materials and methods. N>5 mice/group. Data are expressed as meantSE. * = P5105. E] Ctrl-Saline HIF1orA’A - Saline % Ctrl - 60 pg CoClz l HIF1aA’A - 60 pg CoClz 215 TI .00-0v... 5 days .0080. 3....- r //////////////////////// 2 days .00-0V0. .00-0v... y /d 7o- 60- o- o- o- o. o- o- A_18:9¢ £305 :33. M .0. .n. u. .10. x. _E .8 2.8 RESULTS: BALF protein exudation and cellularity: To characterize the role of HlF1a in cobalt-induced lung injury, control and HlF1aA/A mice were randomly assigned to saline-or cobalt-treated groups. Individual groups received sterile saline or 60 pg cobalt chloride by oropharyngeal aspiration daily for 1, 2, or 5 days (Fig. 5.1). Mice were euthanized 24 hours after last exposure, BALF was collected and lung tissue was processed for light microscopic examination, IHC, and RNA isolation. Total BALF protein concentration was measured as an index of lung epithelial permeability and hence, lung injury. Cobalt-treated control and HlF1aA/A mice showed significantly higher protein concentration indicating that lung injury initiates as soon as 24 hours after the initial exposure (Fig. 5.2A). The level of protein exudation in cobalt-treated control mice remained within narrow range of 35-45 ug/100ul at all the three time points. In contrast, cobalt-treated HlF1aA/A mice showed significantly higher protein exudation (60.71162) at the 48 hours time point as compared to respective control (38.9153) counterparts (Fig. 5.2A). suggesting these mice are more prone to metal-induced lung injury. The total cell count from BALF was also measured to follow the progression of injury. Total cells in BALF showed a significant increase in both control and HIF1aA/A mice following 5 days of cobalt exposure. There was no difference between control and HlF1aA/A mice at any time point. These results suggest that acute cobalt-induced changes in total cell infiltration in the lung are not affected by loss of HIF10L. 217 Figure 5.3. Effect of cobalt treatment on inflammatory cells recovered in bronchoalveolar lavage fluid. Differential cell counts were performed for macrophages (A), lymphocytes (B), neutrophils (C), and eosinophils (D) from BALF of saline (white bars), and cobalt (hatched bars) treated control mice and saline (checkered bars) and cobalt (black bars) treated HlF1aA/A mice. N>5 mice/group. Data are expressed as meaniSE. * = P4105. Ctrl-Saline HlF1ocA’A - Saline % Ctrl - 60 pg CoClz I HIFmNA - 60 pg CoClz 218 V///////////// m — ////////// s = a...“ w S e _ So. ov e 9 _ . ._ .T. 8 So ov .. PVVJ In _3.ova. W p a O O dea d h r Save 2 p C m a mcdva. LY M m. . o e a. e o o 0. o A p25 .5 n“=3. 2.8 B Ame to .E .8. 2.8 2.5- e. a. m. 2 0.5- 219 Figure 5.3. Continued. C. Neutrophils 2.0- III— E . IE 1.01 l 3 D. Eosinophils 1.5- ———| '9 Cells per ml (x105) .° ‘1' 1 day 2 days 5 days 220 To characterize the types of inflammatory cell infiltration that made up the total cells in BALF, differential cell counts was performed. Similar to total cell counts, total macrophages and lymphocytes showed time dependent increase in numbers, however, no significance difference was noticed between cobalt-treated control and HlF1aA/A mice at any of the three time points (Fig. 5.3A and 5.38). In contrast, there was a distinct difference between the two genotypes in the numbers of neutrophils and eosinophils found in the BALF. The control mice displayed a significant increase in neutrophils following cobalt exposure at the 48 hour time point that was not seen in the HlF1a deficient mice. Moreover, this increase was resolved by the 5 day treatment time (Fig. 5.30). The HlF1aA/A mice displayed a large and sustained increase in eosinophil infiltration into the lung following cobalt exposure. This increase was observed as early as 2 days post-treatment and reached significance by 5 days of treatment (Fig. 5.3D). These genotype-specific inflammatory cells infiltrations suggest that epithelium-derived HIF10l is an important regulator in the inflammatory responses to metal insults. Histopathology of Cobalt-induced injury: Histologically, no pulmonary lesions were found in control (Fig 5.4A) or HlF1oA/A (Fig 5.4E) mice that were 221 Figure 5.4. Histopathology staining of control and cobalt-treated control mice. H & E stained lung sections from control (A-D) and HlF1aA/A (E-H) mice that were either saline-treated (A and E) or treated with one (B and E), two (C and G) or 5 consecuitive (D and H) doses of cobalt (60ug). bronchiolar airway (BA), terminal bronchioles (tb), alveolar ducts and adjacent alveoli (a). "Images in this dissertation are presented in color." 222 .. ,......... 3.. EV .. ., fl..H....ll L.‘ . . szVfi.” raw ... u . . am”, {.9 2... \uk ....nw. ... . .. . ., .,.,. . . s \ 1.2.! . ,9... I : ..I .) 223 instilled with saline alone. Cobalt-treatment induced time-dependent increase in lung damage that appeared slightly more pronounced in the HlF1aA/A mice (Fig. 548-0 and 5.4F-G). To verify the cell infiltration, lung tissues from control and HIF10lA/A mice were analyzed via immunohistochemistry using an antibody specific to major basic protein, an eosinophilic specific marker. No eosinophils were observed in saline treated mice of either genotype (data not shown). Control mice, following a single dose of cobalt showed a modest increase in MBP positive cells in the interstitium of proximal lung (Fig. 5.5A). This staining was absent in the 2 and 5 days lung section of control mice (Figs. 5.50 and E). In contrast, HlF1a deficient mice showed a pronounced and prolonged eosinophilia following cobalt exposure (Figs. 5.58, D, and F). The level of neutrophilic infiltration was also assessed using immunohistochemistry. Again, there was little PMN positive staining observed in either control or HIF1aA/A saline treated mice (data not shown). In control mice, there was substantial PMN positive staining as early as 24 hours after the first cobalt exposure (Fig. 5.6A), This staining remained following 2 days of cobalt exposure, however, it was slightly diminished. Following 5 days of exposure, control mice still displayed some neutrophil positive staining but it restricted to peribronchial regions. HlF1aA/A mice also showed strong PMN positive staining following a single dose of cobalt (Fig. 5.63). In contrast to the control mice, this PMN infiltration was absent in the 2 and 5 day treated HlF1a deficient mice. These results are in agreement with the differential cell counts 224 Figure 5.5. Major Basic Protein (MBP) immunohistochemistry in lungs from control and HlFlaA/A mice. Lung sections from control (A, C and E) and HlF1aA/A mice (B, D and F) following exposure to cobalt (60 pg) for 24 hrs (A and B), 48 hrs (C and D), or 120 hrs (E and F) were immunohistochemically stained for MBP, an eosinophil-specific marker, to identify infiltrating eosinophils (red chromagen) and counterstained with hematoxylin. "images in this dissertation are presented in color.” 225 § . u r r ...u \ Wu“? ‘ ... .&‘ .P- . ., . ... uh“ . I PK ...»... («v/:1. l . r ..n, . . v \. . A is. if ‘94 ...M¢.V...4. law I . u , K393)“ . ...a ’ . 226 Figure 5.6. Neutrophil (PMN) immunohistochemistry in lungs from control and HIF1aA/A mice. Lung sections from control (A, C and E) and HlF1aA/A mice (B, D and F) following exposure to cobalt (60 pg) for 24 hrs (A and B), 48 hrs (C and D), or 120 hrs (E and F) were immunohistochemically stained for a 40 kDa antigen, a neutrophil-specific marker, to identify infiltrating neutrophils (red chromagen) and counterstained with hematoxylin. "Images in this dissertation are presented in color." 227 v/ .v . .r A m .11: a g .... . ”MM—«mes. EU»... . ... I «.ht .. . , ......47. .svuvww. ,fiu . u 1.4 ...Htv . a ...HVI. I‘ll ... WV ... ,. V)...“ , .. ...... . . .n _...v...}....-...s.~¢... 228 and suggest that a compromised HlF1a causes a shift in the inflammatory response of the lungs following cobalt exposure. The repeated instillation of cobalt in control (Fig 5.40) as well as HlF1aA/A (Fig 5.4H) mice for five consecutive days resulted in marked bronchopneumonia characterized by a mononuclear cell infiltrate (heteromorphic lymphocytes, monocytes, and occasional plasma cells). However, these lesions were infiltrated with numerous eosinophils specifically in HlF1aA/A mice (Fig. 5.6F) and displayed PMN positive staining specifically in the control mice. Cytokine profiling: The pathology of the lung and the BALF cellularity patterns suggested a change in the inflammatory response upon loss of HlF1a from Type II and Clara cells. To determine the mechanism underlying these patterns, 10 cytokines (viz. lL-2, KC, lL-4, lL-5, lL-13, TNFa, lL-6, lL-10, INF-y, Rantes) were profiled in the cell-free BALF. Out of these profiled cytokines four showed significant difference when compared across genotype or within treatment groups (Fig. 5.7). lL-5, a key mediator in eosinophil activation, was significantly elevated in BALF collected from mice following 1 and 2 day cobalt exposure to HlF1aA/A mice as compared to control mice (Fig. 5.7A). On the other hand, cobalt treated control mice had significantly higher levels of lL-6 and TNF—a at 48 hours time point suggesting their proinfiammatory involvement in cobalt induced acute inflammation and further transition to chronic inflammation (Fig 5.73 and 5.70). . 229 Figure 5.7. Cytokine levels in BALF from cobalt-treated control and HIF to: NA mice. Control and HlF1aA/A mice were exposed to saline or 60 pg CoClz. The levels of cytokines in BALF were assessed using BD CBA Mouse Soluble Protein Flex Sets and FACSCaIibur flow cytometer. N>5 mice/group. Outliers were removed by Grubb’s test and ANOVA was performed with Boneferroni posttest. Asterisks indicate significant difference between cobalt-treated control mice and cobalt—treated HlF10l deficient mice. * =P < 0.05, ** =P < 0.01, *** =P < 0.001 El Ctrl-Saline HIF1CXA’A - Saline % Ctrl - 60 pg CoClz I HlF1alA’A - 60 pg CoCIz 230 IL5 *Wk*' 100' 75I 48 Hours 5 Days 24 Hours 6 H. 5Days 48Ho 24 125- 100 75- 50 25- 0 231 Figure 5.7. Continued. TNF-oc 5 Days 232 Figure 5.7. Continued. RANTES lNF-‘y _. Hm, H ... . a. m I 24 Hours mmwfiaemrhm F ..Eg 233 DISCUSSION: Hard metal lung disease (HMLD) and cobalt asthma are occupational respiratory diseases affecting workers involved in manufacture and maintenance of hard metals (material consisting of tungsten carbide cemented in a matrix of cobalt), diamond polishing, and coal mining. The incidence of HMLD in diamond polishers who were exposed to cobalt-containing dust suggests cobalt as a sole etiological agent in HMLD [1]. These workers manifested ainlvay constriction, alveolitis, fibrosis and associated giant cell interstitial pneumonitis [1]. Despite the countless incidences of cobalt induced toxicities, a complete mechanistic understanding is still lacking. Several in vitro studies have shown that cobalt acts as a stabilizer of HlFs. Hypoxia-like cellular responses have also been observed following cobalt exposure [19, 20]. In addition, a defined role for HlFs in inflammation and tissue injury has been established. HlF1or target genes such as VEGF and MMPs have been shown to be unregulated in inflammatory conditions [21]. Similarly, hypoxia induced activation of genes involved in inflammatory processes suggests links between hypoxia and inflammation [22]. Based on the links between cobalt and hypoxia as well as hypoxia and inflammation, cobalt-induced HlF1or stabilization might be an important player in inflammatory manifestations of metal-induced lung disease. 234 To characterize the progression of cobalt induced inflammation, we performed short-term cobalt exposure studies in a lung epithelium—specific HlF10l deficient mice model. Subchronic cobalt exposure in the same model has elucidated the role of HlF1or in modulation of lung inflammation. These results suggest that airway epithelial-derived HlF1or plays a critical role in modulating the inflammatory response of the lung, however, the signals necessary to instigate the differences in inflammatory response could not be discerned from the single time point tested. The results presented in this study suggest that loss of epithelial derived HlF1or signaling is necessary for establishing the proper Th1 response to cobalt challenge. In recent years, Clinical and experimental studies have generated evidences highlighting various pathological changes in the lungs of exposed humans as well as experimental animals. The pathology observed in cobalt- induced inhalation toxicity includes degeneration of the olfactory epithelium, hyperplasia and squamous metaplasia in the epithelium of the respiratory turbinates and larynx, hemorrhage, macrophage infiltration in the alveolar spaces, lung edema [23] and fibrosing alveolitis [24]. In clinical case of cobalt- fume exposure, positive correlation between TNFor and cobalt pneumoconiosis has been reported that suggests TNFor’s has a potential role in the pathogenesis of interstitial lung disease [25]. However, the similar elevation of TNFor was not seen in rat alveolar macrophage culture as well as in in vivo intranasal exposure studies in rats [26]. Interestingly, levels of TNFor 235 are specifically higher in control mice after 1 and 2 days of cobalt chloride treatments that was not observed in their HIF1CXNA counterparts (Fig. 5.7). These results suggest that HlFlor is capable of regulating the cytokines correlated with manifestations of lung disease. Early in the course of injury, similar to the classical responses to cobalt exposure, neutrophils are seen in cobalt-treated HlF1or deficient as well as cobalt-treated control mice 24 hours after first exposure (Figs. 5.6A and 5.6B). This finding suggests that the initial acute inflammatory events in response to the cobalt exposure are similar between control and HlF1a deficient mice. Cobalt-treated HIF10r deficient mice exhibited less neutrophilic infiltration upon repeated exposure (Figs. 5.60 and 5.60). The possibility of early removal of neutrophils from cobalt-treated HlF1or deficient mice explains the significant reduction of BALF neutrophils 24 hours after second exposure (Fig. 5.30). Neutrophilic influx in to the lung and alveolar space is the characteristic feature of the acute lung injury (ALI) where they act as a key player in the pathogenesis of the injury. Mice deficient in CXCR2, a neutrophil membrane receptor for CXC chemokines KC/CXCL1 and MlP-2/CXCL2/3, showed marked reduction in neutrophilic reduction in response to ventilator-induced lung injury [27]. Neutrophil recruitment is mediated by KC in conjunction with several other factors such as CXC chemokines (Fig 5.7D). Although important for the immune response, neutrophil-predominant inflammatory responses are involved in diffused alveolar tissue damage through the release of proteases 236 (MMPs, protease 3, neutrophil elastase, cathepsin G) and reactive oxygen metabolites (hydrogen peroxide, hypohalous acids, hydroxyl radicals) [28]. Neutrophils generate chemotactic signals that regulate the recruitment of monocytes and dictate macrophage differentiation towards pro- or anti- inflammatory state [29]. Similar to activated alveolar macrophages, alveolar epithelial cells also produced cytokines and chemokines. It has been shown, similar to macrophages, LPS exposed type II cells also act as an important source of cytokines such as MCP-1 and lL-8 [30]. Given the growing evidence of the inflammatory roles of HlFs. It is possible that the loss of HlF1or from the type II cells result in their altered response to the signals received from inflammatory cells such as neutrophils and activated macrophages. All three time points tested in the present study have shown significant interstitial eosinophil infiltration in cobalt-treated HlF1or deficient mice as compared to cobalt-treated control mice (Figs. 5.53, 5.50 and 5.5F). However, the BALF eosinophilia cobalt-treated HlF10l deficient mice reached significance only after 5 exposures (Fig. 5.30). Interestingly, interstitial eosinophilic infiltration was present in cobalt-treated control mice that were exposed to one dose but the degree of infiltration was remarkably less as compared to cobalt-treated HlF1or deficient mice (Fig. 5.5A). Eosinophils are differentiated from myeloid precursor cells under the influence of interleukin IL- 3, granulocyte-macrophage colony-stimulating factor (GM-CSF), and IL5 These cells are selectively recruited into the always through Th2 cytokines. 237 HlF1or deficient mice specific recruitment of eosinophils suggests the epithelial origin of the inflammatory switch. To date, the role of HlFs in eosinophilic inflammation is unclear. Moreover, the role of epithelial-derived mediators as modulator of inflammatory responses has not been examined directly. Taken together, the findings from the current study supplements the data collected in the recent study and suggests critical role of epithelial-derived HlF1a in the observed inflammatory switch. However, further investigation on the chemical signals released by the HlF1or deficient type II and Clara cells will further help us to understand the mechanism underlying the recruitment of eosinophils and other downstream events. The BALF cytokine analyses for lL-2, KC, lL-4, lL-5, lL-13, TNFor, lL-6, IL- 10, INF-y and Rantes have shown the genotype specific changes after cobalt exposure. Control mice showed significant induction of TNFor, a Th1 cytokine secreted by activated macrophages, as well as INF-y (Figs. 5.70 and 5.7F). In accordance to the finding that TNF-or and INF-y stimulates Rantes production, the levels of Rantes in cobalt-treated control mice followed TNFor like pattern (Fig. 5.7E) [31]. lL-6 cytokine that promotes Th2 cells differentiation is elevated in the cobalt-treated control mice (Fig. 5.78). In contrast to control mice, cobalt-treated HlFlor deficient mice had significantly increased lL-5 that mediates Th2 cytokine-producing capacity of eosinophils (Fig. 5.7A). These changes in the cytokine levels further suggest that the loss of HlF1or in lung 238 epithelial cells of the HIF10L deficient mice results in an alteration in the cell’s ability to respond to the incoming signals from the cobalt damaged cells. Taken together, the data collected from the current study consolidates our understanding of the course and nature of cobalt induced inflammation. In conclusion, the data supports a model in which epithelial-derived HIF1or activity regulates the lung’s response to metal challenge by controlling the expression of cytokines necessary to recruit the proper inflammatory response. Loss of this regulatory mechanism leads to the tissue being biased towards a Th2-mediated inflammation. The mechanism that leads to this alteration in the inflammatory response remains unknown and is the focus of new investigations. Acknowledgements: The authors appreciate the gift of the conditional HlF1a mice from Dr. Randall Johnson (University of California San Diego) and the SP-C-rtTA/(tetO)7-CMV-Cre transgenic from Dr. Jeffrey A. Whitsett (Cincinnati Children’s Hospital Medical Center). Grants: This work was supported by funds from the National Institutes of Health (NIH) R01-ES12186 and P42 ESO4911-17 239 REFERENCES 1 Lison, D., Lauwerys, R., Demedts, M. and Nemery, B. (1996) Experimental research into the pathogenesis of cobalt/hard metal lung disease. Eur Respir J. 9, 1024-1028 Vengellur, A., Phillips, J. M., Hogenesch, J. B. and LaPres, J. J. (2005) Gene expression profiling of hypoxia signaling in human hepatocellular carcinoma cells. Physiol. Genomics. 22, 308-318 Jain, M. and Sznajder, J. I. (2005) Effects of hypoxia on the alveolar epithelium. Proceedings of the American Thoracic Society. 2, 202-205 Bunn, H. F. and Poyton, R. O. (1996) Oxygen sensing and molecular adaptation to hypoxia. Phys. Rev. 76, 839-885 Epstein, A. C., et al. (2001) C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell. 107, 43-54. Semenza, G. L., Roth, P. H., Fang, H. M. and Wang, G. L. (1994) Transcriptional regulation of genes encoding glycolytic enzymes by hypoxia-inducible factor 1. J. Biol. Chem. 269, 23757-23763 Forsythe, J. A., et al. (1996) Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol. Cell. Biol. 16, 4604-4613 Jung, Y., et al. (2003) Hypoxia-inducible factor induction by tumour necrosis factor in normoxic cells requires receptor-interacting protein- dependent nuclear factor kappa B activation. Biochem J. 370, 1011- 1017. Mojsilovic-Petrovic, J., et al. (2007) Hypoxia-inducible factor-1 (HIF-1) is involved in the regulation of hypoxia-stimulated expression of monocyte chemoattractant protein-1 (MCP-1/CCL2) and MCP-5 (Ccl12) in astrocytes. J Neuroinflamm. 4, 12 240 10 11 12 13 14 15 16 17 18 19 Salnikow, K., et al. (2004) Depletion of intracellular ascorbate by the carcinogenic metals nickel and cobalt results in the induction of hypoxic stress. J Biol Chem Vengellur, A., Phillips, J. M., Hogenesch, J. B. and LaPres, J. J. (2005) Gene expression profiling of hypoxia signaling in human hepatocellular carcinoma cells. Physiological genomics. 22, 308-318 Lombaert, N., Lison, D., Van Hummelen, P. and Kirsch-Volders, M. (2008) In vitro expression of hard metal dust (WC-Co)-responsive genes in human peripheral blood mononucleated cells. Toxicol Appl Pharrnacol. 227, 299-312 Ryan, H. E., Lo, J. and Johnson, R. S. (1998) HIF-1 alpha is required for solid tumor formation and embryonic vascularization. EMBO Journal. 17, 3005-301 5 Ryan, H. E., et al. (2000) Hypoxia-inducible factor-1 alpha is a positive factor in solid tumor growth. Cancer Res. 60, 4010-4015 Perl, A. K., et al. (2002) Early restriction of peripheral and proximal cell lineages during formation of the lung. Proc Natl Acad Sci U S A. 99, 10482-10487. Lobe, C. G., et al. (1999) Z/AP, a double reporter for ore-mediated recombination. Dev Biol. 208, 281-292. Saini, Y., Harkema, J. R. and LaPres, J. J. (2008) HlF1{alpha} Is Essential for Normal Intrauterine Differentiation of Alveolar Epithelium and Surfactant Production in the Newborn Lung of Mice. J. Biol. Chem. 283, 33650-33657 Bradford, M. M. (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Analytical biochemistry. 72, 248-254 Vengellur, A. and LaPres, J. J. (2004) The role of hypoxia inducible factor 1alpha in cobalt chloride induced cell death in mouse embryonic fibroblasts. Toxicol Sci. 82, 638-646 241 20 21 22 23 24 25 26 27 28 29 Karovic, O., et al. (2007) Toxic effects of cobalt in primary cultures of mouse astrocytes. Similarities with hypoxia and role of HIF-1alpha. Biochemical pharmacology. 73, 694-708 Elson, D. A., et al. (2000) Coordinate up-regulation of hypoxia inducible factor (HlF)-1 alpha and HIF-1 target genes during multi-stage epidermal carcinogenesis and wound healing. Cancer research. 60, 6189-6195 Walmsley, S. R., Cadwallader, K. A. and Chilvers, E. R. (2005) The role of HIF-1alpha in myeloid cell inflammation. Trends in immunology. 26, 434-439 Bucher, J. (1991) NTP technical report on the toxicity studies of Cobalt Sulfate Heptahydrate in F344/N Rats and B6C3F1 Mice (Inhalation Studies) (CAS No. 10026-24-1 ). Toxicity report series. 5, 1-38 Lison, D. (1996) Human toxicity of cobalt-containing dust and experimental studies on the mechanism of interstitial lung disease (hard metal disease). Critical Reviews in Toxicology. 26, 585-616 Rolfe, M. W., Paine, R., Davenport, R. B. and Strieter, R. M. (1992) Hard metal pneumoconiosis and the association of tumor necrosis factor-alpha. The American review of respiratory disease. 146, 1600- 1602 Huaux, F., Lasfargues, G., Lauwerys, R. and Lison, D. (1995) Lung toxicity of hard metal particles and production of interleukin-1, tumor necrosis factor-alpha, fibronectin, and cystatin-c by lung phagocytes. Toxicology and applied pharmacology. 132, 53-62 Belperio, J. A., et al. (2002) Critical role for CXCR2 and CXCR2 ligands during the pathogenesis of ventilator-induced lung injury. The Journal of clinical investigation. 110, 1703-1716 Nathan, C. (2006) Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol. 6, 173-182 Bennouna, S., Bliss, S. K., Curiel, T. J. and Denkers, E. Y. (2003) Cross-talk in the innate immune system: neutrophils instruct recruitment 242 30 31 and activation of dendritic cells during microbial infection. J Immunol. 171, 6052-6058 Thorley, A. J., et al. (2007) Differential regulation of cytokine release and leukocyte migration by Iipopolysaccharide-stimulated primary human lung alveolar type II epithelial cells and macrophages. J Immunol. 178, 463-473 Teran, L. M., et al. (1999) Th1- and Th2-type cytokines regulate the expression and production of eotaxin and RANTES by human lung fibroblasts. American journal of respiratory cell and molecular biology. 20, 777-786 243 CHAPTER 6 CONCLUSIONS The increasing use of disposable electronics and growing industrialization of nation’s economies have drastically raised the risk of exposure to toxic metals. Mining operations, the burning of fossil fuels and globalization have pushed metals into close proximities of neighborhoods and the general population. The most “at risk” group, however, remains workers in various industrial settings, including large-scale smelting operations and alloy manufacturers. One of these metals, cobalt, is used as a coloring agent for ceramics and the production of alloys. ‘Cobalt is used widely in several industries involved in the production of coloring agent for ceramics, hard metal alloys, sintered carbides, drilling and grinding tools. The people who manufacture these metals and use the end products are at particular risk for exposure. It has been estimated that more than 1 million workers in the United States are exposed to cobalt due to the nature of their work. Moreover, cobalt is present in 426 of the 1,636 National Priorities List (NPL) hazardous waste sites identified by the Environmental Protection Agency (EPA). Finally, the correlation of high environmental cobalt levels to a cluster of lung diseases makes the understanding of cobalt-induced signaling and toxicity important to workers and the public at large. 244 Workers exposed to cobalt dust from welding, grinding, or diamond polishing operations have displayed multiple lung pathologies. Cobalt (or hard metal) asthma is one of three occupational respiratory diseases associated with exposure to the transition metal. The other two are hypersensitivity pneumonitis and interstitial lung disease with fibrosis. These diseases are caused by the inhalation of hard metal particles and are characterized by ainlvay constriction, alveolitis, fibrosis and associated giant cell interstitial pneumonitis. Therefore, mechanistic understanding of the toxicities of cobalt and related metals such as nickel is important to devise treatment strategies. The characterizations of these toxicities demands comprehensive understanding of the effects of toxicants on the cell signaling and the downstream changes in the gene expression. It is well established that cobalt is a hypoxia mimic due to its ability to induce hypoxia-like gene expression responses. To enhance our understanding on the involvement of hypoxia- mimicking properties of cobalt in its toxic responses, we developed and used a mouse model system. This mouse model is an inducible lung-specific HlF1or deficient system based on Cre-LoxP recombination strategy. HIFs are the most widely studied family of hypoxia responsive transcription factors and they directly influence the expression of more than 150 genes. Among three known HlFor’s, oxygen labile isoforms, HIF1or is most widely expressed isoform that has been studied extensively. Previous studies in our lab, using an immortalized mouse embryonic fibroblast (MEFs) 245 cell line lacking HlF1a, established the correlation between HIF1a removal and protection against CoCI2 toxicity. Validation of our in vitro understanding of cobalt-induced HlFs signaling necessitates the in vivo studies of cobalt toxicity in HIFs null animals. Our attempt to generate the inducible lung-specific HlF1adeficient mice model led us to an interesting investigation on the roles of HlF1ain lung development. Lung specific embryonic deletion of HlF1a earlier than 3 days prior to parturition led to HlF1q deficient neonates exhibiting cyanosis and respiratory failure soon after birth. The expression of surfactant proteins was significantly lower in HlF1a deficient neonates as compared to Iitterrnate control pups. Examination of the lungs from HlF1q deficient neonates via light and transmission electron microscopy confirmed defects in both alveolar epithelial differentiation and septal development that resulted in the observed respiratory distress. It was interesting to note the decreased expression of HlF20r, another prominent form of HIFor in lungs, in HlF1a deficient neonates that led us to further investigate the roles of both HIF isoforms, individually and collectively. The attempts to generate viable HlF20r mice revealed that the removal of this isoform from the lung does not affect the viability of neonates. However, the removal of both the isoforms from the lungs revealed an interesting rescue phenotype. Microarray analysis of the lungs from HlF1aA/A, HIF2 orA/A and HIF1/2 orA/A mice identified genes and cellular pathways. The majority of these pathways, such as surfactant metabolism and vesicular trafficking, were 246 specifically affected in the HlF1aA/A neonates. However, the expression profile of some genes, such as IL1R—ll and ODS-1, were significantly reduced in the HlF1or deficient mice and regained their expression levels after the simultaneous removal of HlF10r and HIF2 or. Further studies to understand the intricate network of developmentally important factors relative to HlFs necessitates conducting future experiments using rescue or complementation strategies. Given the extremely complex developmental network involved in the lung development, it would be rational to approach to investigate the observed findings through multidimensional approach specific to each of the affected cellular pathway. The upregulation of genes involved in cell cycle processes suggests the shift from cell differentiation pathways to more cell proliferation pathways. This is in agreement with the pathology in which the HIF1or deficient mice exhibited signs of stalled differentiation of immature cuboidal alveolar epithelial cells. It is assumed that these immature cuboidal alveolar epithelial cells are the precursors of differentiated alveolar type II cells that, in turn, differentiate into type | cells. The most striking function of type II cells in the developing lungs is surfactant biosynthesis which necessitates the cell- specific upregulation of genes involved in fatty acid synthesis, cholesterol biosynthesis, surfactant proteins biosynthesis and the specialized vesicular transport system. Surfactant biosynthesis and their vesicular transportation to the cell membrane are characteristic of alveolar type II cells. It is clearly a cell- specific temporal mode of expression of genes involved in these processes. 247 In addition to the cell cycle processes, the genes involved in the vesicular transport and surfactant pathways are downregulated in HIF1or deficient mice. There could be two possible explanation of these observed expression pattern. First, the stalled differentiation might be the consequence of the deletion of HlF1or that leads to the perturbation of the downstream developmentally critical cascades. This effect could be explained by the observed gene expression pattern of transcriptional factors such as B-catenin that were significantly downregulated upon HIF1or deletion. It is quite possible that some other developmentally critical (transcriptional) factors fall directly or indirectly downstream of HlF1or in the cellular differentiation cascades. Thus, the altered expression of the surfactants might be the consequence of stalled differentiation process. The seCond possible explanation of the respiratory distress phenotype observed in HlF1or deficient mice might be more intimately related to the surfactant metabolism. The genes involved in the surfactant metabolism are still not well studied for their hypoxia responsiveness. Further studies on the selected genes will certainly provide avenues for the development of treatment strategies to combat neonatal distress syndrome. The lethality following in utero deletion of HlF1a, led us to adopt postnatal strategy of doxycycline treatment and functional deletion of HlF1a. The post-natal deletion of HIF1or from Type II and Clara cells had no observable pathology. In order to elucidate the role of epithelial derived- HlF1or signaling in cobalt-induced lung injury these mice were exposed to cobalt chloride via oropharyngeal aspiration. Compared to control mice, mice 248 that were HlF1adeficient in their lungs exhibited airway infiltration of eosinophils and airway epithelial changes, including mucus cell metaplasia and increased levels of the chitinase-like proteins YM1 and YM2. Mice deficient in HlF1or also showed a drastic change in cytokine profiles in their lavage fluid when compared to their control. These results suggested that loss of HlF1or from alveolar Type II epithelial and Clara cells of the lungs leads to cellular and molecular processes that are associated with asthma following cobalt exposure. Moreover, the results suggest that ainlvay epithelial-derived HlF1or plays a critical role in modulating the inflammatory response of the lung. These results also suggested a difference in the inflammatory response between the control and HIF10lA/A mice; however, it is based on collected data at a single time point following two weeks of cobalt exposure. Thus, to consolidate our understanding on the course and nature of cobalt induced inflammation further time course studies were planned. In the time course studies, control and HIF1or deficient mice were exposed to cobalt daily for 1, 2, or 5 days. Bronchoalveolar lavage fluid (BALF) cellularity from HlF1adeficient mice displayed a progressive eosinophilic infiltration whereas control mice displayed a transient Increase in neutrophils. Histological analysis revealed accelerated tissue injury following acute cobalt challenge in HlF1or deficient mice. Finally, BALF cytokine analysis showed lL-5 elevation specific to cobalt-treated HIF1or deficient mice. In contrast, control mice showed specific induction of lL-6 and tumor necrosis 249 factor or (TNFa) following cobalt treatment. These results suggests that epithelial-derived HlF1or is essential for regulating early inflammatory events following cobalt challenge and loss of this regulation biases the lung towards a Th2-mediated process and an asthma-like pathology following metal exposure. Taken together, the striking differences observed following cobalt exposure in the two mice suggests that they will be a powerful tool to help understand the relationship between allergy-induced asthma, hypoxia, and inflammation. More importantly, direct comparison of the responses of the control and HIF1orA/A mice in other asthma models (e.g. ovalbumin challenge) and to other inflammatory inhalants will correlate this relationship to specific cytokines. The research also raises several important questions: Does the loss of HIF1or alter an organism’s susceptibility to asthma using other allergens? What role does HlF1or derived from other cell types (e.g. infiltrating inflammatory cells and Type I cells) play in modulating this inflammatory response. Does post-natal deletion of HlF1or alter HlF20r and if so, what impact does this have on the inflammatory response. Most importantly, does a decrease in HlF1or functionality lead to an increased susceptibility to asthma in humans? More specifically, is it possible that loss or decrease in HlF1or function biases the lung towards a Th2 immune polarization and this increases the susceptibility of individuals towards extrinsic asthma? 250 Table A.1. List of differentially expressed genes HIF1 HIF2 HIF1/2 GeneID Gene Name ratio ratio ratio Solute carn'er family 34 (sodium phosphate), 20505 member 1 37.07 -1.00 -l.19 Similar to Ankyrin repeat domain- 546994 containing gotein 26 28.05 -1.00 -1.02 15395 Homeo box A10 21.62 2.30 1.24 56183 Neuromedin U 16.00 1.27 5.62 212070 Clarin3 13.84 -1.19 -1.09 15378 Hepatic nuclear factor 4, alpha 13.40 1.08 -1.82 84112 Succinate receptor 1 11.10 1.40 1.06 228491 Zinc finger protein 770 10.52 -1.16 1.10 14866 Glutathione S-transferase, mu 5 8.64 1.48 5.84 17748 Metallothionein 1 8.42 4.34 3.44 241516 Fibrous sheath-interactigg protein 2 6.46 -1.13 1.47 13653 Early gowth remnse 1 6.25 -1.30 2.35 215627 Zinc finger and BTB domain containing 8 5.54 1.33 2.20 654824 Ankyrin repeat domain 37 5.31 1.04 2.14 75656 RIKEN cdna 1700020A23 gene 5.13 1.22 2.54 230766 Cdna sequence BC030183 4.80 1.10 1.37 66261 Transmembrane 4 L six family member 20 4.23 -1.11 1.27 Methenyltetrahydrofolate synthetase domain 234814 containing 3.88 -1.52 1.24 75512 Glutathione peroxidase 6 3.85 1.12 -1.04 20423 Sonic hedgfig 3.74 1.35 1.08 Chromodomain helicase DNA binding 71389 protein 6 3.72 -1.19 1.24 72650 RIKEN cdna 2810006K23 gene 3.69 1.00 1.13 Cytochrome P450, family 2, subfamily c, 13099 polypeptide 40 3.68 1.51 -1.79 239250 SLIT and NTRK-like family, member 6 3.65 -1.03 1.39 68214 Glutathione S-transferase omeEaZ 3.64 -1.20 -1.29 58909 RIKEN cdna D43OOISBOI gene 3.62 1.08 1.34 67951 Tubulin, beta 6 3.37 1.03 1.46 19434 Retina and anterior neural fold homeobox 3.36 -1.00 -1.31 232339 Ankyn'n repeat domain 26 3.32 -1.48 1.23 20464 Single-minded homolog 1 (Drosophila) 3.31 -1.06 -l .02 53601 Protocadherin 12 3.27 1.28 1.01 12000 Arginine vasopressin receptor 2 3.25 -1.35 -l .00 13193 Doublecortin 3 .24 -1.00 1.05 251 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 207742 Ring finger protein 43 3.14 - l .09 1.03 380773 RIKEN cdna 1810035L17 gene 3.13 -1.52 1.07 Heparan sulfate (glucosamine) 3-0- 54710 sulfotransferase 381 3.10 -1.13 1.42 66950 Transmembrane protein 206 3.09 -1.17 1.07 Haloacid dehalogenase-like hydrolase 72748 domain containing 3 3.09 1.04 1.10 320803 RIKEN cdna C130022MO3 gene 3.06 -l.08 1.31 72080 RIKEN cdna 20103171324 gene 3.04 1.41 1.14 22295 Cadherin 23 (otocadherin) 3.04 1.74 -2.20 56742 Proline/serine-rich coiled-coil 1 3.02 1.57 2.66 68750 Ras responsive element binding protein 1 3.01 1.14 1.22 74043 Peroxisome biogenesis factor 26 3.00 1.20 1.55 18762 Protein kinase C, zeta 2.98 -1.02 1.47 216829 Cdna segrence BC025076 2.96 -1.21 1.34 Anterior pharynx defective lb homolog (C. 208117 Elegg‘») 2.92 1.47 1.63 15438 Homeo box D9 2.88 -1.20 -1.17 215693 Zinc tiger, matrin type 1 2.88 -1.22 1.31 13841 Eph receptor A7 2.87 1.32 1.19 78320 RIKEN cdna 2210415111£ne 2.87 1.06 1.83 71263 Maestro 2.87 1.59 1.49 Proline/serine-n'ch 56742 coiled-coil 1 2.84 1.60 2.03 18828 Phospholipid scramblase 2 2.84 1.13 1.28 RAS-like, estrogen-regulated, growth- 232441 inhibitor 2.84 -1.43 -1.14 16182 Interleukin 18 receptor 1 2.82 -l.25 1.65 18510 Paired box gene 8 2.81 -l.05 1.11 Mediator of RNA polymerase 11 329650 transcription 2.80 1.03 1.29 18828 Phospholipid scramblase 2 2.80 1.14 1.40 328425 Deleted in lymphocytic leukemia, 2 2.78 1.16 1.47 Adaptor-related protein complex 3, mu 1 55946 subunit 2.77 -1.07 1.33 98685 RIKEN cdna 1190005F20 Elle 2.77 1.02 1.12 103573 Exportin 1, CRMl homolog (yeast) 2.76 1.06 1.41 215494 Expressed sequence C85492 2.75 -1.23 1.51 100177 Zinc finger, MYM-type 6 2.74 -1.00 -2.39 218763 Leucine rich repeat containinL33 2.73 -1.00 1.09 Inhibitor of growth family, 69260 member 2 2.73 -1.07 1.38 252 HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio 171282 Acyl-coa thioesterase 4 2.70 1.14 1.07 433940 Cdna sequence BC057022 2.70 -1.52 1.20 66315 SUMOI/sentrin specific peptidase 7 2.70 1.03 1.20 12160 Bone morphogenetic protein 5 2.69 1.52 1.11 244071 ATP/GTP binding protein-like 1 2.68 -1.00 -1.08 320573 RIKEN cdna C130024JOZ gene 2.68 -1.09 1.08 77418 RIKEN cdna C030015Al9jene 2.67 -1.62 1.31 83486 RNA bindingmotif protein 5 2.67 -1.26 1.06 66274 RIKEN cdna 1810012P15 gene 2.65 1.15 1.48 330256 Hypotheticalprotein 9530028C05 2.64 1.46 1.36 56742 Proline/serine-rich coiled-coil 1 2.64 1.63 1.77 228880 Protein kinase C binding protein 1 . 2.64 1.00 1.21 Tumor necrosis factor receptor superfamily, 22163 member 4 2.64 1.12 1.48 DEAH (Asp-Glu-Ala-His) box polypeptide 216877 33 2.64 1.10 -1.22 72852 RIKEN cdna 2900024010 gene 2.64 1.07 1.55 78004 Proline rich 15 2.63 1.37 1.22 67892 RIKEN cdna 1810063B05 gene 2.63 -1.40 1.19 381853 Gastric inhibitory polypeptide receptor 2.63 1.59 1.67 21683 Tectorin alpha 2.63 -1.06 -1.30 218100 Zinc finger protein 322a 2.63 -1.16 1.20 791282 Predicted gene, ENSMUSGOOOOOOS7802 2.61 1.03 -1.01 434179 Predicted gene, EG434179 2.60 1.29 1.34 13423 Deoxyribonuclease II alpha 2.58 -1.23 1.42 71599 SUMO/sentrin specific peptidase 8 2.58 -1.10 1.43 67790 RAB39B, member RAS oncogene family 2.58 -1.18 1.33 RIKEN cdna 77609 C330001K17 gene 2.56 1.58 1.16 217716 Mutl homolog 3 (E coli) 2.56 1.16 1.03 68281 RIKEN cdna 4930430F08 gene 2.56 -1.12 1.24 56335 Methyltransferase-like 3 2.55 -1.04 1.07 16428 IL2-inducible T-cell kinase 2.55 1.02 1.28 UPF3 regulator of nonsense transcripts 68134 homolog B (yeast) 2.55 1.34 1.31 193736 Zinc finger and BTB domain containing 12 2.54 -1.11 1.22 Methylmalonic aciduria (cobalamin 109136 deficiency) type A 2.53 -1.04 1.28 Structural maintenance of 14211 chromosomes 2 2.52 1.09 1.47 67283 Solute carrier family 25 , member 19 2.52 -1.13 1.26 253 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 218850 DNA segment, Chr 14, Abbott 1 expressed 2.51 -l.10 -1.09 THAP domain containing, apoptosis 66816 associated protein 2 2.50 -1.34 1.21 72397 RNA binding motif protein 12B 2.50 1.48 -1.09 Ventricular zone expressed PH domain 72789 homolog 1 (zebrafish) 2.49 -1.05 1.63 22635 Zonadhesin 2.49 1 .09 1 .10 75387 Sirtuin 4 (S. Cerevisiae) 2.49 -1.03 1.42 66522 Pyroflltamyl-peptidase I 2.47 -1.29 -1.04 50780 Rgulator of G-g‘otein signaling 3 2.47 -1.04 1.04 30838 F-box and WD-40 domain protein 4 2.46 -1.15 -1.04 235442 RABSB, member RAS onc0jene famill 2.46 -1.04 1.20 77857 RIKEN cdna 9430065F17 gene 2.45 -1.04 1.22 SH3-domain GRB2-like (endophilin) 73094 interacting protein 1 2.45 -1.12 -1.08 Oxidized low density lipoprotein (lectin- 108078 like) receptor 1 2.43 -1.53 -1.16 625963 Hypothetical protein LOC625963 2.42 1.20 1.36 268853 Gene model 671, (NCBI) 2.41 -1.12 1.01 13176 Deleted in colorectal carcinoma 2.40 -1.17 -1.12 58240 HCLSI bindigg protein 3 2.39 1.02 1.21 58894 RIKEN cdna 4732460K03 Que 2.38 -1.58 1.68 75787 RIKEN cdna 4930471M09 gene 2.38 -1.18 1.05 21808 Transforminggrowth factor, beta 2 2.38 1.05 1.09 Bromodomain and WD repeat domain 93871 containing 1 2.38 1.28 1.24 Golgi-specific brefeldin A-resistance factor 107338 1 2.38 1.05 1.27 74455 NOLl/NOPZ/Sun domain family 6 2.37 1.11 1.14 228880 Protein kinase C binding protein 1 2.37 1.09 1.15 57432 Zinc finger CCCH type containing 8 2.37 -1.01 1.23 434234 RIKEN cdna 2610020H08 gene 2.36 -l.00 1.30 67382 Bromodomain containing3 2.35 -1.35 1.07 225283 Cdna sequence BC021395 2.35 -1.26 1.36 Neural precursor cell expressed, 17997 developmentally down-regulated gene 1 2.34 -1.12 1.16 21848 Tripartite motif-containing 24 2.33 -1.25 -1.10 94281 Sideroflexin 4 2.33 -1.08 1.49 Zinc finger with KRAB and SCAN domains 72739 3 2.32 1.09 1.20 77940 RIKEN cdna A930004D18gg1e 2.31 -1.12 -1.12 78004 Proline rich 15 2.31 -l .02 1.07 254 HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio 72103 RIKEN cdna 2010301N04 gene 2.31 -1.25 -1.03 11994 Protocadherin 15 2.30 -1.21 1.07 77519 RIKEN cdna 5730601FO6 gene 2.29 -1.08 1.23 73242 RIKEN cdna 2610110612 gene 2.28 1.10 1.27 66821 BCSl-like (yeastL 2.28 -1.00 1.26 69008 Calcium bindingprotein 39-1ike 2.27 1.00 1.29 Transmembrane and tetratricopeptide repeat 70551 containing 4 2.27 1.04 1.38 434179 Predicteigme, EG434179 2.27 1.27 1.44 240641 Kinesin family member 20B 2.26 1.05 1.12 Rho guanine nucleotide exchange factor 213649 (GEF) 19 2.26 1.37 1.28 109731 Monoamine oxidase B 2.24 1.18 1.27 Mitogen-activated protein kinase kinase 26404 kinase 12 2.24 1.04 1.24 74087 Solute carrier family 7, member 13 2.24 1.02 -1.21 26414 Mitogen-activated protein kinase 10 2.24 -1.10 -1.00 18205 Neurotrophin 3 2.23 -1.21 1.20 268822 Aarf domain containing kinase 5 2.23 -1.00 1.44 231832 Transmembrane protein 184a 2.23 1.25 1.27 338320 Melanoma inhibitory activity 2 2.23 -1.09 -1.14 76742 Sorting nexin family member 27 2.23 -1 . 14 1.09 387334 Defensin beta 50 2.22 -1.11 1.07 19331 RABl9, member RAS oncogene family 2.22 -1.01 1.25 DEAD (Asp-Glu-Ala-Asp) box polypeptide 230073 58 2.21 1.17 1.22 Enoyl Coenzyme A hydratase domain 67856 containing 3 2.20 1.26 1.25 320376 BCL6 co-repressor-like 1 2.20 -1.26 1.05 113854 Vomeronasal 1 receptor, B4 2.19 -1.03 1.15 52335 Ataxin l-like 2.19 -l.08 1.08 Rap guanine nucleotide exchange factor 268480 (GEF)-like 1 2.19 -1.03 1.40 12858 Cytochrome c oxidase, subunit Va 2.19 1.04 1.16 Thiosulfate sulfurtransferase, 221 17 mitochondrial 2.19 1 .28 1.46 Beta-1,3-glucuronyltransferase 2 280645 (glucuronosyltransferase S) 2.19 -1.06 1.31 Lymphocyte antigen 6 complex, locus 114654 G6D 2.18 -1.08 1.27 240638 Solute carrier family 16 , member 12 2.18 1.20 1.42 245886 Ankyrin repeat domain 27 (V PS9 domaign) 2.18 -1.17 1.03 255 HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio Small nuclear RNA activating complex, 77634 polypeptide 3 2.18 -1.01 -1 .18 Sfil homolog, spindle assembly associated 78887 (yeast) 2.18 1.00 1.09 21778 Testis expressed gene 9 2.17 1.47 1.10 Vezatin, adherens junctions transmembrane 215008 protein 2.17 1.08 1.25 75763 RIKEN cdna 4833418A01 gene 2.17 -l.01 1.08 Poly (ADP-ribose) polymerase family, 101187 member 11 2.16 -1.33 1.22 30054 Ring finger protein 17 2.16 -l .00 1.18 12009 5-azacytidine induced gene 1 2.16 1.02 1.07 269023 Zinc fingr protein 608 2.15 1.04 1.23 22718 Zinc finggr protein 60 2.15 -1.58 1.13 26450 Retinoblastoma binding protein 9 2.15 1.31 1.52 V-set and immunoglobulin domain 57276 containing 2 2.15 1.24 1.26 18044 Nuclear transcription factor-Y alpha 2.14 -1.54 -1.15 381293 Kinesin family member 14 2.14 1.23 1.07 320488 RIKEN cdna D130039L10 gene 2.14 -1.12 -1.07 20689 Sal-like 3 (Drosophila) 2.14 -1.00 -1.14 671 18 Bifunctional apoptosis regulator 2.14 1 .04 1.30 225283 Cdna sequence BC021395 2.14 -1.01 1.18 67459 Nuclear VCP-like 2.13 -1 . 13 1.15 13639 EphrinA4 2.13 1.11 1.16 224055 Receptor transporter protein 2 2.13 -1.20 -1.09 28019 Inhibitor of growth family, member 4 2.13 1.12 1.05 19650 Retinoblastoma-like 1 (p107) 2.13 1.16 1.28 27369 Deoxyguanosine kinase 2.13 -1.02 1.10 RAB3A interacting protein 74760 (rabin3)-like 1 2.13 1.11 1.44 78745 RIKEN cdna 9530097N15 gene 2.12 1.12 -1.03 20585 Helicase-like transcription factor 2.12 1.08 1.34 Zinc finger, RAN-binding domain 53861 containing 2 2.12 -1.06 1.45 100177 Zinc finger, MYM-type 6 2.12 1.03 1.28 SAFB-like, transcription 66660 modulator 2.12 1.03 1.29 231807 Cdna sequence BC037034 2.11 -1.20 1.01 Zinc finger with KRAB and SCAN domains 72739 3 2.11 1.05 1.13 Potassium channel tetramerisation domain 212919 containing 7 2.11 -1.02 -1.06 256 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio Splicing factor, arginine/serine-rich 1 110809 (ASF/SF2) 2.10 -1.03 1.04 237465 Coiled-coil domain containing 38 2.10 -1.31 1.02 57869 Aarf domain containing kinase 2 2.10 -1.03 1.15 77593 Ubiquitin specific petidase 45 2.10 -1.11 1.12 320575 RIKEN cdna C230057HO2 gene 2.10 -1.05 -1.00 21 1329 Nuclear receptor coactivator 7 2.09 -1.32 -1.67 230848 Zinc finger and BTB domain containing 40 2.09 -1.11 1.02 320720 FAST kinase domains 1 2.09 1.05 1.10 19364 RADSl-like 3 (S. Cerevisiae) 2.09 1.16 1.12 240028 Leucyl/cystinyl aminopgptidase 2.09 -1 .57 1 .08 1 1512 Adenylate cyclase 6 2.09 1.17 1.04 57277 Secreted Ly6/Plaur domain containing 1 2.09 1.13 1.17 Ankyrin repeat and sterile alpha motif 72615 domain containing 2.09 1.01 -1.02 54678 Zinc finger protein 108 2.09 -1.03 1.17 17763 Mature T-cell proliferation 1 2.08 1.03 1.35 67288 SFRSlZ-interacting protein 1 2.08 -1.08 1.13 17268 Meis homeobox 1 2.08 -1.02 1.12 PRP38 pre-mma processing factor 38 66921 (yeast) domain containing B 2.08 1.12 1.29 12349 Carbonic anhydrase 2 2.08 1.24 1.12 Metallophosphoesterase domain containing 77015 2 2.08 1.17 1.23 SAM pointed domain containing ets 30051 transcription factor 2.08 -1.54 1.84 242297 RIKEN cdna 1700012H17 gene 2.07 1.29 1.36 494448 Chromobox homolog 6 2.07 -1.00 -1.03 52592 Breast cancer metastasis-sumessor 1-like 2.07 -1.02 1.20 209176 Indoleamine-pyrrole 2,3 dioxygenase-like 1 2.07 -1.05 1.13 BRCA1 interacting protein C-terminal 237911 helicase 1 2.07 -1.18 1.04 16924 Ligand of numb-protein X 1 2.06 1.23 1.68 258472 Olfactory receptor 899 2.06 1.57 1.12 Cholinergic receptor, nicotinic, beta 108015 polypeptide 4 2.06 1.18 -1.05 Serine (or cysteine) peptidase inhibitor, 331535 clade A, member 7 2.05 -1.13 2.53 675815 Similar to quaking type II 2.05 -1.52 1.06 30840 F-box and leucine-rich repeat protein 6 2.05 1.10 1.11 69847 WNK lysine deficient protein kinase 4 2.05 1.12 1.19 230098 RIKEN cdna E130306D19 gene 2.05 1.25 1.22 257 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 73683 Autophagy related 16 like 2 (S. Cerevisiae) 2.05 -1.06 -1.03 108912 Cell division cycle associated 2 2.05 1.14 1.11 Killer cell lectin-like receptor, subfamily A, 16634 member 3 2.04 -1.00 -1.06 76432 RIKEN cdna 2310001H17 gene 2.04 1.05 -1.07 434903 Expressed sequence CN716893 2.04 -1.05 1.04 103733 Tubulin, gamma 1 2.04 1.00 1.23 98463 Expressed sequence A1851716 2.04 1.17 1.12 353165 Taste receptor, type 2, member 136 2.04 -1.29 -1.09 71774 Shroom fanny member 1 2.04 -1.01 -1.04 237073 RNA binding motif Eotein 41 2.04 -1.08 -2.02 78808 Syntaxin binding protein 5 (to_mosyn) 2.04 -1.14 -1 . 10 22619 Sialic acid acgtylesterase 2.04 1.17 1.15 494448 Chromobox homolog 6 2.04 -1.08 1.10 68490 Zinc finger protein 579 2.03 1.02 1.33 NIMA (never in mitosis gene a)-related 23955 expressed kinase 4 2.03 -1.07 1.14 Cystic fibrosis transmembrane conductance 12638 regulator homologfi 2.03 -2.26 1.1 1 229487 PET112-like (yeast) 2.03 1.48 1.41 19653 RNA binding motif protein 4 2.03 -1.06 -1.03 215641 Melanoma antigen family B, 18 2.03 -1.00 1.27 Potassium channel, subfamily V, 240595 member 2 2.02 -1.00 -1.22 56229 Thrombospondin, type 1, domain 1 2.02 1.02 -1.29 14149 F erredoxin reductase 2.02 1.07 1.56 68607 Serine hydrolase-like 2.02 1.44 1.36 56711 Pleiomorphic adenoma gene 1 2.01 -1. 16 -1.17 76511 RIKEN cdna 2010004M13 gene 2.01 -1.00 1.33 General transcription factor II H, 23894 polypeptide 2 2.01 1.07 1.05 68837 Forkhead box K2 2.01 1.01 -1.09 237211 Fanconi anemia, complementatirggroup B 2.01 -1.24 1.03 93728 Poly A binding protein, cytoplasmic 5 2.01 1.16 1.48 54141 Sperm associated antigen 5 2.01 1.61 1.03 67454 RIKEN cdna 1200009F10 gene 2.00 1.02 1.33 225283 Cdna sequence BC021395 2.00 -1.01 1.23 Tial cytotoxic granule-associated RNA 21843 binding protein-like 1 2.00 -l.12 1.21 69754 F-box protein 7 1.99 1.00 -1.01 101563 Expressed sequence A1426330 1.99 1.01 1.23 258 HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio 68053 RIKEN cdna 3110003A22 gene 1.99 1.07 1.41 Potassium channel tetramerisation domain 207474 containing 2b 1.99 1.06 1.22 78248 Armadillo repeat containing, X-linked 1 1.99 -1.04 -1.02 234686 Formin homology 2 domain containing 1 1.99 -1.15 -1.02 Visual system homeobox 1 homolog 114889 (zebrafish) 1.99 -1.06 -1.32 67493 Methyltransferase 10 domain containing 1.98 -1.30 1.20 3-hydroxymethyl-3-methylglutaryl- 208982 Coenzyme A 1yase-like 1 1.98 -1.16 1.50 Threonyl-tma synthetase 2, mitochondrial 71807 (putative) 1.98 -1.12 1.13 14302 bin-related kinase 1.98 -1.26 -1.02 64176 Synaptic vesicle glycoprotein 2 b 1.97 1.03 1.13 Developmentally regulated GTP binding 13494 protein 1 1.97 -1.36 -1.04 72852 RIKEN cdna 2900024010 gene 1.97 1.14 1.16 246293 Kelch-like 8 (Drosophila) 1.97 -1.10 -l.00 74042 RIKEN cdna 4921501E09 gene 1.97 -1.02 -1.07 Ubiquitin protein ligase E3 component n- 22222 recognin l 1.97 -1.05 1.08 68364 RIKEN cdna 0610030E20 gene 1.96 1.03 1.13 ST3 beta-galactoside alpha-2,3- 20454 sialyltransferase 5 1.96 -1.02 1.10 74498 Gplgi reassembly stacking protein 1 1.96 1.12 1.09 213409 LEM domain containing 1 1.96 -l.10 1.20 319806 RIKEN cdna D630022N01 gene 1.96 -1.01 1.13 233424 Transmembrane channel-like gene family 3 1.96 -1.30 -1.55 21390 Thromboxane A2 receptor 1.96 -1.04 -1.08 320230 RIKEN cdna C130023OIO gene 1.95 -1.00 1.19 668173 Peroxisome biogenesis factor 10 1.95 1.49 1.13 13829 Erythrocyte protein band 4.9 1.95 -1. 17 -1.34 104248 Calcineurin binding protein 1 1.95 -1 . 10 -1.11 SH3 domain and tetratricopeptide 225608 repeats 2 1.95 1.04 -1.05 103161 Apolipoprotein F 1.94 1.08 -6.60 Imprinted gene in the Prader-Willi 16353 syndrome gion 1.94 -1.47 -1.12 Gamma-aminobutyric acid (GABA-A) 57249 receptor, subunit theta 1.94 -1.00 -1.01 17684 Cbp/p300-interacting transactivator, 2 1.94 -1.03 1.62 68082 Dual specificity phosphatase 19 1.94 1.09 1.34 246104 Rhomboid, veinlet-like 3 (Drosophilg) 1.94 -1.23 1.06 259 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 245638 TBCl domain family, member 8B 1.94 1.22 1.18 68366 Transmembrane protein 129 1.93 1.01 -1. 15 ATP-binding cassette, sub-family G 192663 (WHITE), member 4 1.93 -1.31 1.01 218734 RIKEN cdna 3830406C13 gene 1.93 1.01 1.12 231642 Alkb, alkylation repair homolog 2 (E. Coli) 1.92 -1.03 1.08 54721 Tyrosine kinase 2 1.92 1.23 -1.04 113859 Vomeronasal 1 receptor, C2 1.92 1.07 -1.06 Musashi homolog 1 17690 (Drosophila) 1.92 -1.20 -1.20 27059 SH3 domain protein D19 1.92 -1.15 1.00 01igonucleotide/oligosaccharide-binding 109019 fold containingZA 1.92 -1.11 1.37 140500 Centaurin, beta 5 1.92 1.09 -1.01 232237 FYVE, rhogef and PH domain containing 1.92 1.07 1.03 18120 Mitochondrial ribosomal protein L49 1.92 1.27 1.04 77781 EPMZflaforin) interacting protein 1 1.91 1.08 -1.07 277463 G protein-coupled receptor 107 1.91 1.12 -1.02 21684 Tectorin beta 1.91 -1.26 -1.26 75425 RIKEN cdna 2610036D13 gene 1.91 1.28 1.08 17222 Anaphase promoting complex subunit 1 1.91 -1.00 1.09 231214 Coiled-coil and C2 domain containing 2A 1.91 1.14 1.28 CDC14 cell division cycle 14 homolog A 229776 (S. Cerevisiae) 1.91 -1.25 -1.00 Src homology 2 domain-containing 20420 transforming protein D 1.90 1.38 1.05 CDC23 (cell division cycle 23, yeast, 52563 homolog) 1.90 1.02 -1.11 Grpe-like 2, 17714 mitochondrial 1.90 1.36 1.85 Major facilitator superfamily domain 213006 containing 4 1.90 -1.20 -1.01 Chromodomain helicase DNA binding 71389 protein 6 1.90 -1.07 1.02 73373 Phosphatase, orphan 2 1.90 -1.00 -1.05 TEL2, telomere maintenance 2, homolog (S. 71718 Cerevisiae) 1.90 -1.10 -1.01 DEAH (Asp-Glu-Ala-Asp/His) box 106794 polypeptide 57 1.89 -1.21 1.06 72915 RIKEN cdna 2900017F054mne 1.89 -1.07 1.08 Fibrous sheath CABYR 623046 bind'fl protein 1.89 -1.00 -1.08 328801 Zinc finger protein 414 1.89 -1.11 1.12 233545 RIKEN cdna 2210018M11 gge 1.89 -1.27 1.18 260 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 224111 UBX domain containing 1.88 —1.04 1.14 70359 GTP binding protein 3 1.88 1.14 1.08 DCNl, defective in cullin neddylation 1, 102323 domain containing 2 1.88 -1.09 1.07 233335 Desmuslin 1.88 -1.46 1.19 20540 Solute carrier family 7, member 7 1.88 -1.07 1.37 101700 Tripartite motif-containing 68 1.88 -1.03 1.07 19213 Pancreas specific transcription factor, 1a 1.87 -1.06 -l .08 56032 Tumor suppressor candidate 4 1.87 1.10 1.15 19769 Ras-like without CAAX 1 1.87 -1.06 1.04 235606 Acylpeptide hydrolase 1.86 1.03 1.34 50915 Growth factor receptor bound motein 14 1.86 1.14 1.41 83563 Ubiquitin specific peptidase 26 1.86 -1.04 -1.00 Mannosyl (alpha-1,3-)-g1ycoprotein beta- 67569 1,4-NAG transferase 1.86 -1.24 -1.06 57890 Interleukin 17 receptor E 1.86 -1.29 -1.20 Trna-yw synthesizing protein 1 homolog (S. 100929 Cerevisiae) 1.86 1.01 -1.04 105351 Expressed sequence AW209491 1.86 -l.09 -1.02 23966 Odd Oz/ten-m homolog 4 (Drosophila) 1.86 1.10 1.01 225912 Cytochrome b, ascorbate dependent 3 1.86 -1.04 1.24 16351 IAP promoted placental gene 1.85 1.10 1.15 71609 TNFRSFlA-associated via death domain 1.85 -1.16 -l .03 DCNI , defective in cullin neddylation 1, 102323 domain containing 2 (S. Cerevisiae) 1.85 1.04 1.07 Pleckstrin homology domain interacting 83946 protein 1.85 1.24 1.37 73238 RIKEN cdna 3110049103 gene 1.85 -1.05 1.07 217026 HEAT repeat containing 6 1.85 -1.10 1.29 105000 Dynein, axonemal, light chain 1 1.85 -1.13 1.29 106064 Expressed sequence AW549877 1.85 1.09 1.36 215789 Phosphatase and actin regulator 2 1.85 1.21 1.33 208760 Aquaporin 12 1.85 -1.23 1.14 Phosphodiesterase 6D, cgmp-specific, rod, 18582 delta 1.84 1.10 1.05 Nudix (nucleoside diphosphate linked 67725 moiety X)-type motif 13 1.84 1.15 1.19 74356 RIKEN cdna 4931428FO4 gene 1.84 -1.07 1.16 Poly(A) binding protein, 241989 cytoplasmic 4-like 1.84 1.10 1.07 213988 Trinucleotide repeat containing6b 1.84 -1.05 -1.08 27059 SH3 domain protein D19 1.84 1.09 -1.07 261 HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio 246317 Neuropilin (NRP) and tolloid (TLL)-like 1 1.83 -1.08 -1.20 80890 Tripartite motif-containing 2 1.83 1.12 1.18 101214 Transformer 2 alpha homolog (Drosrmhila) 1.83 -1.18 1.25 Trna splicing endonuclease 54 homolog 76265 (SEN54, S. Cerevisiae) 1.83 -1.11 1.03 107375 Solute carrier family 25, member 45 1.83 1.04 -1.05 Progressive external ophthalmoplegia 1 226153 (human) 1.82 1.08 1.03 240186 Zinc finger protein 438 1.82 1.04 1.52 Chromobox homolog 1 (Drosophila HPl 12412 beta) 1.82 1.08 -1.13 69401 PLAC8-1ike 1 1.82 -1.02 -1.17 Similar to Spetex-2E 624633 protein 1.82 1.13 -1.01 13829 Erythrocyte protein band 4.9 1.81 -1.01 -1.10 67544 RIKEN cdna 4932442KO8 gene 1.81 -1.29 1.14 72656 Integrator complex subunit 8 1.81 1.08 -1.06 V-erb-b2 erythroblastic leukemia viral 13866 oncogene homolog 2 1.81 1.01 1.10 328801 Zinc finEr protein 414 1.81 -1.08 1.21 ST8 alpha-N-acetyl-neuraminide alpha-2,8- 20452 sialyltransferase 4 1.80 1.19 1.17 COP9 (homolog, subunit 7b (Arabidopsis 26895 thaliana) 1.80 -1.11 1.17 GRAM domain 235283 containing 1B 1.80 1.21 -1.18 16551 Kinesin family member 11 1.80 -1.07 -1.18 52463 Tet oncogene 1 1.80 1.08 -l.06 14625 Glycerol kinase-like 1 1.80 1.03 1.05 Branched chain ketoacid dehydrogenase 12041 kinase 1.80 1.19 -1.09 215951 Lactation elevated 1 1.80 1.19 1.24 Ankyrin repeat and zinc finger domain 52231 containing 1 1.80 1.01 1.12 Adenosine monophosphate deaminase 2 109674 (isoform L) 1.79 -1.21 1.07 212442 Lactamase, beta 2 1.79 1.41 1.14 Oligonucleotide/oligosaccharide-binding 108689 fold containing 1 1.79 1.08 1.07 Ankyrin repeat and zinc finger domain 52231 containing; 1.79 1.37 -1.08 215819 NHS-likel 1.79 1.13 1.21 192976 Cdna sequence BC046404 1.79 1.14 -1.23 66336 Centromere protein P 1.79 -1.01 1.02 262 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio Phosphatidylinositol glycan anchor 228812 biosynthesis, class U 1.79 1.07 1.15 69369 RIKEN cdna 1700017D01 gene 1.78 -l.69 1.12 Hexosaminidase (glycosyl hydrolase family 238023 20, catalytic domapi) containing 1.78 1.17 1.05 69807 Tripartite motif-containing 32 1.78 1.11 1.24 69089 Oxidase assembly 1-1ike 1.78 1.08 1.23 194744 Solute carrier family 25, member 43 1.78 -1.03 -1.13 229759 Olfactomedin 3 1.78 1.11 -1.00 208098 Pannexin 3 1.78 -1.02 -1.35 56771 Mediator complex subunit 20 1.78 1.05 -1.04 Solute carrier family 16 (monocarboxylic 72472 acid transporters), member 10 1.78 -1.12 1.02 2-4-dienoyl-Coenzyme A reductase 2, 26378 peroxisomal 1 .78 1 .24 1 .07 100473 Expressed sequence BBO31773 1.78 1.03 1.53 226539 Aspartyl-trna synthetase 2 (mitochondrial) 1.78 1.04 1.27 NUF2, NDC8O kinetochore complex 66977 component, homolog (S. Cerevisiae) 1.77 -1. 14 1.08 13175 Doublecortin-like kinase 1 1.77 1.30 1.00 Bromodomain adjacent to zinc finger 116848 domain, 2A 1.77 -1.07 -1.05 Far upstream element (FUSE) binding 51886 protein 1 1.77 -1.18 1.02 24071 Synaptojanin 2 binding protein 1.77 1.01 1.30 BCL2/adenovirus EIB interacting protein 1, 224630 NIPl 1.76 1.35 1.06 567 71 Mediator complex subunit 20 1.76 1.21 1.03 Paroxysmal nonkinesiogenic 56695 dyskinesia 1.76 1.24 1.29 Coactivator-associated arginine 59035 methyltransferase 1 1.76 -1.08 1.11 171254 Vomeronasal 1 receptor, 13 1.76 1.06 1.26 80890 Tripartite motif-containirg 2 1.76 1.11 1.00 227624 RIKEN cdna BZ30208H17 gene 1.76 1.01 1.05 RGPl retrograde golgi transport homolog 242406 (S. Cerevisiae) 1.76 -1.04 1.03 SWI/SNF related, matrix associated, 67155 subfamily a, member 2 1.76 -1.38 1.08 54201 Zinc finger protein 316 1.76 1.21 1.21 207213 TD and P02 domain containingl 1.75 -1.08 -1.10 . ATP-binding cassette, sub-family B 74610 (MDR/TAP), member 8 1.75 1.40 1.03 Anaphase promoting complex 17222 subunit 1 1.75 1.07 1.22 263 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 494448 Chromobox homolog 6 1.74 -1.01 1.09 76547 Transmembrane protein 101 1.74 1.16 1.02 Cytidine monophosphate (UMP-CMP) 22169 kinase 2, mitochondrial 1.74 1.22 1.09 380669 Lin-28 homolog B (C. Elegans) 1.74 -1.18 -1.18 68364 RIKEN cdna 0610030E20 Erie 1.74 -l .05 1.11 21685 Thyrotroph embryonic factor 1.73 1.46 1.27 74653 RIKEN cdna 4930444A02 gene 1.73 1.00 1.27 234371 Transmembrane protein 161A 1.73 -1.07 1.11 210035 Transmembrane protein 194 1.73 1.15 1.21 258872 Olfactory receptor 908 1.73 -1.01 -1.00 67230 Zinc finger mtein 329 1.73 1.06 -1.19 RAB7, member RAS oncogene family-like 226422 1 1.73 -1.14 1.09 52710 G protein-coupled receptor 1723 1.72 -1.06 1.18 Cell division cycle 27 homolog (S. 217232 Cerevisiae) 1.72 -1.06 -1.06 73747 RIKEN cdna 1110034G24 gene 1.72 1.02 1.10 78323 RIKEN cdna 2310046006 gene 1.72 -1.03 1.04 245638 TBCI domain family, member SB 1.72 -1.07 1.12 Rap guanine nucleotide exchange factor 76089 QEF) 2 1.72 1.00 -1.11 258752 Olfactory receptor 583 1.72 -3.03 1.18 331195 RIKEN cdna A430089119gene 1.71 1.18 -1.00 76429 RIKEN cdna 2310007H09 gene 1.71 -1.02 1.14 FUS interacting protein (serine-arginine 14105 rich) 1 1.71 -1.26 1.10 76788 RIKEN cdna 2410127E18 gene 1.71 1.05 -1.02 Arginyl aminopeptidase 215615 (aminopeptidase B) 1.71 1.03 1.33 108899 RIKEN cdna 2700081015 gene 1.71 -1.06 -1.07 Phosphatidylinositol glycan anchor 70325 biosynthesis, class W 1.71 1.14 -1.09 78913 Zinc finger protein 294 1.71 -1.05 1.1 1 Mitochondria-associated protein involved in 66449 GM-CSF signal transduction 1.71 -1.01 1.27 208198 BTB (POZ) domain containifl 2 1.70 1.01 1.09 Myelin-associated oligodendrocytic basic 17433 protein 1.70 1.02 -1 . 12 19155 Aminopeptidase puromycin sensitive 1.70 1.06 1.20 56187 Rab geranylgeranyl transferase, a subunit 1.70 1.10 1.11 13185 Down syndrome critical region gene 3 1.70 -1.01 1.08 320727 Importin 8 1.70 -1.05 1.02 264 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 65102 Nggl interacting factor 3-1ike 1 (S. Pombe) 1.70 -1.18 1.03 171235 Vomeronasal 1 receptor, F4 1.70 -1.16 -1.07 Rho/rac guanine nucleotide exchange factor 102098 (GED 18 1.70 -1.00 1.05 68936 RIKEN cdna 1190017012 gene 1.69 -1.08 1.08 17532 Muscle and microspikes RAS 1.69 -1.21 1.25 237636 NPCl-like 1 1.69 -1.01 1.01 14050 Eyes absent 3 homolog(Drosophila) 1.69 1.04 1.05 Phosphodiesterase 4D interacting protein 83679 (myomegalin) 1.69 -1.06 1 .07 Phosphoinositide-3-kinase, regulatory 104709 subunit 6 1.69 1.04 -1 . 17 170753 Zinc finger protein 704 1.69 -1.34 -1.02 67105 RIKEN cdna 1700034H14pgene 1.69 -1.01 1.13 214498 Cell division cycle 73 1.69 -1.16 1.04 Polymerase (DNA—directed), epsilon 4 (p12 66979 subunit) 1.69 -1 . 19 1.15 54131 Interferon regulatory factor 3 1.68 -1.28 -1.11 Par-6 (partitioning defective 6,) homolog 56513 plpha (C. Elegans) 1.68 1.01 1.06 29808 MAX gene associated 1.68 1.02 -1.01 Processing of precursor 5, ribonuclease 117109 PfMRP family LS. Cerevisiae) 1.68 -1.11 1.01 240899 Leucine rich repeat containing 52 1.68 -1.26 -2.55 277562 Olfactory receptor 1286 1.67 -1.00 -1 . 18 71701 Polyribonucleotide nucleotidyltransferase 1 1.67 1.12 1.20 Mitochondrial translational initiation factor 76784 2 1.67 1.20 1.03 76894 Methyltransferase 5 domain containing 1 1.67 1.17 1.29 59015 Nucleoporin 160 1.67 -1.01 -1.08 Solute carrier family 4 (anion exchanger), 20534 member 1, adaptor protein 1.67 -1.04 -1.13 BCL2/adenovirus ElB interacting protein 1, 224630 NIPI 1.67 1.13 1.08 lmmunoglobulin superfamily containing 320563 leucine-rich repeat 2 1.67 -1.52 -1.13 Epidermal grth factor receptor pathway 13859 substrate 15-like 1 1.66 -1.03 -1.03 Solute carrier family 30 (zinc transporter), 210148 member6 1.66 -1.14 1.16 Transcriptional adaptor 2 (ADA2 homolog, 217031 yeast)-like 1.66 -1.01 1.1 1 103012 RIKEN cdna 6720401G13 gene 1.66 1.32 1.16 78408 RIKEN cdna 2900046G09 gfle 1.66 -1.01 1.22 265 HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio 69757 Leukocyte receptor cluster (LRC) member 1 1.65 1.16 1.15 23897 HCLSl associated X-l 1.65 -1.04 1.00 Mitogen-activated protein kinase kinase 26407 kinase 4 1.65 -1.07 1.12 66574 RIKEN cdna 2510017J16g'pne 1.65 1.48 -1.05 13644 Embryonal F yn-associated substrate 1.64 -1.04 1.25 18854 Promyelocgic leukemia 1 .64 1 .06 1. 12 244895 RIKEN cdna C230081A13 gene 1.64 1.11 1.01 CAP-GLY domain containing linker protein 78785 family, member 4 1.64 1.46 1.12 258409 Olfactory receptor 1431 1.64 -1.00 -1.02 74114 Carnitine O-octanoyltransferase 1.64 1.09 1.07 268281 SNF2 histone linker PHD RING helicase 1.64 -1.03 1.09 56695 Paroxysmal nonkinesiogenic dyskinesia 1.64 1.22 1.13 236732 RNA binding motif protein 10 1.63 -l .07 -1.06 71382 Peroxisome biogenesis factor 1 1.63 -1.06 1.04 78323 RIKEN cdna 2310046006 gene 1.63 -1.03 -1.13 547150 Similar to p47 protein 1.63 -1 . 10 1.24 BRCA1/BRCA2-containing complex, 210766 subunit 3 1.63 1.03 1.16 18174 Solute carrier family 11 , member 2 1.63 -1.33 1.13 330963 Predicted gene, EG330963 1.63 1.11 1.23 66050 RIKEN cdna 0610009322 gene 1.63 1.04 1.18 Cytosolic iron-sulfur protein assembly 1 26371 homolog (S. Cerevisiae) 1.63 -1.02 -1.07 72016 RIKEN cdna 1600002H07 gene 1.63 -1.05 -1.11 12338 Calpain 6 1.62 1.66 1.02 66609 Crystallin, zeta (quinone reductase)-like 1 1.62 1.27 1.09 17101 Lysosomal traffickinmulator 1 .62 -1 .07 1.01 Digeorge syndrome critical region 94223 gene 8 1.62 -1.00 -1.10 319953 Tubulin tyrosinejgase-like 1 1.62 1.09 1.12 223483 Hypothetical protein A830021M18 1.62 -1.16 -l.25 15468 Protein gginine N-methyltransferase 2 1.62 -1.24 1.06 Phosphatidylinositol-specific phospholipase 239318 C, X domain containing 3 1.62 -1.20 -1 .05 67892 RIKEN cdna 1810063B05 ggc 1.61 1.01 1.20 NIMA (never in mitosis gene a)-related 23954 expressed kinase 3 1.60 1.14 1.03 Translocase of inner mitochondrial 30057 membrane 8 homologb (yeast) 1.60 1.05 1.23 330050 Expressed sequence A1847670 1.60 1.18 1.22 266 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio Ligand dependent nuclear receptor 209707 corepressor-like 1.60 -1 .03 1 .02 Nicotinamide nucleotide adenylyltransferase 66454 1 1.60 -1.07 1.11 193796 Jumonji domain containingiB 1.60 1.18 -1.17 72320 RIKEN cdna 2510003E01gme 1.60 -1.03 1.11 Protein kinase, camp dependent, catalytic, 18749 beta 1.60 -1.02 1.13 72904 RIKEN cdna 2900034E22 gene 1.59 -l.13 -1.04 442825 RIKEN cdna A230083G16 gene 1.59 1.10 1.00 Insulin-like growth factor 2 ma binding 140486 protein 1 1.59 1.19 -1.01 68223 RIKEN cdna 1700063117 gene 1.59 -1.22 -2.83 HECT domain and ankyrin repeat 209462 containing, E3 ubiquitin protein ligase 1 1.59 1.01 1.31 52856 GTP binding protein 5 1.58 1.16 -1.07 74385 RIKEN cdna 4932432K03 gene 1.58 1.01 1.21 COX19 cytochrome c oxidase assembly 68033 homolog (S. Cerevisiae) 1.58 -1.09 1.00 Cell division cycle 25 homolog A (S. 12530 Pombe) 1.58 1.03 1.11 Thioredoxin domain containing 3 73412 (spermatozoa) 1.58 1.13 -1.10 20892 Stimulated by retinoic acid 13 1.58 1.08 1.15 Zinc finger (CCCH type), RNA binding 22184 motif and serine/arginine rich 2 1.58 1.14 1.16 79264 KRITI, ankyrin repeat containing 1.58 -1.21 -1.16 Pleckstrin homology domain containing, 269608 member 5 1.58 1.14 -1.01 58805 MLX interacting protein-like 1.57 1.42 1.03 171236 Vomeronasal 1 receptor, F5 1.57 -1.00 -1.00 57443 F-box protein 3 1.57 -1.04 1.11 Alkb, alkylation repair homolog 8 67667 (E. Coli) 1.57 1.09 -l .1 1 75632 RIKEN cdna 1700003011 gene 1.57 -1.46 1.01 68691 RIKEN cdna 1110028C15gene 1.57 -1.11 1.26 Ankyrin repeat and SOC S box-containing 78910 protein 15 1.56 -1.04 -1.00 241556 Tetraspanin 18 1.56 1.07 2.29 70733 RIKEN cdna 6330411EO7 gene 1.56 -1.17 -1.15 22061 Transformation related protein 63 1.56 -1.40 -1.08 244417 Gene model 501, (NCBI) 1.56 1.04 -1.19 COX19 cytochrome c oxidase assembly 68033 homolcg (S. Cerevisiae) 1.56 -l .08 1.13 267 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 67784 Plexin D1 1.55 1.28 -1.13 16882 Ligase 111, DNA, ATP-dependent 1.55 -1.16 -1. 18 Phosphatidylinositol glycan anchor 18701 biosynthesis, class F 1.55 -1.10 1.16 Golgi associated, gamma adaptin ear 260302 containing, ARF bindflgprotein 3 1.55 -1.12 1.22 Origin recognition complex, subunit 4-like 26428 (S. Cerevisiae) 1.55 1.11 1.26 CD244 natural killer cell 18106 receptor 2B4 1.55 -1.27 -l.15 Mitogen-activated protein kinase kinase 26407 kinase 4 1.55 -1.07 1.18 CAP-GLY domain containing linker protein 76686 3 1.55 1.21 1.34 75458 Chemokine-like factor 1.55 -1.14 1.06 Amyloid beta (A4) precursor protein- 225372 binding, family B, member 3 1.55 1.10 -1.04 21853 Timeless homolog(Drosophila) 1.55 1.24 -1.11 Melanoma associated antigen 68114 (mutated) 1 1.55 -1.11 -1.08 16650 Karyopherin (importin) alpha 6 1.54 -1.04 1.06 UDP-glcnaczbetagal beta-1 ,3-N- 227327 acetmlucosaminyltransferase 7 1.54 1.10 -1.39 68347 RIKEN cdna 0610011F06 gene 1.54 1.13 1.12 66880 Arginine/serine-rich coiled-coil 1 1.54 1.02 1.06 83429 Cystinosis, nephropathic 1.54 1.04 -1.12 258334 Olfactory receptor 1396 1.54 1.84 1.02 12724 Chloride channel 2 1.53 -1.14 1.05 104859 RIKEN cdna 4930573119 gene 1.53 -1.10 -1.02 MAP/microtubule affmity-regulating kinase 13728 2 1.53 -1.02 -1.04 1E+08 Predicted gene, ENSMUSG00000052469 1.53 1.02 -1.20 57837 Era (G-protein)-like 1 (E. Coli) 1.53 1.07 -1.05 22702 Zinc finger protein 42 1.53 -1.08 1.19 75339 M-phase phosphoprotein 8 1.53 1.15 1.10 258402 Olfactory receptor 1079 1.53 -1.00 -1.80 170762 Nucleoporin 155 1.53 1.01 -1.04 67707 Mitochondrial ribosomal protein L24 1.53 1.25 1.30 56700 RIKEN cdna 0610031106 gene 1.52 1.03 1.10 677296 Fc receptor-like 6 1.52 9.00 1.09 73327 RIKEN cdna 1700040103gpne 1.52 1.18 1.04 321006 Vpr (HIV-l) binding protein 1.52 -1.19 1.05 1E+08 Butyrophilin-like 1 1.52 -1.18 -1.08 268 1 . ‘. HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio 227154 Amyotrophic lateral sclerosis 2 (juvenile) 1.52 1.01 1.22 Nicotinate phosphoribosyltransferase 223646 domain containing 1 1.51 1.27 1.26 NIMA (never in mitosis gene a)-related 59125 expressed kinase 7 1.51 1.35 -l .03 Smg-6 homolog, nonsense mediated mrna 103677 decay factor (C. Elegans) 1.51 1.00 -1.00 320816 Ankyrin repeat domain 16 1.51 1.25 1.36 RNA pseudouridylate synthase domain 71989 containing4 1.51 —1.06 1.10 237943 G patch domain containing 8 1.51 -1.02 1.11 72519 Transmembrane protein 55A 1.51 1.06 1.05 12828 Coflpgen, type IV, alpha 3 1.51 1.98 1.35 60364 Downstream neighbor of SON 1.51 -1.04 -1.02 227326 G protein-coupled receptor 55 1.51 1.03 1.29 67706 Transmembrane protein 179B 1.51 -1.02 1.00 Tubulin, gamma complex associated protein 328580 6 1.50 1.07 1.03 231003 Kelch-like 17 (Drosophila) 1.50 1.06 1.07 216578 Poly(A) polymerase gamma 1.50 1.15 -1.56 216119 RIKEN cdna A130042E20 Elle 1.50 1.05 -1.06 Coagulation factor C homolog (Limulus 12810 polyphemus) 1.50 1.54 1.10 225998 RAR-related orphan receptor beta 1.48 1.85 1.14 258457 Olfactory receptor 108 1.47 1.86 -1.16 Thrombospondin, type 1, domain containing 210417 7B 1.43 -1.00 -1.98 14654 Glycine receptor, alpha 1 subunit 1.43 -l .02 1.83 72892 RIKEN cdna 2900018K06 gene 1.43 1.98 -1.11 70950 RIKEN cdna 49215281014ene 1.38 1.96 1.09 71248 RIKEN cdna 4933428MO9 gene 1.38 -1.17 -1.82 75506 RIKEN cdna 1700017107 gene 1.38 -1.88 1.17 229707 RIKEN cdna 6330569M22 gene 1.37 -1.69 -1.21 13491 Dopamine receptor 4 1.36 -1.90 1.02 Fumarylacetoacetate hydrolase domain 68126 containing 2A 1.36 1.60 1.30 258973 Olfactory receptor 1228 1.35 -2.55 -1.11 Non imprinted in Prader-Willi/Angelman 93790 syndrome 2 homolog (humari 1.35 -1.14 1.55 Apoptosis-inducing, TAF9-like 69928 domain 1 1.33 2.04 1.11 24057 Sh3 domain YSC-like 1 1.33 1.05 2.51 239368 Cdna sequence BC030476 1.30 1.14 -2.05 269 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 27389 Dual specificity phosphatase 13 1.27 -1.51 1,05 258562 Olfactotmceptor 1014 127 -227 -1 .03 Cyclic nucleotide gated 12788 channel alpha 1 1.27 -1.73 -1.33 74967 RIKEN cdna 4930474H20 gene 1.26 1.23 -2.21 67944 RIKEN cdna 1700025DO3 gene 1.26 -1.12 2.89 13176 Deleted in colorectal carcinoma 1.25 2.20 -1.04 320899 RIKEN cdna A430110C17 gene 1.24 1.75 1.15 170725 Calpain 8 1.23 1.58 -1.00 170654 Keratin associated protein 16-4 1.23 -1.67 -1.02 RAB, member of RAS oncogene family-like 67286 5 1.23 1.63 1.05 237310 Interleukin 22 receptor, alpha 2 1.22 -1.18 2.25 Protein tyrosine phosphatase, receptor type, 19264 C 1.22 -2.29 1.07 14177 Fibroblast growth factor 6 1.22 -1.16 -2.17 71229 RIKEN cdna 4933428P19 gene 1.22 1.06 2.56 328918 RIKEN cdna C230097124 gene 1.21 -1.76 1.21 80986 Cytoskeleton associated protein 2 1.21 1.76 1.01 Growth arrest and DNA-damage-inducible, 102060 amma interactig protein 1 1.21 1.38 -1.95 Retinitis pigmentosa gtpase regulator 77945 mteracfipg protein 1 1.21 1.55 -1.08 DNA segment, Chr 16, human D22868OE, 27883 expressed 1.20 1.54 1 . 15 224480 NADPH oxidase 3 1.19 -1.59 -1.21 16184 Interleukin 2 receptor, alpha chain 1.19 -1.24 1.59 Cytoplasmic polyadenylation element 67579 binding protein 4 1.19 -2.11 1.04 Potassium voltage-gated channel, Shal- 16508 related family, member 2 1.19 2.35 1.39 71701 Polyribonucleotide nucleotidyltransferase 1 1.19 2.35 1.08 382059 Defensin related cryptdin 22 1.18 2.01 1.13 111174 Trace arnine-associated receptor 1 1.18 -1.32 -2.33 258356 Olfactory receptor 564 1.18 1.06 -2.74 DEXH (Asp-Glu-X-His) box 80861 polypeptide 58 1.17 1.05 -2.47 ATP-binding cassette, sub-family A 27403 (ABC1), member 7 1.17 -1.78 1.01 71388 RIKEN cdna 5530401A14gene 1.17 -2.03 -1.04 319720 RIKEN cdna 9630028104 gene 1.15 1.04 -1.74 241656 P21 (CDKNIA)-activated kinase 7 1.14 1.92 -1.09 258555 Olfactory receptor 862 1.12 2.06 1.19 270 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio DNA segment, Chr 3, ERATO Doi 300, 56790 expressed 1 .1 1 -1 .00 -2.13 74438 Retinaldehyde binding protein l-like 1 1.11 -1.10 -1.63 70147 RIKEN cdna 2210019111gene 1.09 -1.14 -1.56 ATP-binding cassette, sub-family A 381072 (ABC1), member 17 1.09 5.12 1.17 229521 Synaptotagmin XI 1.09 -1.98 -1.11 101533 Kallilqein related-peptidase 9 1.08 -1.38 1.79 110606 Farnesyltransferase, CAAX box, beta 1.06 1.87 1.14 245847 Amidohydrolase domain containing 2 1.05 2.46 1.29 328839 Predicted gene, EG328839 1.05 -1.85 1.10 14057 Sideroflexin 1 1.04 -1.62 -2.00 70954 RIKEN cdna 4922502801 ggc 1.04 -1.00 -1.69 Chromobox homolog 3 (Drosophila HPl 12417 gamma) 1.04 -2.57 -1.03 258822 Olfactory receptor 39 1.04 1.36 2.62 258944 Olfactory receptor 351 1.03 -1.84 1.02 100647 Uroplakin 33 1.02 1.15 -1.52 258393 Olfactory receptor 1325 1.02 -1.01 1.52 20234 Spermine binding protein 1.02 1.93 1.10 NADH dehydrogenase (ubiquinone) Fe-S 226646 protein 2 1.01 1.71 1.01 76633 RIKEN cdna 1700112E06 gene 1.00 -2.39 1.07 269587 Erythrocyte protein band 4.1 -1.01 -1.08 -1.72 Cytochrome P450, family 2, subfamily j, 13110 polypeptide 6 -1.02 1.57 -1.05 G-rich RNA sequence 231413 binding factor 1 -1.02 1.57 1.06 74916 RIKEN cdna 1700066022 gene -1.04 -1.85 -1.03 75231 RIKEN cdna 4930529122 gene -1.04 3.03 -1.21 Biregional cell adhesion molecule-related 117606 binding protein -1.06 1.51 1.01 12411 Cystathionine beta-synthase -1.06 1.72 1.17 16675 Keratin 27 -1.08 -2.71 1.03 21673 Deoxynucleotidyltransferase, terminal -1 .08 -2.26 -l .24 77900 RIKEN cdna 6720473M11 gene -1.08 1.34 -1.93 Alkb, alkylation repair homolog 3 69113 (E. Coli) -1.09 1.61 -1.14 11898 Argininosuccinate synthetase 1 -1.12 1.61 1.20 Proteasome (prosome, macropain) subunit, 19177 beta type7 -1.12 1.69 1.10 Solute carrier family 6 (neurotransmitter 21366 transporter, taurine), member 6 -1.13 1.11 -1.69 271 HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio 76117 Rho gtpase activat'flprotein 15 -1.16 -243 -1 .00 Transformation related protein 53 inducible 277414 protein 11 -1.17 2.04 -1.07 75089 UHRFl (ICBP90) binding protein 1-like -1.17 -1.19 -1.53 74841 Ubiquitin specific peptidase 38 -1.18 1.19 -1.75 76937 RIKEN cdna 2810429104 gene -1.18 -1.71 -1.05 17339 Major intrinsic protein of eye lens fiber -l.18 -1.23 1.79 Enoyl Coenzyme A hydratase, short chain, 93747 1, mitochondrial -1.19 1.52 -1.17 320500 RIKEN cdna A930001M 12 gene -1.21 1.06 -1.95 Succinate-coa ligase, GDP-forming, alpha 56451 subunit ' -1.21 1.64 1.00 170788 Crumbs homolog 1 (Drosophila) -1.22 -2.56 -1.05 109054 Prefoldin 4 -1.26 1.51 1.29 381062 RIKEN cdna 221040411 1 gene -1.29 1.70 -1.07 Aldehyde dehydrogenase 1 family, member 107747 L1 -1.30 1.83 -1.17 Kv channel interacting protein 3, 56461 calsenilin -1.31 1.98 1.11 102527 RIKEN cdna 6720458D17 gene -1.31 -1.13 -1.76 69354 Solute carrier family 38, member 4 -1.35 2.14 -1.19 548632 Cdna sequence BC023719 -1.35 3.09 1.27 66898 BAIl-associated protein 2-like l -1.39 1.64 -1.13 234875 Tetratricopeptide repeat domain 13 -1.50 1.14 -1.31 20403 Intersectin 2 -1.50 1.12 -1.24 20788 Sterol Egplatory element binding factor 2 -1.51 1.05 -1.23 14390 GA repeat binding protein, alpha -l.51 1.13 -l.13 RNA terminal phosphate cyclase 66368 domain 1 -1.51 1.10 -1.11 212111 Inositol polyphosphate-S-phosfltase A -1.51 1.17 -1.09 Tetratricopeptide repeat, ankyrin repeat and 66860 coiled-coil containing 1 -l.51 1.36 -1.35 216164 Downstream ofStkll -1.52 1.12 -1.04 171580 Microtubule associated monoxygenase, 1 -1.53 -1 . 12 -1.27 18321 Olfactory receptor 23 -1.53 2.05 1.00 75608 Chromatin modifying protein 4B -1 .53 -l.02 -1.10 11566 Adenylosuccinate synthetase, non muscle -1.53 1.07 -1.16 80914 Uridine-cytidine kinase 2 -1.53 -1-22 '1-03 53334 Golgi SNAP receptor complex member 1 -1.54 1.44 -1.19 Translocase of outer mitochondrial 28185 membrane 70 homolog A (yeast) -1.54 1.24 -1.25 21985 Tumor protein D52 -1.54 1.21 -1.16 272 HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio Serine (or cysteine) peptidase inhibitor, 20317 clade F, member 1 -l .55 1.11 -1.32 TRAF family member-associated Nf-kappa 21353 B activator -1.55 1.13 -1.04 100273 Oxysterol binding protein-like 9 -1.55 1.65 -1.32 75007 RIKEN cdna 4930504E06 gene -1.56 1.18 -1.47 Protein phosphatase 2 (formerly 2A),alpha 51792 isoform -1.56 1.38 -1.17 EGF-like repeats and discoidin I-like 13612 domains 3 -1.56 -1.00 1.02 Receptor-interacting serine-threonine kinase 56532 3 -1.56 -1.07 -1.27 Par-6 partitioning defective 6 homolog 93737 gamma (C. Elegans) -1.56 1.17 -1.26 Syntaxin binding protein 5 78808 (tomosyn) -1.56 1.13 -l .26 U2 small nuclear ribonucleoprotein auxiliary 108121 factor (U2AF) 1 -1.57 1.21 -1.09 76273 Nedd4 family interacting protein 2 -1.57 1.21 -1.19 94088 Tripartite motif-containing 6 -1.57 1.57 1.55 Survival motor neuron domain 76479 containing 1 -1.57 1.15 -1.08 Solute carrier family 36 (proton/amino acid 234967 symporter), member 4 -1.58 1.17 -1.15 Suppressor of variegation 4-20 homolog 1 225888 (Drosophila) -1.58 1.01 -1.26 11908 Activating transcription factor 1 -1.58 1.15 -1.22 Taxl (human T-cell leukemia virus type I) 52440 binding protein 1 -1.58 1.39 -1.22 F-box and WD-40 domain protein 7, 50754 archipelago homolog (Drosophila) -1.58 1.23 -1.29 57912 CDC42 small effector 1 -l.59 1.15 -1.33 Shwachman-Bodian-Diamond syndrome 66711 homolog (human) -1.60 1.41 -1.30 70834 Sperm associated antigen 9 -1.60 1.03 -1.49 Src homology 2 domain-containing 214547 transforminflrotein E -1.60 1.07 -1.55 77929 Yipl domain family, member 6 -l .60 1.20 -1.07 71472 Ubiquitin specific peptidase 19 -1.60 1.27 -1.24 50770 Atpase, class VI, type 11A -1 .61 -1.04 -1.40 66674 RIKEN cdna 6330409N04 gene -1.61 1.17 -1.09 233315 Myotubularin related protein 10 -1.61 1.01 -l.23 ' Proteasome (prosome, macropain) 26443 subunit, alpha type 6 -1.61 1.17 -1.08 Acyl-Coenzyme A binding domain 74159 containing 5 -1.62 -1.00 -1.13 273 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 14118 Fibrillinl -1.63 1.12 1.04 224691 Zinc finger protein 472 -1.64 1.19 -1.13 69329 RIKEN cdna 1700003Mlflene -1.64 1.26 1.03 Partner of NOB] homolog 66249 (S. Cerevisiae) -1.64 -1.03 -1.21 Protein tyrosine phosphatase, mitochondrial 66461 1 -1.64 1.01 -1.08 20277 Sodium channel, nonvoltagggated 1 beta -1.64 1.36 -1.19 208449 Sphingomyelin synthase 1 -l .64 1.15 -1.29 16803 Lipopolysaccharide binding protein -1.65 1.33 -1.44 76267 Fatty acid desaturase 1 -1.65 1.43 -1.17 12913 Camp responsive element bindingprotein 3 -1.66 1.24 -1.40 Protein phosphatase 1B, magnesium 19043 dependent, beta isoform -1.66 1.35 -1.42 Castor homolog 1, zinc finger 69743 (Drosophila) -1 .66 -1 .06 -1.27 242691 G patch domain contairfl 3 -1.66 1.18 -1.23 27984 EF hand domain containing 2 -1.66 1.17 -1.28 56389 Syntaxin 5A -1.66 1.26 -1.34 RNA terminal phosphate 59028 cyclase-like 1 -1.66 1.04 -1.55 228859 RIKEN cdna D930001122 gene -1.67 1.00 -1.30 16923 SHZB adaptor protein 3 -1.67 1.09 -1.29 98238 Leucine rich repeat containing59 -1.67 1.22 -1.45 55963 Solute carrier family 1 , member 4 -1.67 1.16 -1.25 General transcription factor 11F, polypeptide 68705 2 -1.67 1.15 -l.22 18570 Programmed cell death 6 -l .67 1.14 -1.20 Ring finger and CHY zinc finger domain 68098 containing 1 -l.67 1.11 -1.15 79555 Cdna sequence BC005537 -1.68 1.12 -1.27 210529 RIKEN cdna G430022H21 gene -1.68 -1.00 1.32 70354 RIKEN cdna 3110001120 gene -1.68 1.16 -1.45 12834 Collagen, type V1, alpha 2 -1.69 -1.20 -1.05 22688 Zinc finger protein 26 -1.69 1.03 -1.01 Regulatory factor X, 2 (influences HLA 19725 class II expression) -1.69 -l .07 -1.06 77446 HEG homologlizebrafish) -1.69 1.15 -1.50 226352 Erythrocyte motein band 4.1-like 5 -1.69 1.21 -1.29 258908 Olfactory receptor 853 -1.70 -l .30 -1.29 66193 RIKEN cdna1110049F12 gene -1.70 1.29 -1.11 22022 Protein-tyrosine sulfotransferase 2 -1.70 1.12 -1.24 274 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio Protein phosphatase 1, regulatory (inhibitor) 232807 subunit 12C -1.70 1.25 -1.31 215449 RAS related protein 1b -1.71 1.07 -l .10 15007 Histocompatibility 2, Q regn locus 10 -1.71 1.44 -1.38 98758 Heterogeneous nuclear ribonucleoirotein F -1.71 1.23 -1.27 54170 Ras-related GTP bind'g C -1.71 1.36 -1.43 Protein phosphatase 1A, magnesium 19042 dependent, alpha isoform -1.72 1.30 -1.49 56494 Golgi SNAP receptor complex member 2 . -1.72 1.26 -1.44 258573 Olfactory receptor 1020 -1.72 -1.00 -1.03 Sema domain, transmembrane domain 214968 (TM), (semaphorin) 6D -1.72 1.24 -1.60 Retinol dehydrogenase 14 (all-trans and 9- 105014 cis) -1.72 -1.13 -1.18 CCR4-NOT transcription complex, subunit 78893 10 -1.73 1.59 -1.13 100494 Zinc finger, ANl-type domain 2A -1.73 1.08 -l.14 Eukaryotic translation initiation factor IA 69860 domain containing -1.73 1.36 -1.51 56722 LPS-induced TN factor -1.73 1.25 -1.23 Protein tyrosine phosphatase, receptor type, 19267 E -1.74 -1.28 -1.39 F asciculation and elongation protein zeta 2 225020 (zygin II) -1.74 1.45 -1.43 20437 Seven in absentia 1A -1.74 1.37 -1.06 Fusion, derived from t(12;l6) malignant 233908 liposarcoma (human) -1.74 1.57 -1.30 Platelet/endothelial cell adhesion 18613 molecule 1 -l.76 1.33 -1.43 68904 Abhydrolase domain containifl 13 -1.76 1.35 -1.41 1 10351 Rapl gtpase-activatingp'otein -1.76 1.35 -1.46 223455 Membrane-associated ring finger (C3HC4) 6 -1.76 1.37 -l .11 108030 Lin-7 homolog A (C. Elegans) -1.76 -1.17 1.02 DEAD (Asp-Glu-Ala-Asp) box polypeptide 56200 21 -1.77 -1.01 -1.29 208659 Cdna sequence BC029169 -1.77 -1.08 -1.25 Zinc finger, MYND domain containing 67187 19 -1.77 1.06 -1.50 242418 WD repeat domain 32 -1.78 1.09 -1.43 217410 Tribbles homolog 2 (Drosophilp) -1.78 1.17 -1.57 192897 Wu beta 4 -1.78 -1.24 -1.08 70611 F-box protein 33 -1.78 -1.06 -1.31 Potassium channel tetramerisation domain 239217 containing 12 -1.79 1.03 -1.07 275 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 22156 Tuftelin 1 -1.79 1.28 -1.21 110651 Ribosomal protein S6 kinase polypeptide 3 -1.79 1.30 -1.35 74504 RIKEN cdna 2410018C17 gene -1 .80 1.20 -1.32 223827 Glycosyltransferase 8 domain containifi 3 -1.80 -1 . 10 1.03 67010 RNA binding motif protein 7 -1.80 1.08 -1.28 17912 Myosin [B -1.80 1.35 -1.20 Heterogeneous nuclear ribonucleoprotein U- 232989 like 1 -1.80 1.29 -1.08 12616 Centromere protein B -l .81 1.37 -l.46 Ubiquitin-conjugating enzyme E2D 3 66105 (UBC4/5 homolog, yeast) -1.81 1.25 -1.22 18196 Neuron specific gene family member 1 -1.81 -1.08 -1.44 28035 Ubiquitin specific peptidase 39 -1.81 1.28 -1.22 Solute carrier family 2 (facilitated glucose 20527 transporter), member 3 -1.81 1.12 -1.18 Protein kinase, camp dependent regulatory, 19087 type 11 alpha -l.82 1.14 -1.13 CLP 1 , cleavage and polyadenylation factor I 98985 subunit, homolog -1.82 1.21 -1.40 56550 Ubiquitin-conjugating enzyme E2D 2 -1.82 1.19 -1.08 50528 Transmembrane protease, serine 2 -1.82 1.08 -1.53 La ribonucleoprotein domain family, 67557 member 6 -1.82 -1.22 -1.08 107702 Ribonuclease/angiogenin inhibitor 1 -l.82 1.16 -l.l9 22034 Tnf recgptor-associated factor 6 -l .83 1.08 -1.13 27965 Spastic paraplpgia 21 homolog (human) -1.83 1.67 -1.27 21985 Tumor protein D52 -1.83 1.09 -1 . 19 75909 Transmembrane protein 49 -1 .84 1.41 -1.56 59069 Tropomyosin 3, gamma -1.84 1.42 -1.19 16885 LIM-domain containing, protein kinase -1.84 -1.22 -l .38 228790 Additional sex combs like 1 (Drosophila) -1.84 1.12 -1.28 20224 SARI gene homolog A (S. Cerevisiae) -1.84 1.11 -1.14 170736 Parvin, beta -1.84 1.20 -1.48 WD repeat and FYVE domain 69368 containipng 1 -1.85 -1.02 -1.31 Nuclear distribution gene E-like homolog 1 83431 (A. Nidulans) -1.85 1.40 -l .40 Protein phosphatase 1A, magnesium 19042 dependent, alpha isoform -1.85 1.25 -1.37 15006 Histocompatibility 2, Q region locus 1 -1.85 1.70 -1.29 16012 Insulin-like growth factor binding protein 6 -1.85 1.57 -1 . 14 Mitochondrial carrier homolog 1 (C. 56462 Eleggns) -1.85 1.37 -1.28 276 HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio 74596 CDP-diaLflglycerol synthase 1 -1.86 1.31 -1.38 Capping protein (actin filament) muscle Z- 12343 line, alpha 2 -1.86 1.26 -l.10 72508 Ribosomal protein S6 kinasemlypeptide l -1.86 1.39 -1.32 16190 Interleukin 4 receptor, alpha -1.86 1.18 -1.46 Ubiquitin-conjugating enzyme E2D 3 66105 (UBC4/5 homolog, yeast) -1.86 1.27 -1.26 3-hydroxy-3-methylglutaryl-Coenzyme A 15357 reductase -1.87 1.23 -l.l7 223433 Cdna sequence BC052328 -1.87 1.32 -1.38 11308 Abl-interactor 1 -1.87 1.29 -1.30 67951 Tubulin, beta 6 -1.87 -1.05 -1.25 109163 RIKEN cdna 3010003L21 gene -1.87 -1.14 -l.58 Fibronectin type 3 and ankyrin repeat 66930 domains 1 -1.87 1.07 -l.03 57439 Transmembrane protein 183A -1.87 1.07 -1.40 69718 Inositol polyphosphate multikinase -1.87 -1.02 -1.14 225280 RIKEN cdna D030070L09 gene -1.88 1.32 -1.37 22433 X-box bindirg protein 1 -1.88 1.19 -1.33 192897 Integrin beta 4 -1.88 -1.42 1.03 Clathrin, light polypeptide 74325 (ch) -1.88 1.14 -1.25 Nuclear undecaprenyl pyrophosphate 52014 synthase 1 homolog -1.88 1.07 -1.41 67946 Spermatogenesis associated 6 -1.89 1.51 -1.28 ADP-ribosylation factor-like 2 binding 107566 protein -1.89 1.23 -1.47 Eukaryotic translation initiation factor 2 15467 alpha kinase 1 -1.89 -1.06 -1.05 208659 Cdna sequence BC029169 -1.89 -1.09 -1.27 29809 RAB gtpase activating protein l-like -1.89 1.14 -1.48 Solute carrier family 30 (zinc transporter), 66500 member 7 -l .89 1.19 -l .24 18008 Nestin -1.90 -1.27 -1.56 MARVEL (membrane-associating) domain 218518 containingZ -1.90 -1.01 -1.35 Dnaj (Hsp40) homolog, subfamily A, 15502 member 1 -1.90 1.20 -1.24 215449 RAS related protein 1b -1.90 1.21 -1.17 Dnaj (Hsp40) homolog, subfamily A, 15502 member 1 -1.90 1.40 -1.22 Histocompatibility 2, K1, 14972 K region -1.92 1.35 -1.10 50523 Large tumor suppressor 2 -1.92 1.12 -1.21 277 HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio 193670 Ring finger protein 185 -1.92 1.38 -1.36 13136 CD55 antigen -1.92 -1.31 -1.22 Serine (or cysteine) peptidase inhibitor, 20708 clade B, member 6b -1.92 -1.06 -1.18 Fusion, derived from t(12; 16) malignant 233908 lipOsarcoma (humap) -1.92 1.71 -1.38 14155 Feminization 1 homolog b (C. Elegans) -1.93 1.39 1.12 Solute carrier family 9 (sodium/hydrogen 236794 exchanger), member 6 -1.93 1.08 -1.23 Frequently rearranged in advanced T-cell 14296 lymphomas -1.93 1.19 -1.42 12827 Collagen, type IV, alpha 2 -1.93 1.22 -1.28 24128 5'-3' exoribonuclease 2 -1.94 1.46 -1.22 66748 RIKEN cdna 4933404M02 gene -1.96 1.15 -1.01 26367 CBA-related cell adhesion molecule 2 -1.96 -1.44 -1.39 Eukaryotic translation initiation factor 3, 78655 subunit .1 -1.97 -1.03 -1.23 Potassium channel tetramerisation domain 239217 containing 12 -1.97 1.15 -1.16 74732 Syntaxin 11 -1.98 1.28 -1.06 13723 Embigin -1.98 -1.02 -1.83 11752 Annexin A8 -1.99 -1.10 -l.13 Serine/threonine kinase 17b (apoptosis- 98267 inducing) -1.99 1.34 -1.08 Pleckstrin homology, Sec7 and coiled-coil 19159 domains 3 -1.99 1.33 -1.30 Zinc finger and BTB domain 268294 containing 24 -1.99 1.50 -1.12 16891 Lipase, endothelial -2.00 1.08 -1.08 80907 Lactamase, beta -2.00 1.42 -1.23 71013 RIKEN cdna 4933400F03 gene -2.00 -1.01 -1.10 70231 Golgi reassembly stacking protein 2 -2.01 1.21 -1.40 243538 Coiled-coil domain containing 37 -2.01 1.04 -1.1 1 319622 RIKEN cdna E030018N11 gene -2.01 1.21 -1.16 381217 Gene model 967, (NCBI) -2.01 1.19 -1.49 Zinc finger, DHHC domain 69035 containing 3 -2.01 1.04 -1.31 Non-metastatic cells 5, (nucleoside- 75533 diphosphate kinase) -2.01 1.20 -1.00 22688 Zinc finger protein 26 -2.01 1.57 -1.17 V-rel reticuloendotheliosis viral oncogene 19697 homolog A (aviary -2.01 1.06 -1.44 330836 Solute carrier family 7 , member 6 -2.02 1.55 -1.40 69863 RIKEN cdna 1810054D07 gene -2.02 1.15 -1.20 278 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 237806 Dynein, axonemal, heavy chain 9 -2.02 -1.00 1.19 13386 Delta-like 1 homolog (Drosophila) -2.02 1.01 -1.62 Tumor necrosis factor, alpha-induced 21927 protein 1 (endothelial) -2.03 1.27 -l.35 21983 Trophoblast glycoprotein -2.03 1 .47 -1 .26 52502 Calcium regulated heat stable protein 1 -2.03 1.08 -1.22 20674 SRY-box containinggene 2 -2.03 -1.14 -1.11 320791 RIKEN cdna A130071D04jene -2.04 -1.27 1.16 Negative regulator of ubiquitin-like protein 53312 1 -2.05 1.57 -l.32 Sparc/osteonectin, cwcv and kazal-like 94214 domains proteoglycan 2 -2.05 1.24 -1.21 74122 Transmembrane protein 43 -2.05 -1.07 -1.42 94242 Tubulointerstitial nephritis antigen-like -2.06 1.15 -1 .3 1 Phorbol- 1 2-myristate- 1 3-acetate-induced 58801 protein 1 -2.06 1.33 1.08 18263 Omithine decarboxylase, structural 1 -2.08 1.81 -1.33 16418 Eukaryotic translation initiation factor 6 -2.08 1.16 -1.] 8 22041 Transferrin -2.08 -1.12 -1.88 70747 Tetraspanin 2 -2.09 -1.04 -1 .09 24109 Ubiguitin-like 3 -2.09 1.25 -1.36 76964 RIKEN cdna 2610028H24 gene -2.09 1.30 1.03 Protein phosphatase 2C, magnesium 381511 dependent, catalytic subunit -2.09 1.40 -1.05 320982 ADP-ribosylation factor-like 4C -2.10 1.38 -1.27 Interferon-related developmental 15982 regulator 1 -2.10 1.20 -1.14 195733 Grainyhead-like 1 (Drosophila) -2.10 1.15 1.06 24063 Sprouty homolog 1 (Drosophila) -2.11 1.04 -1.27 Ring finger and CHY zinc finger domain 68098 containing 1 -2.11 1.25 -1.17 Related RAS viral (r-ras) oncogene homolog 66922 2 -2.11 1.23 -1.57 225363 Eukaryotic translation termination factor 1 -2. 12 1.19 -1.05 Aldehyde dehydrogenase family 1, 11668 subfamily A1 -2. 12 1.10 1.01 l 191 1 Activating transcription factor 4 -2. 12 1.45 -1.28 19883 RAR-related orphan receptor alpha -2. 13 1.34 -1.29 12953 Cryptochrome 2 (photolyase-like) -2.13 1.39 -1.36 69131 Cch-related kinase, arginine/serine-rich -2. 13 1.07 -1.45 Transmembrane BAX inhibitor motif 69660 containing] -2.13 1.18 -1.27 68800 RIKEN cdna 1110059M19 gene -2. 13 -1.24 -1.72 279 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio Endothelial differentiation sphingolipid G- 13609 protein-coupled receptor 1 -2.13 1.25 —1.19 Protein kinase C and casein kinase substrate 23970 in neurons 2 -2.13 1.41 -1.65 68159 Syntaxin 19 -2.14 1.04 -l.10 Protein phosphatase 1, regulatory (inhibitor) 66849 subunit 2 -2. 14 1.01 -1.27 22135 Trans-go_lgi network protein 2 -2. 14 1.01 -1.56 381741 Leucine rich repeat containing 43 -2. 14 -1.07 1.00 Synaptotagmin binding, cytoplasmic RNA 56403 interacting protein -2.15 1.16 -1.21 14461 GATA binding protein 2 -2.15 1.08 -1 .27 68283 RIKEN cdna 9530077C05 gene -2.15 -1.30 -1.27 70019 RIKEN cdna 2410039M03 gene -2.16 -] .00 -1.03 Histocompatibility 2, 14972 K], K region -2. 17 1.22 -1.43 50708 Histone cluster 1, hlc -2.17 1.99 -1.02 435337 Predicted gene, EG435337 -2. 17 1.17 -1.23 110310 Keratin 7 -2.18 1.25 -1.30 269966 Nucleoporin 98 -2.18 1.21 -1.16 Radial spoke head 1 homolog 22092 (Chlamydomonas) -2. 18 1 .3 3 1.01 16782 Laminin, gamma 2 -2.18 1.33 -1.43 Engulfment and cell motility 2, ced-12 140579 homolog (C. Elegans) -2.19 1.25 -l.45 Ras association (ralgds/AF-6) domain 56289 family member 1 -2.20 1.12 -1.52 234797 RIKEN cdna 6430548M08 E116 -2.22 1.15 -1.06 219148 Cdna sequence BC065085 -2.22 1.59 -1.23 3-hydroxy-3-methylglutaryl-Coenzyme A 15357 reductase -2.22 1.14 -1 .24 Lymphatic vessel endothelial hyaluronan 114332 receptor 1 -2.22 1.11 -l .29 Notch-regulated ankyrin 67122 repeat protein -2.23 1.08 -1.33 75564 RIKEN cdna 1700027N10 gene -2.23 -1.06 1.26 72147 Zinc finger and BTB domain containing 46 -2.23 1.17 -1.19 234734 Alanyl-trna smthetase -2.23 1.28 -1.39 Rho GDP dissociation inhibitor (GDI) 14570 gamma -2.23 1.29 -1.07 75137 RIKEN cdna 4930535803 gene -2.23 1.94 -1.50 22436 Xanthine dehydrogenase -2.23 1.40 -1.58 330260 Paraoxonase 2 -2.24 1.19 -1.41 68498 Tetraspanin 11 -2.24 -1.02 -1.98 280 HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio 73075 Peptidylprolyl isomerase (cyclophilin)-like 6 -2.25 1.27 1.08 Heterogeneous nuclear ribonucleoprotein D- 50926 like -2.25 1.98 -1.15 58809 Ribonuclease, rnase A family 4 -2.25 -1.12 -1.50 12492 Scavenger receptor class B, member 2 -2.25 1.07 -1.28 64085 Calsyntenin 2 -2.26 -1.09 -1.16 Triggering receptor expressed on myeloid 71326 cells-like 1 -2.26 -1.20 -1.38 Enhancer of polycomb homolog 1 13831 (Drosophila) -2.26 1.05 -1.18 66540 RIKEN cdna 3110001Al3 gene -2.27 1.04 -1.39 75568 Calcyphosine-like -2.28 1.1 1 1.18 56360 Acyl-coa thioesterase 9 -2.29 1.12 -1.38 383295 Yippee-like 5 (Drosophila) -2.29 1.25 -1.24 74596 CDP-diacylglycerol synthase 1 -2.30 1.07 -1.39 69797 RIKEN cdna 1600029114 gene -2.30 1.07 1.06 14461 GATA binding protein 2 -2.31 1.23 -1.33 194655 Kruppel-like factor 11 -2.32 -1.15 -1.38 20568 Secretory leukocyte peptidase inhibitor -2.32 -1.19 -1.22 19153 Periaxin -2.34 1.18 -1.75 320769 Peroxiredoxin 6, related sequence 1 -2.34 1.08 -1.20 330260 Paraoxonase 2 -2.34 1.26 -1.43 MARVEL (membrane-associating) domain 218518 containing 2 -2.35 1.61 -1.41 71667 RIKEN cdna 0610007L01 gene -2.35 1.10 -] .29 66270 RIKEN cdna 1810015CMjene -2.36 1.72 -1.17 Metal response element binding 17764 transcription factor 1 -2.36 1.33 -1.36 66629 Golgiphosphoprotein 3 -2.37 1.16 -1.31 20104 Ribosomal protein S6 -2.37 1.09 -1.38 71823 RIKEN cdna 3300002A11 gene -2.38 -1.11 1.10 Colony stimulating factor 3 receptor 12986 ( anulocyte) -2.38 -1.08 -1.07 Inositol 1,4,5-trisphosphate 228550 3-kinase A -2.38 1.10 1.12 19243 Protein tyrosine phosphatase 4a] -2.40 1.27 -1.08 Solute carrier family 30 (zinc transporter), 22782 member 1 -2.40 1.23 -1.33 20388 Surfactant associated protein B -2.40 -1.07 -1.81 71648 Optineurin -2.40 1.50 -1.54 78781 Zinc finger CCCH type, antiviral 1 -2.42 1.48 -1.26 17713 Grpe-like 1, mitochondrial -2.42 1.59 -1.52 281 HIF1 HIF 2 HIF1/2 GeneID Gene name ratio ratio ratio 57349 Pro-platelet basic protein -2.44 -1.62 -1.57 104009 Quiescin Q6 sulflrydryl oxidase 1 -2.45 1.28 -1.23 215449 RAS related protein 1b -2.45 1.18 -1.14 317757 thase, IMAP family member 5 -2.45 1.33 -1.66 Protein phosphatase 1, regulatory (inhibitor) 66849 subunit 2 -2.46 1.45 -1.26 230678 Transmembrane protein 125 -2.46 -1.09 -1 . 15 74471 RIKEN cdna 4933440N224ene -2.47 1.15 -1 .60 18286 Outer dense fiber of sperm tails 2 -2.48 1.62 -1.23 11987 Solute carrier family 7 , member 1 -2.48 -] .05 -1.43 Transmembrane BAX inhibitor motif 69660 containing 1 -2.48 1.06 -1.21 170706 Transmembrane protein 37 -2.50 2.14 1.16 64898 Lipin 2 -2.50 1.29 -1.53 218215 Ring finger protein 144B -2.52 1.32 -1.16 72477 Transmembrane protein 873 -2.52 1.36 -1.16 11758 Peroxiredoxin 6 -2.53 1.14 -1.22 20515 Solute carrier family 20, member 1 -2.54 1.00 -1.27 18791 Plasminogen activator, tissue -2.56 1.29 -1.72 18145 Niemann Pick type C] -2.56 1.24 -1.52 Leucine-rich repeats and guanylate kinase 74354 domain containing -2.57 1.08 1.17 13649 Epidermal growth factor receptor -2.57 -1.16 -1.60 Calcitonin/calcitonin-related polypeptide, 12310 alpha -2.58 1.16 1.02 94242 Tubulointerstitial nephritis antigen-like -2.58 1.39 -1.27 75646 Retinoic acid induced 14 -2.58 1.02 -1.82 Par-6 (partitioning defective 6) homolog 58220 beta (C. Elggpns) -2.59 1.23 -1.32 Lymphatic vessel endothelial hyaluronan 114332 receptor 1 -2.63 1.68 -1.66 59043 WD repeat and SOCS box-containing 2 -2.63 1.26 -1.63 67876 Coenzyme Q10 homolfl B (S. Cerevisiae) -2.64 1.28 -1.28 320213 SUMO/sentrin 3&cific peptidase 5 -2.64 1.13 -l.56 1422] Four jointed box 1 (Drosophila) -2.64 1.21 -1.35 78801 Adenylate kinase 7 -2.66 -1.07 1.22 15982 Interferon-related developmental Eulator 1 -2.68 1.33 -1.11 14623 Gap junction protein, beta 6 -2.69 1.23 -1.61 17130 MAD homologpgosophila) -2.70 -1.01 -l .54 Enhancer of polycomb homolog 1 13831 (Drosophila) -2.70 1.16 -1 . 10 282 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio 73472 Spermatogenesis associated 18 -2.72 1.38 -1.24 64085 Calsyntenin 2 -2.73 1.18 -1.27 Sodium channel, nonvoltage-gated 1 20278 amma -2.74 1.32 -1.41 Tumor necrosis factor (ligand) superfamily, 50930 member 14 -2.75 1.01 -1.34 66070 CWC15 homolog (S. Cerevisiae) -2.76 -1.28 -1.03 17395 Matrix metallopeptidase 9 -2.77 -1.27 -1.36 236904 Kelch-like 15 (Drosophila) -2.80 1.21 -1.05 56542 Intestinal cell kinase -2.81 1 .28 -1 .20 21414 Transcription factor 7, T-cell specific -2.81 -1.25 -1.68 101351 RIKEN cdna A1300221M —2.82 -1.11 -1.24 64833 Acyl-coa thioesterase 10 -2.82 1.26 -1.50 17395 Matrix metallopeptidase 9 -2.84 -1.12 -1 .07 66166 $100 calcium binding protein A14 -2.86 -1.12 -1.71 67647 RIKEN cdna 4930523cm -2.87 1.19 -1.23 FK506 binding protein 12-rapamycin 56717 associated protein 1 -2.90 1.63 -1.56 11845 ADP-ribosylation factor 6 -2.92 1.44 -1.63 382137 RIKEN cdna D630004A14 gene -2.92 1.25 -1.10 11669 Aldehyde dehydrogenase 2, mitochondrial -2.93 1.43 -1.12 18260 Occludin -2.93 1.07 -1.55 Feline leukemia virus subgroup C cellular 226844 receptor 1 -2.93 -1 . 14 -1.76 Lysophosphatidylcholine 210992 acyltransferase 1 -2.93 -1-04 '2-52 66102 Chemokine (C-X-C motif) ligand 16 -2.93 1.15 -1.82 231991 Camp responsive element binding protein 5 -2.94 1.10 1.00 14102 Fas (TNF receptor superfamily member. 6) -2.95 1.04 -1.30 CDP-diacylglycerol synthase (phosphatidate 110911 cytidylyltransferase) 2 '2-97 1-25 '1-58 66270 RIKEN cdna 1810015C04 gene -2.98 1.56 .-1.15 67109 Zinc finger protein 787 -2.99 1.28 -1.21 104263 Jumonji domain containing 1A -3.01 1.56 -l .15 84004 Melanoma cell adhesion molecule -3.02 1.36 -] .93 Carbohydrate (N-acetylgalactosamine 4-0) 68947 sulfotransferase 8 -3.04 -1.15 -1.00 20863 Stefin A3 -3.10 -1.29 -1.40 Solute carrier family 7 20539 member 5 -3.13 1.41 -: .6: 55948 Stratifin -3.15 1.01 - . 7 66938 RIKEN cdna 1700029G01 gene -3.16 1.59 -1.6 283 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio Processing of precursor 4, ribonuclease 66161 P/MRP family, (S. Cerevisiae) -3.17 1.41 1.26 Guanine nucleotide binding protein, alpha 14674 13 -3.18 1.34 -1.42 69480 Tetratricopeptide repeat domain 9 -3.20 1.34 -1.87 78781 Zinc fingg CCCH type, antiviral l -3.20 1.23 -1.26 12274 Complement component 6 -3.24 -1.20 -3.52 29815 Breast cancer anti-estrogen resistance 3 -3.26 1.30 -1.13 213208 Interleukin 20 receptor beta -3.28 1.60 -1.39 8100 calcium binding protein A8 2020] (cglggnulin A) -3.28 -1.05 -1.45 319520 Dual specificity phosphatase 4 -3.30 -1.26 -1.53 56195 Polypyn'midine tract binding protein 2 -3.31 1.36 -1.43 66395 AHNAK nucleoprotein (desmoyokin) -3.31 1.70 -1.72 Solute carrier family 34 (sodium phosphate), 2053] member 2 -3.31 -1.01 -2.13 Anterior gradient homolog 3 (Xenopus 403205 laevis) -3.31 1.10 1.19 1E+08 Cdna sequence BC 1 179090 -3.32 -1.37 -1.37 74750 RIKEN cdna 5830410009 gene -3.33 -1.00 1.19 68603 Phosphomevalonate kinase -3.38 1.12 -1.99 C-type lectin domain family 4, 17474 member d -3.41 1.11 -1.41 A disintegrin and metallopeptidase 11501 domain 8 -3.42 -1.45 -1.01 109272 RIKEN cdna 8030451F13 gene -3.42 -1.14 -1.37 17394 Matrix metallopeptidase 8 -4.20 1.34 -1.39 228765 Sgtdecan binding protein (syntenin) 2 -4.26 1.09 -1.86 71908 Claudin 23 -4.29 -1.16 -1.74 Protein phosphatase 1, regulatory (inhibitor) 66849 subunit 2 -4.46 1.56 -1.35 ATP-binding cassette, sub-family A 76184 (ABC1), member 6 -4.46 1.98 -1.48 75137 RIKEN cdna 49305351303 gene 4.73 1.68 -1-58 66895 RIKEN cdna 1300014106 gene -5.03 -1-21 -1-94 77914 Keratin associated protein 17-1 -5.12 -1.20 -l.97 268885 Stefin A2 like 1 f -5.13 -1-11 '1-34 Hi h mobilit on box transcription actor 73389 1 g y gr p -5.30 1.83 -1.17 - ' t d lcium si nal transducer 56753 :umor assoma 6 ca g -5.62 1.10 -1.86 Myosin, heavy polypeptide 1, skeletal 17879 muscle, adult -5.66 -1.46 1.;‘57 14066 Coagplation factor II] -5.78 1.42 -l. 284 HIF1 HIF2 HIF1/2 GeneID Gene name ratio ratio ratio Cystatin 8 (cystatin-related epididyrnal 13012 spermatogenic) -6.32 -1.56 -5.78 12654 Chitinase 3-1ike 1 -6.44 -1.34 -1.94 72432 Serine peptidase inhibitor, Kazal type 5 -6.77 -2.09 -3.26 16178 Interleukin 1 receptor, type II -6.85 1.67 -1.14 245195 Resistin like gamma -9.15 1.06 -1.39 20249 Stearoyl-Coenzyme A desaturase 1 -16.7 -1.08 -3.58 285