DETERMINING THE ROLE OF IRF6 IN T CELL DEVELOPMENT AND FUNCTIONAL COMMITMENT By Tamer Ahmed Mansour A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of Genetics - Doctor of Philosophy Quantitative Biology - Dual Major 2014 ABSTRACT DETERMINING THE ROLE OF IRF6 IN T CELL DEVELOPMENT AND FUNCTIONAL COMMITMENT By Tamer Ahmed Mansour Interferon regulatory factor (IRF) is a protein family with nine members in mammals known to orchestrate the homeostatic mechanisms of host defense. There are functional and/or developmental defects of immune cells in the knockouts of eight family members. Like other family members, IRF6 is involved in regulating the cell cycle but in keratinocytes and mammary epithelial cell with mutations associated with squamous cell carcinomas. However, Irf6 is the only IRF known to be involved in morphogenesis. In humans, rare variants in IRF6 cause autosomal dominant orofacial clefting disorders while common variants contribute risk to non-syndromic forms. IRF6 is the only IRF family member with an as yet undetermined role in immunity. Here, we used publically available microarray data to uncover a dynamic expression pattern for Irf6 during hematopoietic development. We found that Irf6 is expressed early in hematopoiesis in long term hematopoietic stem cells. Also we identified Irf6 expression in T cell lineage, including developing and functionally committed stages. Irf1, 2, 4, 8 are indispensable for a normal T cell development and differentiation. Genetic variants in IRF5, IRF7 and IRF8 are associated to autoimmune disorders of T cells. Furthermore, protein complexes between IRF6/IRF5 and IRF6/IRF8 were described. These data together with DNA conservation among the IRF members and structural homology with IRF5 strongly suggests a role for Irf6 in the immune system, specifically in T-cell development and functional commitment. We utilized a mouse model to show that Irf6 was required for the regulation of thymocyte development. We found that Irf6 was expressed in the subcapsular region and medulla of the thymus. We further found that Irf6 regulated the distribution and proliferation of developing thymocytes. In addition, loss of Irf6 led to an increase in double negative cells with a concomitant increase in TCRγδ. Loss of Irf6 also led to a reduction in double positive cells with no corresponding reduction in single positive cell maturation. Also, we found that Irf6 dose is critical in development of both CD4+ and CD8+ cells in an age-dependent manner. These data suggest a novel gene function for Irf6 in thymocyte development and indicate further studies of IRF6 variants that might increase the risk of autoimmune disease. In the mouse, loss of Irf6 leads to perinatal lethality which hinders the ability to test the necessity of Irf6 in the functionally committed T helper (Th) subsets. In silico analysis suggested a model for Irf6 role in Th17/Treg balance. To test our hypothesis in vivo and overcome perinatal lethality, we employed an adaptive transfer of Irf6 knockout cells into lethally irradiated mice. Mice receiving Irf6 knockout cells had no deficit in restoration of lymphocyte production. In addition, we used two in vitro models to assess the necessity of Irf6 in the commitment of T helper cells. Using a stromal-free culture we found that naive T cells lacking Irf6 could be differentiated efficiently into Th1, Th2, Th17 and Treg using a specific cytokine cocktail. In vitro differentiation of dendritic cells showed significant increase of MHC-II expression after three days of culture. Irf6 might be involved in post-translational regulation of MHC-II. These data indicate that intrinsic Irf6 expression is not essential for T helper subset differentiation. However, a non-cell autonomous role for Irf6 in T cell differentiation through dendritic cells remains plausible. ACKNOWLEDGEMENTS To begin, I would like to extend my deepest gratitude to my mentor, Dr. Brian Schutte. His patience with me and confidence in my intellectual ability has been tantamount to my success. I thank him for teaching my not only how to be a scientist but also how to be a good, fair person. They have both been invaluable lessons. I would also like to thank the members of my committee; Titus Brown, Sungjin Kim, and Sainan Wei for the continued support and scientific contribution to this work. I would like to thank the current and past members of the Schutte Lab: Bill, Walid, Dan, Krysta, Raeuf, Larissa, Julie, Marissa, David, Nicole, Ari, Silas, Soumya, Erica, and Bryana. I could not have asked for a more outstanding group of people to share with both scientifically and socially. Lastly, I would like to thank Dr. Youssef Kousa and Dr. Arriana smith for helping me to grow scientifically, emotionally, and socially. Your guidance, support, and friendship have been invaluable. I must thank the 5th floor community, members of the Friderici, Fyfe, and Yuzbasiyan-Gurkan labs for their constant willingness to help and for making the 5th a wonderful work environment. I would also like to that the Genetics Program staff and my fellow colleagues. A special thank you goes to Jeannine Lee, Dr. Barbara Sears and Dr. Cathy Ernst for their support and willingness to help in any and all circumstances. I truly appreciate it. I would also like to thank the many people who graciously contributed to this work. To Jose Suarez-Martinez who pursed the in vitro differentiation studies of dendritic cells. To Dr. Norbert Kaminski and all his lab members for their help, support iv and scientific contribution. I must also thank members of the Histopathology laboratory, for sharing their expertise and warm attitudes, and members of ULAR, for there diligence and help with all things mouse related. Last, but certainly not least, I would thank my family and friends. I would to thank my parents for a lifetime of love, guidance and support. Without them, none of this would be possible. To my wife Dina and my kids Razan and Adam, thank you for being sources of inspiration and laughter. I owe a great deal of gratitude to all of my friends whom for fear of leaving anyone out, I will refrain from mentioning names. Undoubtedly, you have all been my sanity. I sincerely thank you all for your encouragement, insight, and friendship. This journey would not have been the same without you! v TABLE OF CONTENTS LIST OF TABLES ………..…………………………………………………………….. viii LIST OF FIGURES ...…………………………………………………………………... ix KEY TO SYMBOLS OR ABBREVIATIONS ...…………………………………... xii CHAPTER ONE .……………………………………………………………………….... Literature Review …...…………………………………………………………………… Significance …………………………………………………………………........ Thymus …………………………………………………………………………... T-cells development …………..……………………………………………....... Functional commitment of T helper subsets...………………………………... Interferon regulatory factor family...……………………………………………. Interferon regulatory factor 6…………………………………………………… Roles of IRFs in T-cell development and differentiation…………………….. REFERENCES ...……………………………………………………………….. 1 1 2 2 4 6 11 15 16 21 CHAPTER TWO .………………………………………………………………………… Meta-analysis of hematopoietic expression profiles reveals selective Irf6 expression in stem cells and in T-cells ……………………………………………...... Abstract …………………………………………………………………………... Introduction ………………………………………………………………............ Materials and Methods………………………………………………………….. Results ...………………………………………………………………………..... Discussion………………………………………………………………………… APPENDIX ...………………………………………………………...…………… REFERENCES ………………………………………………………………….. 41 41 42 43 48 50 59 62 78 CHAPTER THREE ……………………………………………………………………… The role of Irf6 in T-cell development in the thymus ..……………………………….. Abstract …………………………………………………………………………… Introduction .……………………………………………………………………… Materials and Methods………………………………………………………….. Mice: ……………………………………………………………………………… Morphological assessment of Irf6 knockout thymi: ………………………….. Western blot analysis: ………………………………………………………….. Immunohistochemistry: …………………………………………………………. Total cell count and Flow cytometric analysis: ………………………………. Results…………………………………………………………………………….. Thymic expression of Irf6: ……………………………………………………… Characterization of thymic changes in Irf6 knock-out embryos: …………… Effect of Irf6 heterozygosity in postnatal thymic proliferation: ……………… 89 89 90 91 95 95 95 95 96 97 100 100 102 107 vi Discussion………………………………………………………………………… REFERENCES ………………………………………………………….............. 110 113 CHAPTER FOUR ...……………………………………………………………………… The role of Irf6 in functional commitment of T-cell subsets………………………….. Abstract……………………………………………………………………………. Introduction……………………………………………………………………….. Materials and Methods…………………………………………........................ Bioinformatic analysis: ………………………………………………………….. Mice and adoptive transfer: …………………………………………………….. Th1, Th2, Th17 and Treg differentiation in vitro: ……………………………... In vitro generation of bone marrow–derived DC: …………………………….. Results…………………………………………………………………………….. Bioinformatic prediction of possible roles of Irf6 in T-helper commitment: ... Efficiency of adoptive transfer: …………………………………………………. Th1, Th2, Th17 and Treg differentiation in vitro: ……………………………... Changes of bone marrow DCs in Irf6 adoptively transferred mice: ………... Discussion………………………………………………………………………… REFERENCES ………………………………………………………………….. 118 118 119 120 122 122 122 123 124 127 127 127 128 131 137 141 CHAPTER FIVE ….…………………………………………………………………….. Summary and Future Directions………………………………………………………. Summary and Future Directions………………………………………………. REFERENCES …………………………………………………………………. 147 147 148 159 CHAPTER SIX ………………………………………………………………………….. Role of endogenous Avian Leukosis Virus in the pathophysiology of spontaneous ALV-like tumors in chickens………………………………………… Abstract…………………………………………………………………………… Introduction………………………………………………………………………. Materials and Methods………………………………………........................... Experimental treatment of chicken: …………………………………………… Next generation sequencing of RNA: …………………………………………. Bioinformatic analysis: Results……………………………………………………………………………. Tumor incidence in the studied groups: ………………………………………. The performance of different preprocessing approaches: ………………….. Expression profile of Tumor samples is closer to bursa tissue than mature splenic B cells: …………………………………………………………………… Differential expression analysis: ……………………………………………….. Pathway enrichment analysis: …………………………………………………. Discussion………………………………………………………………………… APPENDIX ...……………………………………………...……………………… REFERENCES ……………………………………………………..................... 165 vii 165 166 168 170 170 170 173 177 177 177 180 182 184 186 190 226 LIST OF TABLES Table 2.1: Effects of IRFs intrinsic expression in hematopoietic cells..….………… 45 Table 2.2: Mouse expression profiles for hematopoietic cell types………………… 50 Table 2.3: Average rank expression for microarray platforms per gene …………... 58 Table A1: Average rank of IRFs expression for each cell type per experiment ….. 74 Table 6.1: Yield and quality of NGS ...…………………………………………………. 172 Table 6.2: Comparing the performance of different preprocessing approaches …. 179 Table 6.3: Pathway enrichment analysis………………………………………………. 184 Table A2: Genes significantly up regulated in malignant samples over both bursa and spleen samples …………..……………………….….……….. 191 Table A3: Genes significantly down regulated in malignant samples over both bursa and spleen samples ………...……….................................……… 208 viii LIST OF FIGURES Figure 1.1: Histological compartments of a thymic lobule with diagramatic representation of T cell development …..…………………………………... 5 Figure 1.2: Differentiation of T-helper subsets …………………………....……………. 9 Figure 1.3: interferon regulatory factor (IRF) protein family in mammals…………….. 14 Figure 1.4: Roles of IRFs in T-cell development and differentiation …………………. 19 Figure 2.1: Clustering analysis of average Irf gene rank expression ………………… 54 Figure 2.2: Average Irf6 rank expression in early hematopoietic progenitor ...……… 56 Figure 2.3: Average Irf6 rank expression in T cell stages ………………………...…… 57 Figure A1: Expression of Irf genes in immature hematopoietic progenitors at 3 months and 2 years of mouse age ………………..…...……………….… 63 Figure A2a: Expression of Irf genes in immature haematopoietic progenitors ……… 64 Figure A2b: Expression of Irf genes in immature haematopoietic progenitors ……… 64 Figure A2c: Expression of Irf genes in immature haematopoietic progenitors .……... 65 Figure A2d: Expression of Irf genes in immature haematopoietic progenitors …….... 65 Figure A3: Expression of Irf genes in early myeloid development .............................. 66 Figure A4: Expression of Irf genes during granulocytes development ……….....…… 66 Figure A5a: Expression of Irf genes in thymocyte development ………………...……. 67 Figure A5b: Expression of Irf genes in thymocyte development…………………….… 67 Figure A6: Expression of Irf genes in LT-HSC versus terminally differentiated haematopoietic cells ...……………………………………………….………. 68 Figure A7a: Expression of Irf genes in lymphocytes and DCs ………………………... 69 Figure A7b: Expression of Irf genes in lymphocytes and DCs ………………………... 69 Figure A8: Expression of Irf genes in resting and activated lymphocytes …...………. 70 Figure A9: Expression of Irf genes in mast cells and pre-mast cells ………..……….. ix 70 Figure A10a: Expression of Irf genes in T-helper subsets……………………………... 71 Figure A10b: Expression of Irf genes in T-helper subsets……………………………... 71 Figure A10c: Expression of Irf genes in T-helper subsets……………………………... 72 Figure A11: Expression of Irf genes in BM macrophage and whole thymus suspension ……………………………………………………………………. 73 Figure A12: Expression of Irf genes in non-lymphocyte thymic cells…………………. 73 Figure 3.1: Stages of T cell development in the thymus……………………………….. 94 Figure 3.2: Flowcytometric analysis of the thymocyte subpopulations……………….. 98 Figure 3.3: Two-color western blot for Irf6 and Gapdh…………………………………. 100 Figure 3.4: Irf6 expression in the thymus………………………………………………… 101 Figure 3.5: Thymic-cardiac ratio………………………………………………………….. 103 Figure 3.6: BrdU incorporation in developing thymocytes……………………………… 104 Figure 3.7: Irf6 regulates proliferation and cell count of thymocytes…………………. 105 Figure 3.8: Loss of Irf6 leads to an abnormal distribution of proliferating cells………. 106 Figure 3.9: Frequency of TCRγδ…………………………………………………………. 106 Figure 3.10: Loss of Irf6 does not alter total cell death…………………………………. 107 Figure 3.11: Effect of Irf6 dosage on BrdU incorporation in thymocyte populations… 108 Figure 4.1: Gating scheme for identification of DC populations in BM……………….. 126 Figure 4.2: Efficiency of adoptive transfer………………………………………………. 128 Figure 4.3: Th1, Th2, Th17 and Treg differentiation in vitro……………………………. 129 Figure 4.4: Frequency of total DCs and their sub-populations in bone marrow……… 131 Figure 4.5: In vitro differentiation model of DCs………………………………………… 133 Figure 4.6: Expression of maturation markers on DCs in culture……………………… 136 Figure 4.7: Proposed model of Irf6 in Th17/Treg balance……………………………… 137 Figure 6.1: Snap shot from the FASTX toolkit analysis………………………………… 173 Figure 6.2: Snap shot of a genome browser (IGV) comparing the annotations x of different assemblies……………………………………………………...... 174 Figure 6.3: Diagrammatic representation of the TopHat/Cufflinks analysis…………. 175 Figure 6.4: Work flow for the pipeline of NGS analysis………………………………… 176 Figure 6.5a: Boxplot of expression profiles …..…………………………………………. 181 Figure 6.5b: Ward Hierarchical Clustering using Euclidean distance…………………. 181 Figure 6.6: Differential expression Analysis……………………………………………… 183 Figure 6.7: TUSC2 Molecular pathway…………………………………………………… 188 xi KEY TO SYMBOLS OR ABBREVIATIONS IRF - Interferon regulatory factor Th – T helper TEC – thymic epithelial cells CK – cytokeratin DN - double negative TCR - T cell receptor DP - double positive TGFβ - Transforming Growth Factor β SP - single positive Treg - regulatory T-cells T-bet - T-box expressed in T-cells APC - Antigen Presenting Cell GATA3 - GATA-binding protein 3 c-MAF - transcription factor Maf RORγt - retinoic acid receptor–related orphan receptor γt Klf4 - Kruppel-like factor 4 SLE - systemic lupus erythematosus FOXP3 - Forkhead box P3 DBD - DNA-binding domain IAD – Interferon Association Domain ChIP - Chromatin immunoprecipitation xii HSC - Haematopoietic stem cells RMA - Robust Multi-array Average algorithm LT-HSC - long term hematopoietic stem cells ST-HSC - short term hematopoietic stem cells LMPP - lymphoid multipotent progenitors CMP - common myeloid progenitors CLP - common lymphoid progenitors MEP - megakaryocyte-erythroid progenitor GMP - granulocyte monocyte progenitors BLP - B cell-biased lymphoid progenitor NK - nature killer cells iTreg - induced regulatory T cells nTreg - natural regulatory T cell DC - dendritic cells pDC - plasmacytoid dendritic cells (pDC) tDC - thymic dendritic cells BM.MPh - bone marrow macrophages tMPh - thymic macrophages Nu RBCs - nucleated erythrocytes cTEC - cortical thymic epithelial cells mTEC - modularly thymic epithelial cells BrdU - Bromodeoxyuridine GM-CSF - granulocyte macrophage-colony stimulating factors xiii BM – Bone marrow LC - Langerhans cells LL - Lymphoid leukosis ALV - Avian Leukosis Virus TUSC2 - Tumor suppressor candidate 2 EIF4E - Eukaryotic translation initiation factor 4E MDV2 - Marek's disease virus serotype 2 xiv CHAPTER ONE Literature Review 1 Significance T-cells are the most abundant subset of blood lymphocytes and serve as the core of the adaptive immune response. Changes in T-cell number or function can lead to autoimmune diseases, immune deficiency, inflammatory disorders and cancer. Evolution of lymphocytes with a cadre of highly diverse antigen-recognition receptors is necessary for immune surveillance but also requires stringent screening for autoreactive clones. T-cells develop in the thymus, where a highly specialized microenvironment educates the evolving T-cells (Janeway et al., 2001; Paul, 2008 ). The Interferon regulatory factor (IRF) family of transcription factors are indispensable for functional and developmental regulation of immune cells. While Irf6 shares DNA conservation and predicted structural homology with the IRF family members, its role in immunity is unknown. However, we already know that IRF6 is involved in protein-protein interactions with IRF5 and IRF8 (Li et al., 2011), both of which regulate T-cell development and T helper differentiation. Preliminary bioinformatic analysis shows expression of Irf6 during thymocyte development and functional commitment of T helper subsets. These preliminary findings support the investigation of Irf6 in T-cell development. Furthermore, it provides a new diagnostic and therapeutic target in autoimmune disorders such as psoriasis and systemic lupus erythematosus. Thymus Evolutionary studies show that thymus development began with jawed vertebrates (Boehm & Bleul, 2007; Litman & Cooper, 2007). Mammals in general, and 2 humans in particular, have a single thymus located superior to the heart at the thoracic inlet and is composed of bilateral lobes (Dooley et al., 2006; Rodewald, 2008; Terszowski et al., 2006). Histologically, the thymus is composed of an inner medulla and a peripheral cortex surrounded by an outer capsule. Thymus tissue is composed of lymphoid cells (CD45+CD7+) and stromal cells with a ratio of 50 lymphoid cells for each stromal cell (Rodewald, 2008; Singer et al., 1986). Non-hematopoietic stromal cells can be further classified into thymic epithelial cells (TEC, Keratin+) and mesenchymal cells (Keratin−) (Anderson et al., 1993). Dendritic cells and macrophages are CD45+ thymic stromal cells, thus they constitute the hematopoietic component of the stromal mesh (Rodewald, 2008) (Figure1.1). In the mouse, thymus organogenesis starts around 10.5 days after fertilization (E10.5) when endodermal epithelial cells of the third pharyngeal pouch initiate the thymic primordium (Gordon et al., 2004; Hollander et al., 2006). Neural crest cells migrate into the thymic capsule, interlobular septae, and stromal cell network to regulate the early proliferation and differentiation of immature TECs (Ambrosiani et al., 1996; Itoi et al., 2007; Jenkinson et al., 2003; Jenkinson et al., 2007; Johnston, 1966; Yamazaki et al., 2005). Migration of lymphoid precursors (next section) to the thymus starts by E11.5 (Haynes & Heinly, 1995; Liu et al., 2006; Owen & Ritter, 1969). The interaction between lymphoid cells and TECs is critical for normal development of both the lymphoid and epithelial cell compartments (Anderson & Jenkinson, 2001). By E13.5, two thymic epithelial populations can be appreciated by cytokeratin (CK) markers; cortical epithelium (CK8+CK5−) and medullary epithelium (CK8−CK5+) (Klug et al., 2002). 3 However, additional expansion of medullary islands is observed as late as E18.5 and corresponds with the emergence of mature T-cells (Irla et al., 2008). T-cells development T-cell precursors migrate from either the fetal liver or bone marrow to seed the thymus. T-cell precursors are initially called double negative (DN) thymocytes because they lack expression of both CD4 and CD8 (Godfrey et al., 1993; Pearse et al., 1989). DN thymocytes undergo T cell receptor (TCR) rearrangement under guidance of cortical TECs (Raulet et al., 1985; Shinkai et al., 1992; Takahama, 2006; Tourigny et al., 1997; von Boehmer & Fehling, 1997; Xu et al., 1996). Cells with functional TCRs start to express both CD4 and CD8 and are in turn called double positive (DP) thymocytes (Irving et al., 1998; Michie & Zuniga-Pflucker, 2002). In the subcapsular region, transforming growth factor β (TGFβ) signaling suppresses the proliferation of pre-DP thymocytes to regulate the production of DP cells (Benz et al., 2004). DP cells migrate back through the cortex where positive and negative selections occur. In total, only 35% of cells survive and reach the thymic medulla. DP cells lose either CD4 or CD8 to reach the single positive (SP) stage. SP thymocytes, either CD4+ (T helper) cells or CD8+ (T cytotoxic) cells, continue their maturation and central tolerance in the medulla before being shuttled out of the thymus (Blackburn & Manley, 2004; Germain, 2002; Hoffmann et al., 2003; Lind et al., 2001; Plotkin et al., 2003; Prockop & Petrie, 2000) (Figure1.1). 4 Figure 1.1: Histological compartments of a thymic lobule with diagramatic representation of T cell development. Haematopoietic precursors seed the thymus at medullary cortical junction. Recent thymic immigrants lack both CD4 and CD8 and thus called double negative (DN) thymocytes. CD44 and CD25 are two surface markers which mark 4 major developmental sub-populations of DN thymocytes (DN1, CD44+CD25-; DN2, CD44+CD25+; DN3, CD44-CD25+; and DN4, CD44-CD25-). Transition from DN4 (also called pre-DP) to DP cells occurs in the subcapsular region. DP cells migrate back through the cortex and reach the thymic medulla. Meanwhile, DP cells lose either CD4 or CD8 to reach the single positive (SP) stage. (Figure is modified from Blackburn and Manley, 2004) 5 Functional commitment of T helper subsets T-cells execute their designated functions by either secreting soluble cytokines or through direct cell-cell interaction. T helper (Th) lymphocytes are widely understood to function as the conductors of the adaptive immune orchestra. Upon antigen exposure, T helper cells differentiate into specialized subsets. Each T helper subset differentiates under a unique signaling pathway and lineage-specific transcription factors to produce a characteristic cytokine milieu (Fietta & Delsante, 2009; Hirahara et al., 2011). T helper subsets include Th1, Th2, Th17, regulatory T-cells (Treg), T follicular helper cells, Th9 and Th22 cells (Bluestone et al., 2009; Shevach, 2010). The balance between different T helper cells is most typically defined by mutually exclusive expression of lineagespecific transcription factors. Relative to all T helper subsets, Th1 and Th2 development and function has been most clearly elucidated. Differentiation of Th1 initially requires expression of a transcription factor called T-box expressed in T-cells (T-bet). Subsequent exposure to IL12 and IL18 among other cytokines produced by Antigen Presenting Cells (APCs) induces completion of the differentiation process. Th1 cells regulate cellular immunity and are essential for the eradication of intracellular pathogens (Matsuoka et al., 2004; Rautajoki et al., 2008). Alternatively, Th2 differentiation necessitates IL4 mediated signaling and expression of the lineage specific transcription factors GATA-binding protein 3 (GATA3) and transcription factor Maf (c-MAF). Th2 cells are responsible for regulating humoral immunity and are implicated in the pathophysiology of allergy (O'Garra & Arai, 2000; Rautajoki et al., 2008). 6 Th17, a T helper subset producing IL17, has pro-inflammatory effects and protects against bacterial infections in the intestine and the airways (Miossec et al., 2009; Mitsdoerffer et al., 2010). TGFβ in the presence of IL6 can initiate Th17 commitment (Bettelli et al., 2006; Dong, 2006; McGeachy et al., 2009). The Th17 lineage specific transcription factor is retinoic acid receptor–related orphan receptor γt (RORγt) (Ivanov et al., 2006). Kruppel-like factor 4 (Klf4) is another important transcription factor required for full commitment of Th17. T-cell-specific Klf4-knockout mice show 24% reduction of IL-17+ CD4+ T-cells (Botti et al., 2011). Defects in Th17 development can lead to several autoimmune diseases, including rheumatoid arthritis, asthma and systemic lupus erythematosus (SLE) (Maddur et al., 2013; Oukka, 2008; Tesmer et al., 2008). Additional roles for Th17 in graft rejection and inflammatory bowel disease have been described (Agorogiannis et al., 2012; Dong, 2008; Kolls & Linden, 2004). Treg is a suppressor T helper cell subset that controls the amplitude of the immune response and prevents the development of autoimmune diseases. Impairment of the reciprocal differentiation between Th17 and Treg has been implicated in several autoimmune disorders such as experimental autoimmune encephalomyelitis; a mouse model of multiple sclerosis, and type I diabetes mellitus (Pan et al., 2011). Moreover, Th17/Treg imbalance is associated with tumors (Hu et al., 2011) and graft-versus host rejection(Dander et al., 2009). While there are different subpopulations of Tregs, Forkhead box P3 (FOXP3) is a common marker in this lineage (Green et al., 1983). Two subsets of Tregs are determined by their developmental origin, whereas nTregs arise naturally in the thymus, iTregs are induced peripherally. Treg subtypes might have 7 similar or overlapping functions but are not identical. For example, Foxp3 deficiency causes fatal systemic autoimmune disease due to preferential Th1 and Th17 induction (Bennett et al., 2001; Brunkow et al., 2001). However selective deficiency of iTreg is associated with Th2 allergic response at mucosal sites when systemic autoimmunity of Th1 or Th17 has not been implicated (Josefowicz et al., 2012). Importantly, prior work showed that TGFβ signaling inhibits both Th1 and Th2 differentiation. More recent work shows that TGFβ signaling is essential for induction of Foxp3 expression and commitment of Treg, either nTreg or iTreg (Chen & Wahl, 2002). The ability of TGFβ signaling to induce FOXP3 while concomitantly suppressing the Th17 cell lineage is mediated by the protein Inhibitor of DNA binding 3 (ID3) (Chen & Wahl, 2002; Maruyama et al., 2011; Pan et al., 2011) (Figure 1.2). 8 Figure 1.2: Differentiation of T-helper subsets. Naïve T cells undergo functional differentiation under cytokine direction form antigen presenting cells (APC). 9 Figure 1.2 (cont’d): Th1 requires expression of the transcription factor T-box (Tbet). IL12 and IL18 induce STAT4 signaling to induce IFN-γ production to complete the differentiation and expression of IL2, IFN-γ and TNF. Th1 cells invoke cell mediated immunity and induce destruction of intracellular pathogens. IL4 and IL6 induce STAT6 signaling and expression of GATA-binding protein3 (GATA3) and transcription factor Maf (c-MAF) to allow Th2 differentiation. Th2 cells secrete IL4, IL5, IL6, IL10 and IL13 to initiate humoral immunity. Th2 response is required for control of helminthes infection. TGFβ and IL6 initiate STAT3 signaling and expression of retinoic acid receptor–related orphan receptor γt (RORγt) and Kruppel-like factor 4 (Klf4) to induce Th17 commitment. Th17 cells produce IL21, IL17, IL22 and TNF. Th17 cytokines have proinflammatory effect and protect against bacterial infections. TGFβ can induce STAT5 signaling and mediate expression of FOXP3 causing Treg commitment. Tregs produce more TGFβ and IL10 to suppress the response. Figure is modified from Fietta and Delsante, 2009. 10 Interferon regulatory factor family The interferon regulatory factor (IRF) family of transcription factors has nine members in mammals (Huang et al., 2010; Nehyba et al., 2009). All family members share a highly conserved N-terminal DNA-binding domain (DBD) that possesses five conserved tryptophan residues, each separated by 10–18 amino acids (Kondo et al., 2002; Lohoff & Mak, 2005; Tamura et al., 2008). There is also a shared, but less conserved protein-binding domain at the C-terminus (Kondo et al., 2002; Lohoff & Mak, 2005) (Figure 1.3). The IRF family is known to orchestrate homeostasis of host defense (Tamura et al., 2008; Taniguchi et al., 2001). The functions of different IRFs can be categorized into three main targets. First category is transcriptional regulation of type I interferon responses which is an indispensible downstream target of IRFs (Barnes et al., 2001; Fujita et al., 1989; Honda et al., 2005a; Honda et al., 2005b; Honda et al., 2006; Honda & Taniguchi, 2006; Hoshino et al., 2006; Izaguirre et al., 2003; Marie et al., 1998; Matsui et al., 2006; Matsuyama et al., 1993; Moynagh, 2005; Negishi et al., 2005; Sato et al., 1998; Sato et al., 2000; Tailor et al., 2007; Takaoka et al., 2005; Taniguchi et al., 2001; Taniguchi & Takaoka, 2002; Tsujimura et al., 2004; Yoneyama et al., 1998; Zhao et al., 2006). Second broad category of IRF family functions is their necessary roles in development and function of a cadre of immune cell types, innate immune cells such as phagocytes (Hida et al., 2005; Holtschke et al., 1996; Kamijo et al., 1994; Salkowski et al., 1999; Scheller et al., 1999; Tamura et al., 2000; Tamura et al., 2005b; Testa et al., 2004; Tsujimura et al., 2002) and natural killers (Duncan et al., 1996; Lohoff et al., 2000; Ogasawara et al., 1998; Taki et al., 2005) or adaptive immune cells for example dendritic cells (Gabriele et al., 2006; Honda et al., 2004; Ichikawa et al., 2004; Schiavoni 11 et al., 2002; Schiavoni et al., 2004; Suzuki et al., 2004; Tamura et al., 2005a; Tsujimura et al., 2003) and lymphocytes (Brien et al., 2011; Brustle et al., 2007; Fragale et al., 2008; Klein et al., 2006; Lee et al., 2006; Lohoff et al., 1997; Lohoff et al., 2002; Lu et al., 2003; Ma et al., 2011; Ma et al., 2006; Mittrucker et al., 1997; Ouyang et al., 2011; Penninger et al., 1997; Penninger & Mak, 1998; Scharton-Kersten et al., 1997; Sciammas et al., 2006; Taki et al., 1997; Tian et al., 2012; Tominaga et al., 2003; White et al., 1996; Zhang et al., 2011; Zheng et al., 2009). Lastly, IRF family members are also involved in the regulation of cell cycle control and oncogenic pathogenesis. IRF1 (Bouker et al., 2005; Connett et al., 2005; Giatromanolaki et al., 2004; Harada et al., 1993; Kano et al., 1999; Passioura et al., 2005a; Romeo et al., 2002; Tanaka et al., 1994; Tanaka et al., 1996; Yim et al., 2003), IRF3 (Duguay et al., 2002; Heylbroeck et al., 2000; Kim et al., 1999; Kim et al., 2003), IRF5 (Barnes et al., 2003; Hu et al., 2005; Mori et al., 2002; Yanai et al., 2007) and IRF8 (Deng & Daley, 2001; Hao & Ren, 2000; Liu & Abrams, 2003; Yang et al., 2007a; Yang et al., 2007b) are negative regulators of cell proliferation with known pro-apoptotic and tumor suppressor activities. On the other hand, IRF2 antagonizes the tumor suppressor effect of IRF1 (Connett et al., 2005; Passioura et al., 2005a; Passioura et al., 2005b; Yim et al., 2003). The role of IRF4 in oncogenesis is context dependent. For example, IRF4 is oncogenic in late developmental stages of lymphoid lineage (Hrdlickova et al., 2001; Iida et al., 1997; Ito et al., 2002; Shaffer et al., 2008; Tsuboi et al., 2000) and a tumor suppressor in myeloid leukemia (Jo & Ren, 2011; Ortmann et al., 2005; Schmidt et al., 2000) and B cell malignancies of early developmental stages (Acquaviva et al., 2008; Pathak et al., 12 2011). Combined deficiency of IRF4 and IRF8 produces both myeloid and lymphoid tumors (Jo et al., 2010). 13 Figure 1.3: interferon regulatory factor (IRF) protein family in mammals (Modified from Lohoff & Mak. Nat.Immunol. 2005). IRF family has nine members in mammals. IRFs are characterized by a highly conserved DNA binding domain (DBD) and a much less conserved protein association domain (regulatory domain; IAD – Interferon Association Domain). 14 Interferon regulatory factor 6 IRF6 has the canonical family DBD and its protein-binding domain is most closely related to IRF5 (Huang et al., 2010; Nehyba et al., 2009). Like other family members, IRF6 is involved in regulating the cell cycle with an anti-proliferative function in keratinocytes and mammary epithelial cell (Bailey et al., 2008; Ingraham et al., 2006; Richardson et al., 2006). Consistently, mutations in IRF6 have also been found in patients with squamous cell carcinomas, re-emphasizing tumor suppressor function (Bailey et al., 2009; Botti et al., 2011; Stransky et al., 2011). However, Irf6 is the only IRF known to be involved morphogenesis (Ingraham et al., 2006; Richardson et al., 2006; Richardson et al., 2009; Thomason et al., 2010). In humans, haploinsufficiency of IRF6 causes syndromic orofacial clefting (Kondo et al., 2002). Furthermore, a common DNA variant at the IRF6 locus accounts for 12% of isolated orofacial clefting risk worldwide (Rahimov et al., 2008; Zucchero et al., 2004). Despite sequence conservation and structural similarity, Irf6 is the only IRF family member with an as yet undetermined role in immunity. In 2005, Lohoff and Mak wondered if IRF6 is even expressed by haematopoietic cells (Lohoff & Mak, 2005). Furthermore, functional and genetic studies of Irf6 in the immune system are hindered because the knockout mouse model is perinatal lethal (Ingraham et al., 2006). Thus refined technical strategies are required to test the role of IRF6 in the immune system. IRF6 is a putative transcription factor. The DBD of IRF6 binds a sequence highly analogous to the IRF family consensus (Botti et al., 2011; Little et al., 2009). We know that the DBD is critical for IRF6 function because mutations in this region can lead to more severe forms of orofacial clefting, e.g. popliteal pterygium syndrome (Kondo et al., 15 2002). Evidence for the transcriptional activity of IRF6 is also shown in Sabel et al. 2009. Expression of the IRF DBD in xenopus embryos results in failure of gastrulation. Co-expression of IRF6 rescues the phenotype suggesting specific competition at the DNA (Sabel et al., 2009). Chromatin immunoprecipitation (ChIP) sequencing for IRF6 binding sites confirmed the direct DNA binding of Irf6 to many genes (Botti et al., 2011). As a result of these studies, we know that IRF6 binds to and regulates an important transcriptional network Roles of IRFs in T-cell development and differentiation IRF family members regulate T-cell biology either by intrinsic transcriptional activities in T-cells or through extrinsic roles in non-T-cells like thymic stromal cells and other immune cells. While IRFs have an important role in immunity, their role in T helper cell development and differentiation is of particular interest for both biological and clinical applications (Figure 1.4). Although several Irfs are expressed in thymocytes (Colantonio et al., 2011; Hrdlickova et al., 2001; Nordang et al., 2011; Simon et al., 1997), Irf1 is the only family member with functional studies supporting a role in T-cell development (Lee et al., 1999; White et al., 1996). Non-cell autonomous Irf1 reduces expression of the major histocompatibility complex related genes in the thymic microenvironment (Lee et al., 1999; White et al., 1996). However it is the Irf1 intrinsic activity in T-cells that is required for development and thymic selection of naïve CD8 T-cells (Matsuyama et al., 1993; Penninger et al., 1997; Penninger & Mak, 1998). In Irf4 knockout mice, while cell count changes during thymocyte development appear to be unaffected, the proliferative 16 capacity, antiviral cytotoxicity, allogenic graft rejection, tumor surveillance and cytokine production are markedly impaired (Mittrucker et al., 1997). Likewise, Irf1, Irf2 and Irf8 are critical for mounting a Th1 response (Lohoff et al., 1997; Lohoff et al., 2000; Scharton-Kersten et al., 1997; Taki et al., 1997) mainly through transcriptional activation of Il12 (Coccia et al., 1999; Galon et al., 1999; Giese et al., 1997; Liu et al., 2003; Maruyama et al., 2003; Salkowski et al., 1999); a macrophage derived cytokine mandatory for Th1 differentiation (Murphy & Reiner, 2002). Th1 is further supported by IRF-mediated suppression of IL4 production in Th2 and basophils (Elser et al., 2002; Lohoff et al., 1997; Taki et al., 1997) and activation of APCs (Fantuzzi et al., 2001; Lohoff et al., 2000; Niedbala et al., 2002; Ogasawara et al., 1998; Oppmann et al., 2000). Similarly, Irf4 is indispensable for a Th2 response (Lohoff et al., 2002; Tominaga et al., 2003). Irf4 induces intrinsic transcription of Il4 (Hu et al., 2002; Rengarajan et al., 2002) and enhances key transcriptional regulators of Th2 (e.g. GATA3 (Lohoff et al., 2002) and GFI1 (Tominaga et al., 2003)). Irf4 knockout mice also have abnormal dendritic cell development, potentiating the T-cell defect (Suzuki et al., 2004; Tamura et al., 2005a). The Irf family also has a prominent role in Th17 commitment. For example, Irf4 is essential for Th17 differentiation (Brustle et al., 2007; Zhang et al., 2011) by direct transcriptional induction of Il17A and Il21 (Chen et al., 2008; Fanzo et al., 2006). In contrast, Irf8 is capable of suppressing Th17 differentiation by direct repression of RORγt; the lineage-specific transcription factor of Th17 (Ouyang et al., 2011; Tian et al., 2012). Similar patterns of balanced regulation can be seen in Treg cells. Irf1 suppresses the production of both nTreg in the thymus and iTreg in the periphery by direct 17 transcriptional repression of the Foxp3 promoter through a highly conserved Irf binding site (Fragale et al., 2008; Ma et al., 2011). On the contrary, Irf4 is a direct downstream target of Foxp3 and mediates the immune suppressive effect of Tregs (Zheng et al., 2009). TGF- β1 is known to control the differentiation of both Th17 and Treg (Bettelli et al., 2006; Chen & Wahl, 2002; McGeachy et al., 2009). Previous studies in the palate show Irf6 as a downstream target to Tgf-β signaling (Le et al., 2012; Xu et al., 2006). However, this interaction has not been tested in T helper subsets. 18 Figure 1.4: Roles of IRFs in T-cell development and differentiation. Irfs are required for intra thymic development and functional commitment in peripheral lymphoid organs. 19 Finally, autoimmune disease association studies have further implicated the IRF family in orchestrating T-cell development and function. Genetic variants in IRF5, IRF7 and IRF8 are associated to psoriasis, multiple sclerosis and SLE susceptibility (De Jager et al., 2009; Demirci et al., 2007; Gateva et al., 2009; Graham et al., 2006; Harley et al., 2008; Leppa et al., 2011; Patel, 2011; Sanchez et al., 2008). Shitao Li at al 2011 performed a proteomic study to define the candidate protein complexes involved in regulating interferon type I. Interestingly, affinity purification identified protein complex formation between IRF6/IRF5 and IRF6/IRF8, the latter was further confirmed by coimmunoprecipitation in HEK293 cells (Li et al., 2011). Protein-protein interactions between IRF6 and more canonically described immune IRF family members, DNA conservation of the DBD and structural homology with IRF5 strongly suggests a role for Irf6 in the immune system, specifically in T-cell development and functional commitment. 20 REFERENCES 21 REFERENCES Acquaviva, J., Chen, X. & Ren, R. (2008). IRF-4 functions as a tumor suppressor in early B-cell development. Blood 112, 3798-3806. Agorogiannis, E. I., Regateiro, F. S., Howie, D., Waldmann, H. & Cobbold, S. P. (2012). Th17 cells induce a distinct graft rejection response that does not require IL17A. Am J Transplant 12, 835-845. Ambrosiani, J., Armengol, J. A., Martinez, S. & Puelles, L. (1996). The avian inferior olive derives from the alar neuroepithelium of the rhombomeres 7 and 8: an analysis by using chick-quail chimeric embryos. Neuroreport 7, 1285-1288. Anderson, G., Jenkinson, E. J., Moore, N. C. & Owen, J. J. (1993). MHC class IIpositive epithelium and mesenchyme cells are both required for T-cell development in the thymus. Nature 362, 70-73. Anderson, G. & Jenkinson, E. J. (2001). Lymphostromal interactions in thymic development and function. Nat Rev Immunol 1, 31-40. Bailey, C. M., Abbott, D. E., Margaryan, N. V., Khalkhali-Ellis, Z. & Hendrix, M. J. (2008). Interferon regulatory factor 6 promotes cell cycle arrest and is regulated by the proteasome in a cell cycle-dependent manner. Mol Cell Biol 28, 2235-2243. Bailey, C. M., Margaryan, N. V., Abbott, D. E., Schutte, B. C., Yang, B., KhalkhaliEllis, Z. & Hendrix, M. J. (2009). Temporal and spatial expression patterns for the tumor suppressor Maspin and its binding partner interferon regulatory factor 6 during breast development. Dev Growth Differ 51, 473-481. Barnes, B. J., Moore, P. A. & Pitha, P. M. (2001). Virus-specific activation of a novel interferon regulatory factor, IRF-5, results in the induction of distinct interferon alpha genes. J Biol Chem 276, 23382-23390. Barnes, B. J., Kellum, M. J., Pinder, K. E., Frisancho, J. A. & Pitha, P. M. (2003). Interferon regulatory factor 5, a novel mediator of cell cycle arrest and cell death. Cancer Res 63, 6424-6431. Bennett, C. L., Christie, J., Ramsdell, F. & other authors (2001). The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet 27, 20-21. Benz, C., Heinzel, K. & Bleul, C. C. (2004). Homing of immature thymocytes to the subcapsular microenvironment within the thymus is not an absolute requirement for T cell development. Eur J Immunol 34, 3652-3663. 22 Bettelli, E., Carrier, Y., Gao, W., Korn, T., Strom, T. B., Oukka, M., Weiner, H. L. & Kuchroo, V. K. (2006). Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441, 235-238. Blackburn, C. C. & Manley, N. R. (2004). Developing a new paradigm for thymus organogenesis. Nat Rev Immunol 4, 278-289. Bluestone, J. A., Mackay, C. R., O'Shea, J. J. & Stockinger, B. (2009). The functional plasticity of T cell subsets. Nat Rev Immunol 9, 811-816. Boehm, T. & Bleul, C. C. (2007). The evolutionary history of lymphoid organs. Nat Immunol 8, 131-135. Botti, E., Spallone, G., Moretti, F. & other authors (2011). Developmental factor IRF6 exhibits tumor suppressor activity in squamous cell carcinomas. Proc Natl Acad Sci U S A 108, 13710-13715. Bouker, K. B., Skaar, T. C., Riggins, R. B., Harburger, D. S., Fernandez, D. R., Zwart, A., Wang, A. & Clarke, R. (2005). Interferon regulatory factor-1 (IRF-1) exhibits tumor suppressor activities in breast cancer associated with caspase activation and induction of apoptosis. Carcinogenesis 26, 1527-1535. Brien, J. D., Daffis, S., Lazear, H. M., Cho, H., Suthar, M. S., Gale, M., Jr. & Diamond, M. S. (2011). Interferon regulatory factor-1 (IRF-1) shapes both innate and CD8(+) T cell immune responses against West Nile virus infection. PLoS Pathog 7, e1002230. Brunkow, M. E., Jeffery, E. W., Hjerrild, K. A. & other authors (2001). Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet 27, 68-73. Brustle, A., Heink, S., Huber, M. & other authors (2007). The development of inflammatory T(H)-17 cells requires interferon-regulatory factor 4. Nat Immunol 8, 958966. Chen, Q., Yang, W., Gupta, S., Biswas, P., Smith, P., Bhagat, G. & Pernis, A. B. (2008). IRF-4-binding protein inhibits interleukin-17 and interleukin-21 production by controlling the activity of IRF-4 transcription factor. Immunity 29, 899-911. Chen, W. & Wahl, S. M. (2002). TGF-beta: receptors, signaling pathways and autoimmunity. Curr Dir Autoimmun 5, 62-91. Coccia, E. M., Passini, N., Battistini, A., Pini, C., Sinigaglia, F. & Rogge, L. (1999). Interleukin-12 induces expression of interferon regulatory factor-1 via signal transducer 23 and activator of transcription-4 in human T helper type 1 cells. J Biol Chem 274, 66986703. Colantonio, A. D., Epeldegui, M., Jesiak, M., Jachimowski, L., Blom, B. & Uittenbogaart, C. H. (2011). IFN-alpha is constitutively expressed in the human thymus, but not in peripheral lymphoid organs. PLoS One 6, e24252. Connett, J. M., Badri, L., Giordano, T. J., Connett, W. C. & Doherty, G. M. (2005). Interferon regulatory factor 1 (IRF-1) and IRF-2 expression in breast cancer tissue microarrays. J Interferon Cytokine Res 25, 587-594. Dander, E., Balduzzi, A., Zappa, G. & other authors (2009). Interleukin-17-producing T-helper cells as new potential player mediating graft-versus-host disease in patients undergoing allogeneic stem-cell transplantation. Transplantation 88, 1261-1272. De Jager, P. L., Jia, X., Wang, J. & other authors (2009). Meta-analysis of genome scans and replication identify CD6, IRF8 and TNFRSF1A as new multiple sclerosis susceptibility loci. Nat Genet 41, 776-782. Demirci, F. Y., Manzi, S., Ramsey-Goldman, R. & other authors (2007). Association of a common interferon regulatory factor 5 (IRF5) variant with increased risk of systemic lupus erythematosus (SLE). Ann Hum Genet 71, 308-311. Deng, M. & Daley, G. Q. (2001). Expression of interferon consensus sequence binding protein induces potent immunity against BCR/ABL-induced leukemia. Blood 97, 34913497. Dong, C. (2006). Diversification of T-helper-cell lineages: finding the family root of IL17-producing cells. Nat Rev Immunol 6, 329-333. Dong, C. (2008). TH17 cells in development: an updated view of their molecular identity and genetic programming. Nat Rev Immunol 8, 337-348. Dooley, J., Erickson, M., Gillard, G. O. & Farr, A. G. (2006). Cervical thymus in the mouse. J Immunol 176, 6484-6490. Duguay, D., Mercier, F., Stagg, J., Martineau, D., Bramson, J., Servant, M., Lin, R., Galipeau, J. & Hiscott, J. (2002). In vivo interferon regulatory factor 3 tumor suppressor activity in B16 melanoma tumors. Cancer Res 62, 5148-5152. Duncan, G. S., Mittrucker, H. W., Kagi, D., Matsuyama, T. & Mak, T. W. (1996). The transcription factor interferon regulatory factor-1 is essential for natural killer cell function in vivo. J Exp Med 184, 2043-2048. 24 Elser, B., Lohoff, M., Kock, S., Giaisi, M., Kirchhoff, S., Krammer, P. H. & Li-Weber, M. (2002). IFN-gamma represses IL-4 expression via IRF-1 and IRF-2. Immunity 17, 703-712. Fantuzzi, G., Reed, D., Qi, M., Scully, S., Dinarello, C. A. & Senaldi, G. (2001). Role of interferon regulatory factor-1 in the regulation of IL-18 production and activity. Eur J Immunol 31, 369-375. Fanzo, J. C., Yang, W., Jang, S. Y., Gupta, S., Chen, Q., Siddiq, A., Greenberg, S. & Pernis, A. B. (2006). Loss of IRF-4-binding protein leads to the spontaneous development of systemic autoimmunity. J Clin Invest 116, 703-714. Fietta, P. & Delsante, G. (2009). The effector T helper cell triade. Riv Biol 102, 61-74. Fragale, A., Gabriele, L., Stellacci, E. & other authors (2008). IFN regulatory factor-1 negatively regulates CD4+ CD25+ regulatory T cell differentiation by repressing Foxp3 expression. J Immunol 181, 1673-1682. Fujita, T., Kimura, Y., Miyamoto, M., Barsoumian, E. L. & Taniguchi, T. (1989). Induction of endogenous IFN-alpha and IFN-beta genes by a regulatory transcription factor, IRF-1. Nature 337, 270-272. Gabriele, L., Fragale, A., Borghi, P. & other authors (2006). IRF-1 deficiency skews the differentiation of dendritic cells toward plasmacytoid and tolerogenic features. J Leukoc Biol 80, 1500-1511. Galon, J., Sudarshan, C., Ito, S., Finbloom, D. & O'Shea, J. J. (1999). IL-12 induces IFN regulating factor-1 (IRF-1) gene expression in human NK and T cells. J Immunol 162, 7256-7262. Gateva, V., Sandling, J. K., Hom, G. & other authors (2009). A large-scale replication study identifies TNIP1, PRDM1, JAZF1, UHRF1BP1 and IL10 as risk loci for systemic lupus erythematosus. Nat Genet 41, 1228-1233. Germain, R. N. (2002). T-cell development and the CD4-CD8 lineage decision. Nat Rev Immunol 2, 309-322. Giatromanolaki, A., Koukourakis, M. I., Ritis, K., Mimidis, K. & Sivridis, E. (2004). Interferon regulatory factor-1 (IRF-1) suppression and derepression during endometrial tumorigenesis and cancer progression. Cytokine 26, 164-168. Giese, N. A., Gabriele, L., Doherty, T. M., Klinman, D. M., Tadesse-Heath, L., Contursi, C., Epstein, S. L. & Morse, H. C., 3rd (1997). Interferon (IFN) consensus sequence-binding protein, a transcription factor of the IFN regulatory factor family, regulates immune responses in vivo through control of interleukin 12 expression. J Exp Med 186, 1535-1546. 25 Godfrey, D. I., Kennedy, J., Suda, T. & Zlotnik, A. (1993). A developmental pathway involving four phenotypically and functionally distinct subsets of CD3-CD4-CD8- triplenegative adult mouse thymocytes defined by CD44 and CD25 expression. J Immunol 150, 4244-4252. Gordon, J., Wilson, V. A., Blair, N. F., Sheridan, J., Farley, A., Wilson, L., Manley, N. R. & Blackburn, C. C. (2004). Functional evidence for a single endodermal origin for the thymic epithelium. Nat Immunol 5, 546-553. Graham, R. R., Kozyrev, S. V., Baechler, E. C. & other authors (2006). A common haplotype of interferon regulatory factor 5 (IRF5) regulates splicing and expression and is associated with increased risk of systemic lupus erythematosus. Nat Genet 38, 550555. Green, D. R., Flood, P. M. & Gershon, R. K. (1983). Immunoregulatory T-cell pathways. Annu Rev Immunol 1, 439-463. Hao, S. X. & Ren, R. (2000). Expression of interferon consensus sequence binding protein (ICSBP) is downregulated in Bcr-Abl-induced murine chronic myelogenous leukemia-like disease, and forced coexpression of ICSBP inhibits Bcr-Abl-induced myeloproliferative disorder. Mol Cell Biol 20, 1149-1161. Harada, H., Kitagawa, M., Tanaka, N., Yamamoto, H., Harada, K., Ishihara, M. & Taniguchi, T. (1993). Anti-oncogenic and oncogenic potentials of interferon regulatory factors-1 and -2. Science 259, 971-974. Harley, J. B., Alarcon-Riquelme, M. E., Criswell, L. A. & other authors (2008). Genome-wide association scan in women with systemic lupus erythematosus identifies susceptibility variants in ITGAM, PXK, KIAA1542 and other loci. Nat Genet 40, 204-210. Haynes, B. F. & Heinly, C. S. (1995). Early human T cell development: analysis of the human thymus at the time of initial entry of hematopoietic stem cells into the fetal thymic microenvironment. J Exp Med 181, 1445-1458. Heylbroeck, C., Balachandran, S., Servant, M. J., DeLuca, C., Barber, G. N., Lin, R. & Hiscott, J. (2000). The IRF-3 transcription factor mediates Sendai virus-induced apoptosis. J Virol 74, 3781-3792. Hida, S., Tadachi, M., Saito, T. & Taki, S. (2005). Negative control of basophil expansion by IRF-2 critical for the regulation of Th1/Th2 balance. Blood 106, 20112017. Hirahara, K., Vahedi, G., Ghoreschi, K., Yang, X. P., Nakayamada, S., Kanno, Y., O'Shea, J. J. & Laurence, A. (2011). Helper T-cell differentiation and plasticity: insights from epigenetics. Immunology 134, 235-245. 26 Hoffmann, R., Bruno, L., Seidl, T., Rolink, A. & Melchers, F. (2003). Rules for gene usage inferred from a comparison of large-scale gene expression profiles of T and B lymphocyte development. J Immunol 170, 1339-1353. Hollander, G., Gill, J., Zuklys, S., Iwanami, N., Liu, C. & Takahama, Y. (2006). Cellular and molecular events during early thymus development. Immunol Rev 209, 2846. Holtschke, T., Lohler, J., Kanno, Y. & other authors (1996). Immunodeficiency and chronic myelogenous leukemia-like syndrome in mice with a targeted mutation of the ICSBP gene. Cell 87, 307-317. Honda, K., Mizutani, T. & Taniguchi, T. (2004). Negative regulation of IFN-alpha/beta signaling by IFN regulatory factor 2 for homeostatic development of dendritic cells. Proc Natl Acad Sci U S A 101, 2416-2421. Honda, K., Ohba, Y., Yanai, H., Negishi, H., Mizutani, T., Takaoka, A., Taya, C. & Taniguchi, T. (2005a). Spatiotemporal regulation of MyD88-IRF-7 signalling for robust type-I interferon induction. Nature 434, 1035-1040. Honda, K., Yanai, H., Negishi, H. & other authors (2005b). IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature 434, 772-777. Honda, K., Takaoka, A. & Taniguchi, T. (2006). Type I interferon [corrected] gene induction by the interferon regulatory factor family of transcription factors. Immunity 25, 349-360. Honda, K. & Taniguchi, T. (2006). IRFs: master regulators of signalling by Toll-like receptors and cytosolic pattern-recognition receptors. Nat Rev Immunol 6, 644-658. Hoshino, K., Sugiyama, T., Matsumoto, M., Tanaka, T., Saito, M., Hemmi, H., Ohara, O., Akira, S. & Kaisho, T. (2006). IkappaB kinase-alpha is critical for interferonalpha production induced by Toll-like receptors 7 and 9. Nature 440, 949-953. Hrdlickova, R., Nehyba, J. & Bose, H. R., Jr. (2001). Interferon regulatory factor 4 contributes to transformation of v-Rel-expressing fibroblasts. Mol Cell Biol 21, 63696386. Hu, C. M., Jang, S. Y., Fanzo, J. C. & Pernis, A. B. (2002). Modulation of T cell cytokine production by interferon regulatory factor-4. J Biol Chem 277, 49238-49246. Hu, G., Mancl, M. E. & Barnes, B. J. (2005). Signaling through IFN regulatory factor-5 sensitizes p53-deficient tumors to DNA damage-induced apoptosis and cell death. Cancer Res 65, 7403-7412. 27 Hu, J., Mao, Y., Li, M. & Lu, Y. (2011). The profile of Th17 subset in glioma. Int Immunopharmacol 11, 1173-1179. Huang, B., Qi, Z. T., Xu, Z. & Nie, P. (2010). Global characterization of interferon regulatory factor (IRF) genes in vertebrates: glimpse of the diversification in evolution. BMC Immunol 11, 22. Ichikawa, E., Hida, S., Omatsu, Y., Shimoyama, S., Takahara, K., Miyagawa, S., Inaba, K. & Taki, S. (2004). Defective development of splenic and epidermal CD4+ dendritic cells in mice deficient for IFN regulatory factor-2. Proc Natl Acad Sci U S A 101, 3909-3914. Iida, S., Rao, P. H., Butler, M., Corradini, P., Boccadoro, M., Klein, B., Chaganti, R. S. & Dalla-Favera, R. (1997). Deregulation of MUM1/IRF4 by chromosomal translocation in multiple myeloma. Nat Genet 17, 226-230. Ingraham, C. R., Kinoshita, A., Kondo, S. & other authors (2006). Abnormal skin, limb and craniofacial morphogenesis in mice deficient for interferon regulatory factor 6 (Irf6). Nat Genet 38, 1335-1340. Irla, M., Hugues, S., Gill, J. & other authors (2008). Autoantigen-specific interactions with CD4+ thymocytes control mature medullary thymic epithelial cell cellularity. Immunity 29, 451-463. Irving, B. A., Alt, F. W. & Killeen, N. (1998). Thymocyte development in the absence of pre-T cell receptor extracellular immunoglobulin domains. Science 280, 905-908. Ito, M., Iida, S., Inagaki, H. & other authors (2002). MUM1/IRF4 expression is an unfavorable prognostic factor in B-cell chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL). Jpn J Cancer Res 93, 685-694. Itoi, M., Tsukamoto, N., Yoshida, H. & Amagai, T. (2007). Mesenchymal cells are required for functional development of thymic epithelial cells. Int Immunol 19, 953-964. Ivanov, II, McKenzie, B. S., Zhou, L., Tadokoro, C. E., Lepelley, A., Lafaille, J. J., Cua, D. J. & Littman, D. R. (2006). The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 126, 11211133. Izaguirre, A., Barnes, B. J., Amrute, S. & other authors (2003). Comparative analysis of IRF and IFN-alpha expression in human plasmacytoid and monocyte-derived dendritic cells. J Leukoc Biol 74, 1125-1138. Janeway, C. A., Travers, P., Walport, M. & Shlomchik, M. J. (2001). Immunobiology, 5th edition edn. New York: Garland Science 28 Jenkinson, W. E., Jenkinson, E. J. & Anderson, G. (2003). Differential requirement for mesenchyme in the proliferation and maturation of thymic epithelial progenitors. J Exp Med 198, 325-332. Jenkinson, W. E., Rossi, S. W., Parnell, S. M., Jenkinson, E. J. & Anderson, G. (2007). PDGFRalpha-expressing mesenchyme regulates thymus growth and the availability of intrathymic niches. Blood 109, 954-960. Jo, S. H., Schatz, J. H., Acquaviva, J., Singh, H. & Ren, R. (2010). Cooperation between deficiencies of IRF-4 and IRF-8 promotes both myeloid and lymphoid tumorigenesis. Blood 116, 2759-2767. Jo, S. H. & Ren, R. (2011). IRF-4 suppresses BCR/ABL transformation of myeloid cells in a DNA binding-independent manner. J Biol Chem 287, 1770-1778. Johnston, M. C. (1966). A radioautographic study of the migration and fate of cranial neural crest cells in the chick embryo. Anat Rec 156, 143-155. Josefowicz, S. Z., Niec, R. E., Kim, H. Y., Treuting, P., Chinen, T., Zheng, Y., Umetsu, D. T. & Rudensky, A. Y. (2012). Extrathymically generated regulatory T cells control mucosal TH2 inflammation. Nature 482, 395-399. Kamijo, R., Harada, H., Matsuyama, T. & other authors (1994). Requirement for transcription factor IRF-1 in NO synthase induction in macrophages. Science 263, 16121615. Kano, A., Haruyama, T., Akaike, T. & Watanabe, Y. (1999). IRF-1 is an essential mediator in IFN-gamma-induced cell cycle arrest and apoptosis of primary cultured hepatocytes. Biochem Biophys Res Commun 257, 672-677. Kim, T., Kim, T. Y., Song, Y. H., Min, I. M., Yim, J. & Kim, T. K. (1999). Activation of interferon regulatory factor 3 in response to DNA-damaging agents. J Biol Chem 274, 30686-30689. Kim, T. Y., Lee, K. H., Chang, S., Chung, C., Lee, H. W., Yim, J. & Kim, T. K. (2003). Oncogenic potential of a dominant negative mutant of interferon regulatory factor 3. J Biol Chem 278, 15272-15278. Klein, U., Casola, S., Cattoretti, G., Shen, Q., Lia, M., Mo, T., Ludwig, T., Rajewsky, K. & Dalla-Favera, R. (2006). Transcription factor IRF4 controls plasma cell differentiation and class-switch recombination. Nat Immunol 7, 773-782. Klug, D. B., Carter, C., Gimenez-Conti, I. B. & Richie, E. R. (2002). Cutting edge: thymocyte-independent and thymocyte-dependent phases of epithelial patterning in the fetal thymus. J Immunol 169, 2842-2845. 29 Kolls, J. K. & Linden, A. (2004). Interleukin-17 family members and inflammation. Immunity 21, 467-476. Kondo, S., Schutte, B. C., Richardson, R. J. & other authors (2002). Mutations in IRF6 cause Van der Woude and popliteal pterygium syndromes. Nat Genet 32, 285289. Le, M., Naridze, R., Morrison, J., Biggs, L. C., Rhea, L., Schutte, B. C., Kaartinen, V. & Dunnwald, M. (2012). Transforming growth factor Beta 3 is required for excisional wound repair in vivo. PLoS One 7, e48040. Lee, C. H., Melchers, M., Wang, H. & other authors (2006). Regulation of the germinal center gene program by interferon (IFN) regulatory factor 8/IFN consensus sequence-binding protein. J Exp Med 203, 63-72. Lee, C. K., Gimeno, R. & Levy, D. E. (1999). Differential regulation of constitutive major histocompatibility complex class I expression in T and B lymphocytes. J Exp Med 190, 1451-1464. Leppa, V., Surakka, I., Tienari, P. & other authors (2011). The genetic association of variants in CD6, TNFRSF1A and IRF8 to multiple sclerosis: a multicenter case-control study. PLoS One 6, e18813. Li, S., Wang, L., Berman, M., Kong, Y. Y. & Dorf, M. E. (2011). Mapping a dynamic innate immunity protein interaction network regulating type I interferon production. Immunity 35, 426-440. Lind, E. F., Prockop, S. E., Porritt, H. E. & Petrie, H. T. (2001). Mapping precursor movement through the postnatal thymus reveals specific microenvironments supporting defined stages of early lymphoid development. J Exp Med 194, 127-134. Litman, G. W. & Cooper, M. D. (2007). Why study the evolution of immunity? Nat Immunol 8, 547-548. Little, H. J., Rorick, N. K., Su, L. I. & other authors (2009). Missense mutations that cause Van der Woude syndrome and popliteal pterygium syndrome affect the DNAbinding and transcriptional activation functions of IRF6. Hum Mol Genet 18, 535-545. Liu, C., Saito, F., Liu, Z. & other authors (2006). Coordination between CCR7- and CCR9-mediated chemokine signals in prevascular fetal thymus colonization. Blood 108, 2531-2539. Liu, J., Cao, S., Herman, L. M. & Ma, X. (2003). Differential regulation of interleukin (IL)-12 p35 and p40 gene expression and interferon (IFN)-gamma-primed IL-12 production by IFN regulatory factor 1. J Exp Med 198, 1265-1276. 30 Liu, K. & Abrams, S. I. (2003). Coordinate regulation of IFN consensus sequencebinding protein and caspase-1 in the sensitization of human colon carcinoma cells to Fas-mediated apoptosis by IFN-gamma. J Immunol 170, 6329-6337. Lohoff, M., Ferrick, D., Mittrucker, H. W., Duncan, G. S., Bischof, S., Rollinghoff, M. & Mak, T. W. (1997). Interferon regulatory factor-1 is required for a T helper 1 immune response in vivo. Immunity 6, 681-689. Lohoff, M., Duncan, G. S., Ferrick, D. & other authors (2000). Deficiency in the transcription factor interferon regulatory factor (IRF)-2 leads to severely compromised development of natural killer and T helper type 1 cells. J Exp Med 192, 325-336. Lohoff, M., Mittrucker, H. W., Prechtl, S. & other authors (2002). Dysregulated T helper cell differentiation in the absence of interferon regulatory factor 4. Proc Natl Acad Sci U S A 99, 11808-11812. Lohoff, M. & Mak, T. W. (2005). Roles of interferon-regulatory factors in T-helper-cell differentiation. Nat Rev Immunol 5, 125-135. Lu, R., Medina, K. L., Lancki, D. W. & Singh, H. (2003). IRF-4,8 orchestrate the pre-Bto-B transition in lymphocyte development. Genes Dev 17, 1703-1708. Ma, H., Lu, C., Ziegler, J., Liu, A., Sepulveda, A., Okada, H., Lentzsch, S. & Mapara, M. Y. (2011). Absence of Stat1 in donor CD4(+) T cells promotes the expansion of Tregs and reduces graft-versus-host disease in mice. J Clin Invest 121, 2554-2569. Ma, S., Turetsky, A., Trinh, L. & Lu, R. (2006). IFN regulatory factor 4 and 8 promote Ig light chain kappa locus activation in pre-B cell development. J Immunol 177, 78987904. Maddur, M. S., Miossec, P., Kaveri, S. V. & Bayry, J. (2013). Th17 cells: biology, pathogenesis of autoimmune and inflammatory diseases, and therapeutic strategies. Am J Pathol 181, 8-18. Marie, I., Durbin, J. E. & Levy, D. E. (1998). Differential viral induction of distinct interferon-alpha genes by positive feedback through interferon regulatory factor-7. EMBO J 17, 6660-6669. Maruyama, S., Sumita, K., Shen, H., Kanoh, M., Xu, X., Sato, M., Matsumoto, M., Shinomiya, H. & Asano, Y. (2003). Identification of IFN regulatory factor-1 binding site in IL-12 p40 gene promoter. J Immunol 170, 997-1001. Maruyama, T., Li, J., Vaque, J. P. & other authors (2011). Control of the differentiation of regulatory T cells and T(H)17 cells by the DNA-binding inhibitor Id3. Nat Immunol 12, 86-95. 31 Matsui, K., Kumagai, Y., Kato, H., Sato, S., Kawagoe, T., Uematsu, S., Takeuchi, O. & Akira, S. (2006). Cutting edge: Role of TANK-binding kinase 1 and inducible IkappaB kinase in IFN responses against viruses in innate immune cells. J Immunol 177, 57855789. Matsuoka, K., Inoue, N., Sato, T. & other authors (2004). T-bet upregulation and subsequent interleukin 12 stimulation are essential for induction of Th1 mediated immunopathology in Crohn's disease. Gut 53, 1303-1308. Matsuyama, T., Kimura, T., Kitagawa, M. & other authors (1993). Targeted disruption of IRF-1 or IRF-2 results in abnormal type I IFN gene induction and aberrant lymphocyte development. Cell 75, 83-97. McGeachy, M. J., Chen, Y., Tato, C. M., Laurence, A., Joyce-Shaikh, B., Blumenschein, W. M., McClanahan, T. K., O'Shea, J. J. & Cua, D. J. (2009). The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17producing effector T helper cells in vivo. Nat Immunol 10, 314-324. Michie, A. M. & Zuniga-Pflucker, J. C. (2002). Regulation of thymocyte differentiation: pre-TCR signals and beta-selection. Semin Immunol 14, 311-323. Miossec, P., Korn, T. & Kuchroo, V. K. (2009). Interleukin-17 and type 17 helper T cells. N Engl J Med 361, 888-898. Mitsdoerffer, M., Lee, Y., Jager, A., Kim, H. J., Korn, T., Kolls, J. K., Cantor, H., Bettelli, E. & Kuchroo, V. K. (2010). Proinflammatory T helper type 17 cells are effective B-cell helpers. Proc Natl Acad Sci U S A 107, 14292-14297. Mittrucker, H. W., Matsuyama, T., Grossman, A. & other authors (1997). Requirement for the transcription factor LSIRF/IRF4 for mature B and T lymphocyte function. Science 275, 540-543. Mori, T., Anazawa, Y., Iiizumi, M., Fukuda, S., Nakamura, Y. & Arakawa, H. (2002). Identification of the interferon regulatory factor 5 gene (IRF-5) as a direct target for p53. Oncogene 21, 2914-2918. Moynagh, P. N. (2005). TLR signalling and activation of IRFs: revisiting old friends from the NF-kappaB pathway. Trends Immunol 26, 469-476. Murphy, K. M. & Reiner, S. L. (2002). The lineage decisions of helper T cells. Nat Rev Immunol 2, 933-944. Negishi, H., Ohba, Y., Yanai, H., Takaoka, A., Honma, K., Yui, K., Matsuyama, T., Taniguchi, T. & Honda, K. (2005). Negative regulation of Toll-like-receptor signaling by IRF-4. Proc Natl Acad Sci U S A 102, 15989-15994. 32 Nehyba, J., Hrdlickova, R. & Bose, H. R. (2009). Dynamic evolution of immune system regulators: the history of the interferon regulatory factor family. Mol Biol Evol 26, 2539-2550. Niedbala, W., Wei, X. Q., Campbell, C., Thomson, D., Komai-Koma, M. & Liew, F. Y. (2002). Nitric oxide preferentially induces type 1 T cell differentiation by selectively upregulating IL-12 receptor beta 2 expression via cGMP. Proc Natl Acad Sci U S A 99, 16186-16191. Nordang, G. B., Viken, M. K., Amundsen, S. S., Sanchez, E. S., Flato, B., Forre, O. T., Martin, J., Kvien, T. K. & Lie, B. A. (2011). Interferon regulatory factor 5 gene polymorphism confers risk to several rheumatic diseases and correlates with expression of alternative thymic transcripts. Rheumatology (Oxford) 51, 619-626. O'Garra, A. & Arai, N. (2000). The molecular basis of T helper 1 and T helper 2 cell differentiation. Trends Cell Biol 10, 542-550. Ogasawara, K., Hida, S., Azimi, N., Tagaya, Y., Sato, T., Yokochi-Fukuda, T., Waldmann, T. A., Taniguchi, T. & Taki, S. (1998). Requirement for IRF-1 in the microenvironment supporting development of natural killer cells. Nature 391, 700-703. Oppmann, B., Lesley, R., Blom, B. & other authors (2000). Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity 13, 715-725. Ortmann, C. A., Burchert, A., Holzle, K., Nitsche, A., Wittig, B., Neubauer, A. & Schmidt, M. (2005). Down-regulation of interferon regulatory factor 4 gene expression in leukemic cells due to hypermethylation of CpG motifs in the promoter region. Nucleic Acids Res 33, 6895-6905. Oukka, M. (2008). Th17 cells in immunity and autoimmunity. Ann Rheum Dis 67 Suppl 3, iii26-29. Ouyang, X., Zhang, R., Yang, J. & other authors (2011). Transcription factor IRF8 directs a silencing programme for TH17 cell differentiation. Nat Commun 2, 314. Owen, J. J. & Ritter, M. A. (1969). Tissue interaction in the development of thymus lymphocytes. J Exp Med 129, 431-442. Pan, F., Fan, H., Lu, L., Liu, Z. & Jiang, S. (2011). The yin and yang of signaling in Tregs and TH17 cells. Sci Signal 4, mr4. Passioura, T., Dolnikov, A., Shen, S. & Symonds, G. (2005a). N-ras-induced growth suppression of myeloid cells is mediated by IRF-1. Cancer Res 65, 797-804. 33 Passioura, T., Shen, S., Symonds, G. & Dolnikov, A. (2005b). A retroviral library genetic screen identifies IRF-2 as an inhibitor of N-ras-induced growth suppression in leukemic cells. Oncogene 24, 7327-7336. Patel, D. R. (2011). Linking interferon regulatory factors to the pathogenesis of human lupus. Transl Res 157, 323-325. Pathak, S., Ma, S., Trinh, L., Eudy, J., Wagner, K. U., Joshi, S. S. & Lu, R. (2011). IRF4 is a suppressor of c-Myc induced B cell leukemia. PLoS One 6, e22628. Paul, W. E. (2008 ). Fundamental Immunology, sixth edition edn: Lippincott Williams & Wilkins. Pearse, M., Wu, L., Egerton, M., Wilson, A., Shortman, K. & Scollay, R. (1989). A murine early thymocyte developmental sequence is marked by transient expression of the interleukin 2 receptor. Proc Natl Acad Sci U S A 86, 1614-1618. Penninger, J. M., Sirard, C., Mittrucker, H. W. & other authors (1997). The interferon regulatory transcription factor IRF-1 controls positive and negative selection of CD8+ thymocytes. Immunity 7, 243-254. Penninger, J. M. & Mak, T. W. (1998). Thymocyte selection in Vav and IRF-1 genedeficient mice. Immunol Rev 165, 149-166. Plotkin, J., Prockop, S. E., Lepique, A. & Petrie, H. T. (2003). Critical role for CXCR4 signaling in progenitor localization and T cell differentiation in the postnatal thymus. J Immunol 171, 4521-4527. Prockop, S. & Petrie, H. T. (2000). Cell migration and the anatomic control of thymocyte precursor differentiation. Semin Immunol 12, 435-444. Rahimov, F., Marazita, M. L., Visel, A. & other authors (2008). Disruption of an AP2alpha binding site in an IRF6 enhancer is associated with cleft lip. Nat Genet 40, 13411347. Raulet, D. H., Garman, R. D., Saito, H. & Tonegawa, S. (1985). Developmental regulation of T-cell receptor gene expression. Nature 314, 103-107. Rautajoki, K. J., Kylaniemi, M. K., Raghav, S. K., Rao, K. & Lahesmaa, R. (2008). An insight into molecular mechanisms of human T helper cell differentiation. Ann Med 40, 322-335. Rengarajan, J., Mowen, K. A., McBride, K. D., Smith, E. D., Singh, H. & Glimcher, L. H. (2002). Interferon regulatory factor 4 (IRF4) interacts with NFATc2 to modulate interleukin 4 gene expression. J Exp Med 195, 1003-1012. 34 Richardson, R. J., Dixon, J., Malhotra, S., Hardman, M. J., Knowles, L., BootHandford, R. P., Shore, P., Whitmarsh, A. & Dixon, M. J. (2006). Irf6 is a key determinant of the keratinocyte proliferation-differentiation switch. Nat Genet 38, 13291334. Richardson, R. J., Dixon, J., Jiang, R. & Dixon, M. J. (2009). Integration of IRF6 and Jagged2 signalling is essential for controlling palatal adhesion and fusion competence. Hum Mol Genet 18, 2632-2642. Rodewald, H. R. (2008). Thymus organogenesis. Annu Rev Immunol 26, 355-388. Romeo, G., Fiorucci, G., Chiantore, M. V., Percario, Z. A., Vannucchi, S. & Affabris, E. (2002). IRF-1 as a negative regulator of cell proliferation. J Interferon Cytokine Res 22, 39-47. Sabel, J. L., d'Alencon, C., O'Brien, E. K., Van Otterloo, E., Lutz, K., Cuykendall, T. N., Schutte, B. C., Houston, D. W. & Cornell, R. A. (2009). Maternal Interferon Regulatory Factor 6 is required for the differentiation of primary superficial epithelia in Danio and Xenopus embryos. Dev Biol 325, 249-262. Salkowski, C. A., Kopydlowski, K., Blanco, J., Cody, M. J., McNally, R. & Vogel, S. N. (1999). IL-12 is dysregulated in macrophages from IRF-1 and IRF-2 knockout mice. J Immunol 163, 1529-1536. Sanchez, F. O., Linga Reddy, M. V., Mallbris, L., Sakuraba, K., Stahle, M. & Alarcon-Riquelme, M. E. (2008). IFN-regulatory factor 5 gene variants interact with the class I MHC locus in the Swedish psoriasis population. J Invest Dermatol 128, 17041709. Sato, M., Tanaka, N., Hata, N., Oda, E. & Taniguchi, T. (1998). Involvement of the IRF family transcription factor IRF-3 in virus-induced activation of the IFN-beta gene. FEBS Lett 425, 112-116. Sato, M., Suemori, H., Hata, N. & other authors (2000). Distinct and essential roles of transcription factors IRF-3 and IRF-7 in response to viruses for IFN-alpha/beta gene induction. Immunity 13, 539-548. Scharton-Kersten, T., Contursi, C., Masumi, A., Sher, A. & Ozato, K. (1997). Interferon consensus sequence binding protein-deficient mice display impaired resistance to intracellular infection due to a primary defect in interleukin 12 p40 induction. J Exp Med 186, 1523-1534. Scheller, M., Foerster, J., Heyworth, C. M., Waring, J. F., Lohler, J., Gilmore, G. L., Shadduck, R. K., Dexter, T. M. & Horak, I. (1999). Altered development and cytokine responses of myeloid progenitors in the absence of transcription factor, interferon consensus sequence binding protein. Blood 94, 3764-3771. 35 Schiavoni, G., Mattei, F., Sestili, P., Borghi, P., Venditti, M., Morse, H. C., 3rd, Belardelli, F. & Gabriele, L. (2002). ICSBP is essential for the development of mouse type I interferon-producing cells and for the generation and activation of CD8alpha(+) dendritic cells. J Exp Med 196, 1415-1425. Schiavoni, G., Mattei, F., Borghi, P., Sestili, P., Venditti, M., Morse, H. C., 3rd, Belardelli, F. & Gabriele, L. (2004). ICSBP is critically involved in the normal development and trafficking of Langerhans cells and dermal dendritic cells. Blood 103, 2221-2228. Schmidt, M., Hochhaus, A., Konig-Merediz, S. A. & other authors (2000). Expression of interferon regulatory factor 4 in chronic myeloid leukemia: correlation with response to interferon alfa therapy. J Clin Oncol 18, 3331-3338. Sciammas, R., Shaffer, A. L., Schatz, J. H., Zhao, H., Staudt, L. M. & Singh, H. (2006). Graded expression of interferon regulatory factor-4 coordinates isotype switching with plasma cell differentiation. Immunity 25, 225-236. Shaffer, A. L., Emre, N. C., Lamy, L. & other authors (2008). IRF4 addiction in multiple myeloma. Nature 454, 226-231. Shevach, E. D., T. (2010). Regulatory T cells. Nature reviews of Immunology 10. Shinkai, Y., Rathbun, G., Lam, K. P. & other authors (1992). RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell 68, 855867. Simon, A. K., Desrois, M. & Schmitt-Verhulst, A. M. (1997). Interferon-regulatory factors during development of CD4 and CD8 thymocytes. Immunology 91, 340-345. Singer, K. H., Wolf, L. S., Lobach, D. F., Denning, S. M., Tuck, D. T., Robertson, A. L. & Haynes, B. F. (1986). Human thymocytes bind to autologous and allogeneic thymic epithelial cells in vitro. Proc Natl Acad Sci U S A 83, 6588-6592. Stransky, N., Egloff, A. M., Tward, A. D. & other authors (2011). The mutational landscape of head and neck squamous cell carcinoma. Science 333, 1157-1160. Suzuki, S., Honma, K., Matsuyama, T. & other authors (2004). Critical roles of interferon regulatory factor 4 in CD11bhighCD8alpha- dendritic cell development. Proc Natl Acad Sci U S A 101, 8981-8986. Tailor, P., Tamura, T., Kong, H. J., Kubota, T., Kubota, M., Borghi, P., Gabriele, L. & Ozato, K. (2007). The feedback phase of type I interferon induction in dendritic cells requires interferon regulatory factor 8. Immunity 27, 228-239. 36 Takahama, Y. (2006). Journey through the thymus: stromal guides for T-cell development and selection. Nat Rev Immunol 6, 127-135. Takaoka, A., Yanai, H., Kondo, S. & other authors (2005). Integral role of IRF-5 in the gene induction programme activated by Toll-like receptors. Nature 434, 243-249. Taki, S., Sato, T., Ogasawara, K. & other authors (1997). Multistage regulation of Th1-type immune responses by the transcription factor IRF-1. Immunity 6, 673-679. Taki, S., Nakajima, S., Ichikawa, E., Saito, T. & Hida, S. (2005). IFN regulatory factor2 deficiency revealed a novel checkpoint critical for the generation of peripheral NK cells. J Immunol 174, 6005-6012. Tamura, T., Nagamura-Inoue, T., Shmeltzer, Z., Kuwata, T. & Ozato, K. (2000). ICSBP directs bipotential myeloid progenitor cells to differentiate into mature macrophages. Immunity 13, 155-165. Tamura, T., Tailor, P., Yamaoka, K., Kong, H. J., Tsujimura, H., O'Shea, J. J., Singh, H. & Ozato, K. (2005a). IFN regulatory factor-4 and -8 govern dendritic cell subset development and their functional diversity. J Immunol 174, 2573-2581. Tamura, T., Thotakura, P., Tanaka, T. S., Ko, M. S. & Ozato, K. (2005b). Identification of target genes and a unique cis element regulated by IRF-8 in developing macrophages. Blood 106, 1938-1947. Tamura, T., Yanai, H., Savitsky, D. & Taniguchi, T. (2008). The IRF family transcription factors in immunity and oncogenesis. Annu Rev Immunol 26, 535-584. Tanaka, N., Ishihara, M., Kitagawa, M. & other authors (1994). Cellular commitment to oncogene-induced transformation or apoptosis is dependent on the transcription factor IRF-1. Cell 77, 829-839. Tanaka, N., Ishihara, M., Lamphier, M. S. & other authors (1996). Cooperation of the tumour suppressors IRF-1 and p53 in response to DNA damage. Nature 382, 816-818. Taniguchi, T., Ogasawara, K., Takaoka, A. & Tanaka, N. (2001). IRF family of transcription factors as regulators of host defense. Annu Rev Immunol 19, 623-655. Taniguchi, T. & Takaoka, A. (2002). The interferon-alpha/beta system in antiviral responses: a multimodal machinery of gene regulation by the IRF family of transcription factors. Curr Opin Immunol 14, 111-116. Terszowski, G., Muller, S. M., Bleul, C. C. & other authors (2006). Evidence for a functional second thymus in mice. Science 312, 284-287. 37 Tesmer, L. A., Lundy, S. K., Sarkar, S. & Fox, D. A. (2008). Th17 cells in human disease. Immunol Rev 223, 87-113. Testa, U., Stellacci, E., Pelosi, E. & other authors (2004). Impaired myelopoiesis in mice devoid of interferon regulatory factor 1. Leukemia 18, 1864-1871. Thomason, H. A., Zhou, H., Kouwenhoven, E. N. & other authors (2010). Cooperation between the transcription factors p63 and IRF6 is essential to prevent cleft palate in mice. J Clin Invest 120, 1561-1569. Tian, Y., Wang, C., Ye, Z., Xiao, X., Kijlstra, A. & Yang, P. (2012). Effect of 1,25dihydroxyvitamin D3 on Th17 and Th1 response in patients with Behcet's disease. Invest Ophthalmol Vis Sci 53, 6434-6441. Tominaga, N., Ohkusu-Tsukada, K., Udono, H., Abe, R., Matsuyama, T. & Yui, K. (2003). Development of Th1 and not Th2 immune responses in mice lacking IFNregulatory factor-4. Int Immunol 15, 1-10. Tourigny, M. R., Mazel, S., Burtrum, D. B. & Petrie, H. T. (1997). T cell receptor (TCR)-beta gene recombination: dissociation from cell cycle regulation and developmental progression during T cell ontogeny. J Exp Med 185, 1549-1556. Tsuboi, K., Iida, S., Inagaki, H. & other authors (2000). MUM1/IRF4 expression as a frequent event in mature lymphoid malignancies. Leukemia 14, 449-456. Tsujimura, H., Nagamura-Inoue, T., Tamura, T. & Ozato, K. (2002). IFN consensus sequence binding protein/IFN regulatory factor-8 guides bone marrow progenitor cells toward the macrophage lineage. J Immunol 169, 1261-1269. Tsujimura, H., Tamura, T. & Ozato, K. (2003). Cutting edge: IFN consensus sequence binding protein/IFN regulatory factor 8 drives the development of type I IFN-producing plasmacytoid dendritic cells. J Immunol 170, 1131-1135. Tsujimura, H., Tamura, T., Kong, H. J., Nishiyama, A., Ishii, K. J., Klinman, D. M. & Ozato, K. (2004). Toll-like receptor 9 signaling activates NF-kappaB through IFN regulatory factor-8/IFN consensus sequence binding protein in dendritic cells. J Immunol 172, 6820-6827. von Boehmer, H. & Fehling, H. J. (1997). Structure and function of the pre-T cell receptor. Annu Rev Immunol 15, 433-452. White, L. C., Wright, K. L., Felix, N. J., Ruffner, H., Reis, L. F., Pine, R. & Ting, J. P. (1996). Regulation of LMP2 and TAP1 genes by IRF-1 explains the paucity of CD8+ T cells in IRF-1-/- mice. Immunity 5, 365-376. 38 Xu, X., Han, J., Ito, Y., Bringas, P., Jr., Urata, M. M. & Chai, Y. (2006). Cell autonomous requirement for Tgfbr2 in the disappearance of medial edge epithelium during palatal fusion. Dev Biol 297, 238-248. Xu, Y., Davidson, L., Alt, F. W. & Baltimore, D. (1996). Function of the pre-T-cell receptor alpha chain in T-cell development and allelic exclusion at the T-cell receptor beta locus. Proc Natl Acad Sci U S A 93, 2169-2173. Yamazaki, H., Sakata, E., Yamane, T., Yanagisawa, A., Abe, K., Yamamura, K., Hayashi, S. & Kunisada, T. (2005). Presence and distribution of neural crest-derived cells in the murine developing thymus and their potential for differentiation. Int Immunol 17, 549-558. Yanai, H., Chen, H. M., Inuzuka, T., Kondo, S., Mak, T. W., Takaoka, A., Honda, K. & Taniguchi, T. (2007). Role of IFN regulatory factor 5 transcription factor in antiviral immunity and tumor suppression. Proc Natl Acad Sci U S A 104, 3402-3407. Yang, D., Thangaraju, M., Browning, D. D., Dong, Z., Korchin, B., Lev, D. C., Ganapathy, V. & Liu, K. (2007a). IFN regulatory factor 8 mediates apoptosis in nonhemopoietic tumor cells via regulation of Fas expression. J Immunol 179, 47754782. Yang, D., Thangaraju, M., Greeneltch, K. & other authors (2007b). Repression of IFN regulatory factor 8 by DNA methylation is a molecular determinant of apoptotic resistance and metastatic phenotype in metastatic tumor cells. Cancer Res 67, 33013309. Yim, J. H., Ro, S. H., Lowney, J. K., Wu, S. J., Connett, J. & Doherty, G. M. (2003). The role of interferon regulatory factor-1 and interferon regulatory factor-2 in IFNgamma growth inhibition of human breast carcinoma cell lines. J Interferon Cytokine Res 23, 501-511. Yoneyama, M., Suhara, W., Fukuhara, Y., Fukuda, M., Nishida, E. & Fujita, T. (1998). Direct triggering of the type I interferon system by virus infection: activation of a transcription factor complex containing IRF-3 and CBP/p300. EMBO J 17, 1087-1095. Zhang, X., Tao, Y., Troiani, L. & Markovic-Plese, S. (2011). Simvastatin inhibits IFN regulatory factor 4 expression and Th17 cell differentiation in CD4+ T cells derived from patients with multiple sclerosis. J Immunol 187, 3431-3437. Zhao, J., Kong, H. J., Li, H. & other authors (2006). IRF-8/interferon (IFN) consensus sequence-binding protein is involved in Toll-like receptor (TLR) signaling and contributes to the cross-talk between TLR and IFN-gamma signaling pathways. J Biol Chem 281, 10073-10080. 39 Zheng, Y., Chaudhry, A., Kas, A. & other authors (2009). Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T(H)2 responses. Nature 458, 351-356. Zucchero, T. M., Cooper, M. E., Maher, B. S. & other authors (2004). Interferon regulatory factor 6 (IRF6) gene variants and the risk of isolated cleft lip or palate. N Engl J Med 351, 769-780. 40 CHAPTER TWO Meta-analysis of hematopoietic expression profiles reveals selective Irf6 expression in stem cells and in T-cells 41 Abstract The IRF family of transcription factors regulates critical immune functions, including development and differentiation of progenitors and commitment of effecter cells. IRF6 regulates cutaneous, craniofacial and limb development. Here, we used publically available microarray data to uncover a dynamic expression pattern for Irf6 during hematopoietic development and functional commitment. We found that Irf6 is expressed early in hematopoiesis, especially in long term hematopoietic stem cells making Irf6 a target for further investigation in long term engraftment studies. Consistent with a role in differentiation, we found abrupt attenuation of Irf6 expression in hematopoietic lineage committed progenitors. Moreover, we observed an age dependent increase of expression in common myeloid progenitor and myeloid erythroid progenitor. Also we identified Irf6 expression in T cell lineage, including developing and functionally committed stages. Thymic expression of Irf6 in double positive and single positive thymocytes suggests a possible role in T cell development. We found high Irf6 expression in naïve T cells with persistence in functionally committed T cell subsets except Th1. Considering co-expression and dynamic changes of Irf6 with other Irf family members, this work suggests a complex, interwoven network that regulates T cell lineage. This data further highlights the importance of developing an animal model to study Irf6 function in hematopoiesis. 42 Introduction Haematopoiesis is the process of production of all blood cells from a common pluripotent stem cell. Haematopoietic stem cells (HSC) differentiate into both myeloid and lymphoid lineages. While the myeloid lineages gives rise to erythrocytes, platelets, granulocytes, macrophages, and dendritic cells, the lymphoid lineages gives rise to Tcells, B-cells, and NK-cells. T-cells are the most abundant subset of blood lymphocytes and serve as the core of the adaptive immune response. Changes in T-cell number or function can lead to autoimmune diseases, immune deficiency, inflammatory disorders and cancer (Janeway et al., 2001; Paul, 2008 ). The interferon regulatory factor (IRF) family of transcription factors has nine members in mammals (Huang et al., 2010; Nehyba et al., 2009). All family members share a highly conserved N-terminal DNA-binding domain (DBD) and a less conserved C-terminal protein-binding domain (Kondo et al., 2002; Lohoff & Mak, 2005; Tamura et al., 2008). The functions of IRFs in the hematopoietic and immune system are indispensable. IRFs perform their functions by either intrinsic expression in their target cells (Table 2.1) or indirectly by influencing the environment of these cells (Tamura et al., 2008). Intrinsic expression of IRFs are usually constitutive in haematopoietic cells however it can be further induced or activated by external signals (Tamura et al., 2008). Common and rare IRF6 variants cause and contribute risk toward craniofacial defects. While rare variants lead to Van der Woude and Popliteal Pterygium Syndrome (Kondo et al., 2002), common IRF6 variants contribute 12% of orofacial clefting risk (Zucherro et al., 2004). The Irf6 knockout mouse revealed its role in craniofacial and limb development. Regulation of keratinocyte proliferation and differentiation contributes 43 to the cutaneous defects. However perinatal mortality hinders our ability to study the role of Irf6 in the haematopoietic system (Ingraham et al., 2006; Richardson et al., 2006; Richardson et al., 2009; Thomason et al., 2010). In 2005, Lohoff and Mak wondered if IRF6 is even expressed by haematopoietic cells (Lohoff & Mak, 2005). Shitao Li at al 2011 performed a proteomic study to define the candidate protein complexes involved in regulating interferon type I. Interestingly, affinity purification identified protein complex formation between IRF6/IRF5 and IRF6/IRF8, the latter was further confirmed by coimmunoprecipitation in HEK293 cells (Li et al., 2011). Proteinprotein interactions between IRF6 and more canonically described immune IRF family members, DNA conservation of the DBD and structural homology with IRF5 strongly suggests a role for Irf6 in the haematopoietic system. Haematopoietic system is characterized by having different cell lineages each one of them goes through several developmental stages. Many of the developmental stages have rare frequencies and require several markers or cumbersome procedures for isolation. Bench work required for assessment of expression for a given gene throughout the whole haematopoietic system is devastating. In the current era with the explosion of the publically available whole transcriptomic data for almost every cell type, bioinformatic analysis is a very promising approach to replace the classical tedious techniques. In this study, we are trying to profile the expression of Irf6 in different haematopoietic cells to predict the stages likely to be affected by its deficiency. Also we are identifying the pattern of expression of all other family members in the same cell types to identify the possible genetic interactions which are commonly seen between different members of Irf family. 44 IRF Intrinsic function in hematopoietic cells IRF1 * Required for CD8α+ (Penninger et al., 1997) and suppress pDCs (Gabriele et al., 2006) * Development of myeloid lineage (Testa et al., 2004) * Required for macrophage functions (Blanco et al., 2000; Brien et al., 2011) * Induction of NK cell-mediated cytotoxicity (Duncan et al., 1996) * Regulation of thymocyte development (Simon et al., 1997) * Induce Th1 differentiation (Kano et al., 2008; Liu et al., 2003; Lohoff et al., 1997; Taki et al., 1997). * Suppress IL4 production by Th2 (Elser et al., 2002) * Suppress Treg cells (Fragale et al., 2008) * Differentiation of CD8+ T cells (Brien et al., 2011) IRF2 * Self-renewal HSCs (Sato et al., 2009) * Development of epidermal and CD4+ DCs (Ichikawa et al., 2004) * Suppresses basophil expansion (Hida et al., 2005) * Megakaryocytic differentiation (Stellacci et al., 2004) * Regulates macrophage function (Cuesta et al., 2007; Salkowski et al., 1999) * Development of NK cells (Lohoff et al., 2000; Taki et al., 2005) * Regulation of thymocyte development (Simon et al., 1997) * Suppress IL4 production by Th2 (Elser et al., 2002; Zheng et al., 2009) * Possible oncogenic effect in leukemic cells (Passioura et al., 2005) * Suppression of CD8+ CTL activity (Hida et al., 2000) Table 2.1: Effects of IRFs intrinsic expression in hematopoietic cells 45 Table 2.1 (cont’d) IRF3 * Required for apoptosis of macrophages (Hsu et al., 2004) * Induction of IFN-β in stimulated DCs (Kim et al., 2014; Sakaguchi et al., 2003; Sato et al., 2000) * Prostaglandin E2 production in LPS-primed monocyte (Endo et al., 2014) IRF4 * Development of CD11bhi CD8α- DCs (Suzuki et al., 2004) and CD4+ DCs (Tamura et al., 2005a) * Induce IL4 production by Th2 (Hu et al., 2002; Lohoff et al., 2002; Rengarajan et al., 2002) * Th17 differentiation (Brustle et al., 2007; Chen et al., 2008; Zhang et al., 2011) * B cell development (Lu et al., 2003; Ma et al., 2006) * Plasma cell differentiation (Klein et al., 2006; Sciammas et al., 2006) * Oncogenic effect in multiple myeloma (Iida et al., 1997; Shaffer et al., 2008) & CLL (Ito et al., 2002; Tsuboi et al., 2000) * Tumor suppressor in early B-cell development (Acquaviva et al., 2008; Pathak et al., 2011) & myeloid transformation (Jo & Ren, 2011) IRF5 * Production of type I IFNs and IL6 in macrophages after viral infection (Yanai et al., 2007) and pDCs after CpG-A stimulation (Yasuda et al., 2007). * Required in apoptosis in DCs (Couzinet et al., 2008) * Monocytes and B-cells from Irf5-/- mice have an intrinsic defect in their response to pristane-induced lupus (Savitsky et al., 2010; Stone et al., 2012; Yang et al., 2012) 46 Table 2.1 (cont’d) IRF6 No intrinsic functions identified in hematopoietic cells IRF7 * Induction of type I IFN in stimulated DCs and macrophage (Honda et al., 2005; Hsieh et al., 2014; Kim et al., 2013; Tamura et al., 2008) * Control monocyte differentiation to macrophage (Lu & Pitha, 2001) * Induction of IL33 in monocytes and macrophages (Sun et al., 2014) * Contradictory effect on antiviral response of CD8 T cells (Gracias et al., 2013; Li et al., 2013; Zhou et al., 2013) IRF8 * Development of CD8α+ DCs (Schiavoni et al., 2002) and pDCs (Becker et al., 2012; Tamura et al., 2005a) * Development and trafficking of Langerhans cells and dermal DCs (Schiavoni et al., 2004) * Macrophage development (Tamura et al., 2005b) * Suppression of neutrophil production (Becker et al., 2012) * B cell development (Lee et al., 2006; Lu et al., 2003; Ma et al., 2006) * Knock out is associated with CML-like disease (Holtschke et al., 1996) * Suppress Th17 differentiation (Ouyang et al., 2011; Tian et al., 2012). IRF9 * Regulation of B cell activity and isotype switch in response to self antigen (Thibault et al., 2008) * Modulation of IFN-I and IFN-II responsiveness in macrophages (Farlik et al., 2012; Weiden et al., 2000) 47 Materials and methods Gene Expression Omnibus repository (www.ncbi.nlm.nih.gov/geo/), Gene Expression Atlas (www.ebi.ac.uk/gxa/), and the murine haematopoietic data base Blood Express (Miranda-Saavedra et al., 2009) are publically available repositories of microarray experiments. I searched for experiments covering one or more mouse developmental hematopoietic lineages. Raw data of each experiment was analyzed independently using R computing environment (http://www.r-project.org/). The probe intensities were subjected to background correction and quantile normalization using Robust Multi-array Average algorithm (RMA) (Irizarry et al., 2003). Probe numbers for Irf genes were identified in the tested microarray platforms using the BioMart ID conversion tool (Kasprzyk, 2011). Relative expression of Irf genes was plotted for each experiment. To ensure the reliability of comparison, average expression ranks of Irf genes were calculated for every cell type per experiment. To study the co-expression pattern, expression ranks of Irf genes in all experiments were pooled and clustered using “gplots” package in R computing environment. Four Affymatrix (Santa Clara, California, USA) microarray platforms were used in the studied experiments; Murine Genome U74Av2 [MG_U74Av2], Mouse Genome 430 2.0 [Mouse430_2], Mouse Expression 430A [MOE430A], and Mouse Gene 1.0 ST Array [MoGene1]. Studied experiments covered the expression profiles of hematopoietic stem cells (HSC) and its two main subpopulations; long term hematopoietic stem cells (LTHSC) and short term hematopoietic stem cells (ST-HSC). Several lineage-committed precursors were also compiled, including lymphoid multipotent progenitors (LMPP), common myeloid progenitors (CMP), common lymphoid progenitors (CLP), 48 megakaryocyte-erythroid progenitor (MEP) and granulocyte monocyte progenitors (GMP). Lymphocyte lineages were heavily covered during early development and after maturation in resting and activated conditions. Early developmental stages included CD4+ CD8+ double negative thymocytes (DN), CD4- CD8- double positive thymocytes (DP), B cell-biased lymphoid progenitor (BLP), and pre-ProB cells. Also mature splenic T cells and B cells and nature killer cells (NK) were tested. Resting T cells were identified by being negative for B220 marker and resting B cells were indentified by being negative for CD43 marker. Also naïve and activated T cells with its main subpopulations; T helper and T cytotoxic were analyzed. Functionally committed T helper subpopulations were covered including T-helper 1 (Th1), T-helper 2 (Th2), Thelper 17 (Th17), induced regulatory T cells (iTreg) and natural regulatory T cell (nTreg). BM precursors of myeloid linage were also covered like promyelocytes and myelocytes. Other tested terminally differentiated cells included dendritic cells (DCs), plasmacytoid dendritic cells (pDC), thymic dendritic cells (tDC), monocytes, bone marrow macrophages (BM.MPh), thymic macrophages (tMPh), granulocytes, nucleated erythrocytes (Nu RBCs), and precursor and mature mast cells. Whole thymocytes as well as cortical and modularly thymic epithelial cells (cTEC,mTEC) were included. 49 Results Several members of the IRF family of transcription factors are essential for hematopoeisis. However, there are no data on IRF6 because mice that lack Irf6 die shortly after birth from abnormal morphogenesis. As a first step in identifying a potential function for IRF6 in hematopoeisis, we performed a meta analysis on gene expression profiles in murine hematopoietic tissues. We identified 20 such studies (Table 2.2) that included 241 microarrays from 50 unique hematopoietic cell types. Publication lineages Affymetrix Chip LT-HSC, NK, Naïve & activated CD4 & CD8 T-cells, B-cells, (Chambers et al., 2007) Mouse Genome 430-2 Monocytes, Granulocytes, Nu. Erythrocytes Resting and activated T cells and (Holwerda et al., 2013) Mouse Gene 1.0 ST B cell (Derbinski et al., 2005) Thymic stromal cells Murine Genome U74Av2 DC, B cells, CD4 T cells. CD8 T (Dudziak et al., 2007) Mouse Genome 430-2 cells (Lin et al., 2014) Th1,Th2, Th17 Mouse Gene 1.0 ST (Ficara et al., 2008) LT-HSC, ST-HSC Mouse Genome 430-2 mast cell precursors and mature (Haddon et al., 2009) Mouse Expression 430A mast cells Table 2.2: Mouse expression profiles for hematopoietic cell types. 50 Table 2.2 (Cont’d) (Mansson et al., 2007) HSC Mouse Genome 430-2 (Robbins et al., 2008) DCs, NK, B cells, CD8 T cells Mouse Genome 430-2 (Rodriguez et al., 2007) Th1 vs Th2 Mouse Genome 430-2 (Tothova et al., 2007) HSC, MLP Mouse Genome 430-2 LT-HSC, ST-HSC, MPP Mouse Genome 430-2 (Vigano et al., 2013) HSC, pro Tcells, DP T cells Mouse Gene 1.0 ST (Wang et al., 2010) HSC vs CMP and GMP Mouse Genome 430-2 (Venkatraman et al., 2013) Naïve T-helper, Th1, Th2, Th17, (Wei et al., 2009) Mouse Genome 430-2 iTreg, nTreg (Weischenfeldt et al., macrophage vs T-cells Mouse Genome 430-2 2008) LT-HSC,ST-HSC, LMPP, CMP, (Beerman et al., 2014) Mouse Genome 430-2 CLP, GMP, MEP, BLP, pre-ProB HSC, Promyelocytes, Myelocytes, (Wong et al., 2014) Mouse Gene 1.0 ST Granulocytes (Egawa & Littman, 2011) DN, DP, CD4SP, CD8SP Mouse Genome 430-2 (Kawazu et al., 2007) Mouse Genome 430-2 DN cells 51 Plotting of non-logarithmic normalized data for each study allowed us to compare the level of expression for all 9 Irf genes in 50 mouse hematopoietic cells types (Supplementary Figures A1 - A12). However, testing the reproducibility across experiments, following the expression changes along the haematopoietic tree, and prediction of possible family member interactions required a more integrative approach. To facilitate inter-experimental comparison of expression of Irf genes, we calculated and ranked the relative expression for each Irf gene in each cell type per experiment (Supplementary table A1). We then performed cluster analysis for the average rank expression of Irf genes (figure 2.1). We observed two main patterns of differential expression for family members across the studied cell lineages. First, Irf1, Irf3 and Irf9 have consistently high expression levels in most studied cell lineages with average rank expressions of 93, 83 and 84, respectively. All other family members showed more variable expression among different cell types. Irf6 was expressed mainly in two stages of hematopoietic development. Early in development, Irf6 was expressed in HSC and their immediate downstream progeny LMPP. Irf6 expression was maintained in CLP with apparent down regulation in all myeloid capable progenitors (CMP, MEP, and GMP). However, suppression of Irf6 expression in CMP and MEP was lost in progenitors obtained from 2 year-old mice (figure 2.2). Regulation of Irf6 expression was also observed in development of T cells. In two out of three studies, Irf6 expression was barely detectable in early developing DN thymocytes. We found an increase of Irf6 expression in DP thymocytes and even more in single positive CD4 and CD8 progeny. Expression of Irf6 peaked to exceed 80% in naïve T-helper and cytotoxic populations. No changes in Irf6 expression were seen in activated T-helper cells, however the rank 52 expression profile lost about 10 percentiles in activated T-cytotoxic cells. Terminally differentiated T-helper sub-populations maintained a 63-71% expression rank except TH-1, where Irf6 rank of expression went down to the 30th percentile for 2 out of 3 ranked studies. Finally, the thymic-derived natural Treg came at the 79th percentile on the rank of expression (Figure 2.3). 53 L M PP,M a n s s o n 2 0 0 7 L M PP,Be e rm a n 2 0 1 4 L M PP.Ol d _ m i c e ,Be e rm a n 2 0 1 4 L M PP,Ve n k a tra m a n 2 0 1 3 ST-HSC,Fi c a ra 2 0 0 8 ST-HSC,Ve n k a tra m a n 2 0 1 3 CL P,Be e rm a n 2 0 1 4 CL P.Ol d _ m i c e ,Be e rm a n 2 01 4 CM P.Ol d _ m i c e ,Be e rm a n 2 0 1 4 HSC,Vi g a n o 2 0 1 3 HSC,Wo n g 2 0 1 4 Pro m y e l o c y te s ,Wo n g 2 0 1 4 Gra n u l o c y te s ,Wo n g 2 0 1 4 M y e l o c y te s ,Wo n g 2 0 1 4 M o n o c y te ,Ch a m b e rs 2 0 0 7 CM P,To th o v a 2 0 0 7 BL P.Ol d _ m i c e ,Be e rm a n 2 01 4 BL P,Be e rm a n 2 0 1 4 tM Ph ,De rb i n s k i 2 0 0 5 p re -Pro B.Ol d _ m i c e ,Be e rm an 2 0 1 4 p re -Pro B,Be e rm a n 2 0 1 4 Th 1 ,We i 2 0 0 9 NK,Ro b b i n s 2 0 0 8 NK,Ch a m b e rs 2 0 0 7 CD1 1 c +CD8 +DCs ,Du d z i a k 2 0 0 7 BM .M Ph ,We i s c h e n fe l d t 2 00 8 CD8 +DCs ,Ro b b i n s 2 0 0 8 p DCs ,Ro b b i n s 2 0 0 8 CD1 1 b +DCs ,Ro b b i n s 2 0 0 8 tDC,De rb i n s k i 2 0 0 5 B-Ce l l ,Ro b b i n s 2 0 0 8 B-Ce l l ,Du d z i a k 2 0 0 7 Re s ti n g B-Ce l l ,Ho l we rd a 2 01 3 Ac t. B-Ce l l ,Ho l we rd a 2 0 1 3 B-Ce l l ,Ch a m b e rs 2 0 0 7 Th 1 ,Ro d ri g u e z 2 0 0 7 CD1 1 c +CD8 -DCs ,Du d z i a k 20 0 7 Re s ti n g T-Ce l l ,Ho l we rd a 2 01 3 DN th y m o c y te s ,Vi g a n o 2 0 1 3 DP th y m o c y te s ,Vi g a n o 2 0 1 3 Gra n u l o c y te s ,Ch a m b e rs 2 0 0 7 Sp l e n i c T-c y to ,Ro b b i n s 2 00 8 Nu .RBCs ,Ch a m b e rs 2 0 0 7 L T-HSC,Ve n k a tra m a n 2 0 1 3 Wh o l e .Th y m u s ,We i s c h e n fe l d t 2 0 0 8 L T-HSC.Fe ta l _ L i v e r,Be e rm a n 2 0 1 4 n Tre g ,We i 2 0 0 9 Na ïv e T-h e l p e r,We i 2 0 0 9 Na ïv e T-h e l p e r,Ch a m b e rs 2 0 0 7 Na ïv e T-c y to ,Ch a m b e rs 2 00 7 L T-HSC,Ch a m b e rs 2 0 0 7 HSC,To th o v a 2 0 0 7 L T-HSC,M a n s s o n 2 0 0 7 L T-HSC,Be e rm a n 2 0 1 4 L T-HSC.Ol d _ m i c e ,Be e rm a n 2 0 1 4 ST-HSC,M a n s s o n 2 0 0 7 ST-HSC,Be e rm a n 2 0 1 4 ST-HSC.Ol d _ m i c e ,Be e rm a n 2 0 1 4 CD4 SP th y m o c y te s ,Eg a wa 2 0 1 1 CD8 SP th y m o c y te s ,Eg a wa 2 0 1 1 Sp l e n i c T-h e l p e r,Du d z i a k 2 0 0 7 Sp l e n i c T-c y to ,Du d z i a k 2 00 7 i Tre g ,We i 2 0 0 9 L T-HSC,Fi c a ra 2 0 0 8 Th 2 ,Ro d ri g u e z 2 0 0 7 Ac t. T-h e l p e r,Ch a m b e rs 2 00 7 Ac t. T-c y to ,Ch a m b e rs 2 0 0 7 Th 1 7 ,We i 2 0 0 9 Th 2 ,We i 2 0 0 9 Ac t. T-Ce l l ,Ho l we rd a 2 0 1 3 Th 2 ,L i n 2 0 1 4 Th 1 7 ,L i n 2 0 1 4 Th 1 ,L i n 2 0 1 4 M EP.Ol d _ m i c e ,Be e rm a n 2 0 1 4 HSC,Wa n g 2 0 1 0 DP th y m o c y te s ,Eg a wa 2 0 11 DN th y m o c y te s ,Ka wa z u 2 00 7 DN th y m o c y te s ,Eg a wa 2 0 11 M EP,Be e rm a n 2 0 1 4 CM P,Be e rm a n 2 0 1 4 GM P,Be e rm a n 2 0 1 4 GM P.Ol d _ m i c e ,Be e rm a n 2 0 1 4 CM P,Wa n g 2 0 1 0 GM P,Wa n g 2 0 1 0 p re -m a s t.c e l l s ,Ha d d o n 2 0 0 9 m a s t.c e l l s ,Ha d d o n 2 0 0 9 Color Key Irf9 Irf3 Irf1 Irf8 Irf7 100 Irf5 80 Irf2 60 Irf4 40 Irf6 20 Figure 2.1: Clustering analysis of average Irf gene rank expression. 54 Figure 2.1 (Cont’d): Irf1, Irf3, and Irf9 show non-selective high rank of expression in most haematopoietic cell types. The other family members have high variability between cell types, especially Irf6 and Irf4. Irf6 expression is almost exclusively in early haematopoietic progenitors and T cell developmental and functional cells. 55 100 80 60 40 0 20 CLP.Old_mice,Beerman 2014 CLP,Beerman 2014 GMP.Old_mice,Beerman 2014 GMP,Beerman 2014 GMP,Wang 2010 MEP.Old_mice,Beerman 2014 MEP,Beerman 2014 CMP.Old_mice,Beerman 2014 CMP,Beerman 2014 CMP,Wang 2010 CMP,Tothova 2007 LMPP.Old_mice,Beerman 2014 LMPP,Beerman 2014 LMPP,Venkatraman 2013 LMPP,Mansson 2007 ST-HSC.Old_mice,Beerman 2014 ST-HSC,Beerman 2014 ST-HSC,Venkatraman 2013 ST-HSC,Ficara 2008 ST-HSC,Mansson 2007 LT-HSC.Old_mice,Beerman 2014 LT-HSC.Fetal_Liver,Beerman 2014 LT-HSC,Beerman 2014 LT-HSC,Venkatraman 2013 LT-HSC,Ficara 2008 LT-HSC,Mansson 2007 LT-HSC,Chambers 2007 HSC,Wong 2014 HSC,Vigano 2013 HSC,Wang 2010 HSC,Tothova 2007 Figure 2.2: Average Irf6 rank expression in early hematopoietic progenitors. Every bar represents the Irf6 rank of expression in a given microarray. The horizontal axis represents the percentile rank while the vertical axis shows the names of the studied cell type appended to the names of first author and year of publication. Cells are arranged from the most immature (at the origin of the figure) to the mature stages. 56 100 80 60 40 20 0 nTreg,Wei 2009 iTreg,Wei 2009 Th17,Lin 2014 Th17,Wei 2009 Th2,Lin 2014 Th2,Wei 2009 Th2,Rodriguez 2007 Th1,Lin 2014 Th1,Wei 2009 Th1,Rodriguez 2007 Act. T-cyto,Chambers 2007 Act. T-helper,Chambers 2007 Naïve T-cyto,Chambers 2007 Naïve T-helper,Wei 2009 Naïve T-helper,Chambers 2007 Splenic T-cyto,Robbins 2008 Splenic T-cyto,Dudziak 2007 Splenic T-helper,Dudziak 2007 Act. T-Cell,Holwerda 2013 Resting T-Cell,Holwerda 2013 CD8SP thymocytes,Egawa 2011 CD4SP thymocytes,Egawa 2011 DP thymocytes,Vigano 2013 DP thymocytes,Egawa 2011 DN thymocytes,Vigano 2013 DN thymocytes,Egawa 2011 DN thymocytes,Kawazu 2007 Figure 2.3: Average Irf6 rank expression in T cell stages. Every bar represents the Irf6 rank of expression in a given microarray. The horizontal axis represents the percentile rank while the vertical axis shows the names of the studied cell type appended to the names of first author and year of publication. Cells are arranged from the most immature (at the origin of the figure) to the mature stages. 57 To assess a possible bias of probes across arrays, biological replicates done using different microarray platforms were identified. Average rank expression was calculated for each platform per gene. Paired t-test analysis showed significant bias for Irf1 and Irf2 but insignificant changes for the other Irf genes (Table 2.3). Irf1 Mouse Mo430-2 Gene1 HSC 92 86 DN 95 91 DP 87 85 Th1 95 68 Th2 93 74 Th17 93 66 p-value = 0.030* Irf2 Mouse Mo430-2 Gene1 HSC 76 90 DN 57 93 DP 58 93 Th1 79 88 Th2 76 88 Th17 79 78 p-value = 0.034* Mouse Mo430-2 Gene1 HSC 73 77 DN 81 78 DP 84 83 Th1 80 79 Th2 76 82 Th17 83 82 p-value = 0.661 Irf4 MoMouse 430-2 Gene1 HSC 52 35 DN 39 57 DP 35 53 Th1 86 96 Th2 90 95 Th17 86 97 p-value = 0.217 Irf5 Mouse Mo430-2 Gene1 HSC 59 72 DN 56 75 DP 46 55 Th1 73 66 Th2 72 65 Th17 64 66 p-value = 0.319 MoMouse 430-2 Gene1 HSC 61 53 DN 19 59 DP 51 64 Th1 29 63 Th2 70 62 Th17 71 63 p-value = 0.298 Irf7 Irf8 Mouse Mo430-2 Gene1 HSC 70 86 DN 70 78 DP 44 76 Th1 86 90 Th2 79 77 Th17 79 76 p-value = 0.149 Mouse Mo430-2 Gene1 HSC 56 72 DN 71 74 DP 63 76 Th1 72 37 Th2 62 37 Th17 72 37 p-value = 0.330 Irf3 Irf6 Irf9 Mouse Mo430-2 Gene1 HSC 85 83 DN 76 85 DP 69 88 Th1 85 61 Th2 80 60 Th17 86 60 p-value = 0.384 Table 2.3: Average rank expression for microarray platforms per gene 58 Discussion In this study, we uncovered a novel expression pattern for Irf6 in murine hematopoietic cell lineages. Our results showed that Irf6 expression is selectively expressed in hematopoietic cell progenitors and terminally differentiated cells. We found that hematopoietic stem cells and T cells are the two main developmental windows where Irf6 expression was evident. Highest expression of Irf6 is seen in LT-HSC which requires further investigation for a possible role of IRF6 in long term BM engraftment. Interferons directly stimulate HSC proliferation and differentiation (Schuettpelz & Link, 2013). Intrinsic expression of Irf2 was shown to preserve the self-renewal and multilineage differentiation capacity of HSCs by suppression of INF-I signaling (Sato et al., 2009). Our meta-analysis showed that expression of both Irf2 and Irf6 were very comparable in HSCs and Irf6 was even higher in the long term compartment. Loss of Irf6 expression in more differentiated myeloid progenitors with persistent expression in CLP suggested an important role in the lineage commitment of these critical progenitor cells (Figure 2.1). Regulation of early lineage commitment is one of the documented functions in the family. For example, Irf8 promotes DC lineage commitment over neutrophil production in myeloid progenitors (Becker et al., 2012) while Irf4 favors macrophage commitment (Yamamoto et al., 2011). Recently, age dependent increase of activated Irf3 was shown to promote the inflammatory response in aging kidney cells (Xi et al., 2014). The relative increase of Irf6 in CMP and MEP obtained from old mice might have a similar effect on these progenitors. 59 In T cells, the expression was evident during thymic development, T-cell activation and terminal functional commitment (Figure 2.2). A possible role of Irf6 in T cell development can be predicted from intrinsic expression in evolving thymocytes as well as expression in cortical and medullary thymic epithelium. Persistent Irf6 expression in activated and functionally committed T cell subsets except Th1 suggests a potential role for Irf6 in functional commitment and terminal differentiation of T cells. Variability of expression in the same cell type among different experiments was sometimes striking. Two obvious examples were Irf6 expression in DN thymocytes and Th1 cells, where 2 out of 3 experiments show very low rank of expression (< 30%) while 3rd experiment is uniquely high (> 60%). One possible explanation is a difference between probe sensitivities across the platforms. However comparing all replicates using the two commonly used platforms (Mouse430_2 & MoGene1), we failed to observe a consistent bias. Another possible explanation is that DN cells are a rare population and can be easily contaminated by thymocytes with low expression of CD4 and CD8 (Lucas & Germain, 1996). Finally, induction of Th1 cell in vitro is difficult without contamination from other functional subsets. These biological obstacles may explain the high variability we see in our analysis. Non-linear noise signature of microarray experiments is another important factor that always should be considered in these meta-analysis studies (Chen et al., 2011; Leek et al., 2010). Publically available expression profile data sets are an underutilized tool that can help researchers to understand the expression pattern of their target genes as well as possible interactions with other co-expressed genes. Transcriptional expression of Irf members is constitutive and IFN-inducible in most targets tissues (Tamura et al., 2008). 60 IRFs are considered master regulators of hematopoietic development and differentiation. Until now, the expression pattern of Irf6 in haematopoiesis has not been studied. Also, perinatal lethality of Irf6 knockout mouse hindered efforts to study its function in haematopoiesis. We found that Irf6 expression is selectively expressed early HSC and in various T-cell lineages, including Th1, Th2 and Th17. This work strongly supports a regulatory function of Irf6 in haematopoiesis. 61 APPENDIX 62 LT-HSC.Fetal_Liver Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 LT-HSC LT-HSC.Old_mice ST-HSC ST-HSC.Old_mice LMPP LMPP.Old_mice CMP CMP.Old_mice MEP MEP.Old_mice GMP GMP.Old_mice CLP CLP.Old_mice BLP BLP.Old_mice pre-ProB 8000 6000 4000 2000 0 pre-ProB.Old_mice Figure A1: Expression of Irf genes in immature hematopoietic progenitors at 3 months and 2 years of mouse age. Expression data obtained from Beerman et al (2014). 63 Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 HSC Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 LT-HSC 1500 1000 500 0 1400 1200 1000 800 600 400 200 ST-HSC 0 CMP Figure A2a: Expression of Irf genes in Figure A2b: Expression of Irf genes in immature haematopoietic progenitors. immature haematopoietic progenitors. Expression data obtained from Tothova et Expression data obtained from Ficara et al al 2007. 2008. 64 Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 LT-HSC Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 LT-HSC ST-HSC ST-HSC 8000 6000 4000 0 600 400 200 0 2000 LMPP LMPP Figure A2c: Expression of Irf genes in Figure A2d: Expression of Irf genes in immature haematopoietic progenitors. immature haematopoietic progenitors. Expression data obtained from Mansson et Expression data obtained from al 2007. Venkatraman et al 2013. 65 Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 HSC Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 HSC Promyelocytes CMP Myelocytes GMP 800 600 400 200 0 1400 1200 1000 800 600 400 200 0 Granulocytes Figure A3: Expression of Irf genes in Figure A4: Expression of Irf genes early myeloid development. Expression during granulocytes development. data obtained from Wang et al 2010. Expression data obtained from Wong et al 2014. 66 Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 HSC Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 DN thymocytes DP thymocytes DN Thymocytes CD4SP thymocytes DP Thymocytes 1500 1000 500 0 2000 1500 1000 500 0 CD8SP thymocytes Figure A5a: Expression of Irf genes in Figure A5b: Expression of Irf genes in thymocyte development. Expression data thymocyte development. Expression data obtained from Vigano et al 2013. obtained from Egawa et al 2011. 67 Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 LT-HSC NK Naïve T-helper Naïve T-cyto Act.T-helper Act.T-cyto B-Cell Monocyte Granulocytes 10000 8000 6000 4000 2000 0 Nu.RBCs Figure A6: Expression of Irf genes in LT-HSC versus terminally differentiated haematopoietic cells. Expression data obtained from Chambers et al 2007. 68 Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 CD11c+CD8-DCs CD11c+CD8+DCs Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 CD8+DCs CD11b+DCs pDCs B-cells NK Splenic T-helper B-Cell Splenic T-cyto 8000 6000 4000 2000 0 7000 6000 5000 4000 3000 2000 1000 0 Splenic T-cyto Figure A7a: Expression of Irf genes in Figure A7b: Expression of Irf genes in lymphocytes and DCs. Expression data lymphocytes and DCs. Expression data obtained from Dudziak et al 2007. obtained from Robbins et al 2008. 69 Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 Resting B-Cell Act.B-Cell Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 pre-mast.cells Resting T-Cell mast.cells 120 100 80 60 40 20 0 1000 800 600 400 200 0 Act.T-Cell Figure A8: Expression of Irf genes in Figure A9: Expression of Irf genes in resting and activated lymphocytes. mast cells and pre-mast cells. Expression data obtained from Holwerda Expression data obtained from Haddon et et al 2013. al 2009. 70 Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 Th1 Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 TH1 TH2 Th2 1000 800 600 400 200 0 1400 1200 1000 800 600 400 200 0 TH17 Figure A10a: Expression of Irf genes Figure A10b: Expression of Irf genes in T- in T-helper subsets. Expression data helper subsets. Expression data obtained obtained from Rodriguez et al 2007 from Lin et al 2014. 71 Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 Naïve T-helper Th1 Th2 Th17 iTreg 4000 3000 2000 1000 0 nTreg Figure A10c: Expression of Irf genes in T-helper subsets. Expression data obtained from Wei et al 2009. 72 Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 BM.MPh Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 Irf9 cTEC mTEC tDC Whole.Thymus 20000 15000 10000 5000 3000 2500 2000 1500 1000 500 0 0 tMPh Figure A11: Expression of Irf genes in Figure A12: Expression of Irf genes in BM macrophage and whole thymus non-lymphocyte thymic cells. suspension. Expression data obtained Expression data obtained from Derbinski from Weischenfeldt et al 2008. et al 2005 (No probes for Irf2). 73 Author Chip type Cell Irf1 Irf2 Irf3 Irf4 Irf5 Irf6 Irf7 Irf8 and year lineage Tothova et al Mouse430_2 HSC 95 98 73 76 65 81 53 58 2007 Wang et al Mouse430_2 HSC 89 55 74 28 54 41 60 83 2010 Vigano et al MoGene1 HSC 81 91 78 36 73 53 70 89 2013 Wong et al MoGene1 HSC 91 90 76 34 71 54 75 83 2014 Chambers et Mouse430_2 LT-HSC 96 83 87 65 64 98 64 65 al 2007 Mansson et al Mouse430_2 LT-HSC 98 70 86 26 74 90 75 62 2007 Ficara et al Mouse430_2 LT-HSC 98 70 86 98 83 66 76 84 2008 Venkatraman Mouse430_2 LT-HSC 94 60 86 59 60 64 69 70 et al 2013 Beerman et al Mouse430_2 LT-HSC 96 78 85 32 70 86 68 62 2014 Beerman et al Mouse430_2 LT-HSC 94 66 81 47 68 77 64 73 2014 (Fetal Liver) Beerman et al Mouse430_2 LT-HSC 98 65 86 29 66 85 75 53 2014 (Old mice) Mansson et al Mouse430_2 ST-HSC 96 73 86 34 71 71 71 70 2007 Ficara et al Mouse430_2 ST-HSC 94 73 84 49 65 60 68 79 2008 Venkatraman Mouse430_2 ST-HSC 96 63 86 39 63 56 65 85 et al 2013 Beerman et al Mouse430_2 ST-HSC 95 73 84 28 71 70 66 67 2014 Beerman et al Mouse430_2 ST-HSC 96 73 90 26 70 71 73 58 2014 (Old mice) Mansson et al Mouse430_2 LMPP 97 72 85 34 72 64 74 94 2007 Venkatraman Mouse430_2 LMPP 97 71 87 45 65 55 74 90 et al 2013 Beerman et al Mouse430_2 LMPP 95 72 86 32 77 68 73 86 2014 Beerman et al Mouse430_2 LMPP 96 71 87 38 63 69 70 88 2014 (Old mice) Tothova et al Mouse430_2 CMP 89 94 75 48 61 30 46 74 2007 Table A1: Average rank of IRFs expression for each cell type per experiment 74 Irf9 87 84 85 81 76 85 78 75 86 78 90 80 82 71 83 89 85 78 85 88 82 Table A1 (Cont’d) Wang et al 2010 Beerman et al 2014 Beerman et al 2014 Beerman et al 2014 Beerman et al 2014 Wang et al 2010 Beerman et al 2014 Beerman et al 2014 Beerman et al 2014 Beerman et al 2014 Beerman et al 2014 Beerman et al 2014 Beerman et al 2014 Beerman et al 2014 Chambers et al 2007 Dudziak et al 2007 Robbins et al 2008 Holwerda et al 2013 Holwerda et al 2013 Weischenfeldt et al 2008 Derbinski et al 2005 Derbinski et al 2005 Mouse430_2 CMP 85 44 73 28 50 25 50 92 78 Mouse430_2 CMP 95 60 83 29 72 26 69 98 82 Mouse430_2 CMP (Old mice) MEP 93 69 86 27 60 47 65 94 87 83 47 81 35 60 72 52 79 88 71 87 31 52 44 71 74 87 Mouse430_2 MEP (Old mice) GMP 83 34 71 28 53 23 49 92 72 Mouse430_2 GMP 93 47 81 33 77 13 64 97 75 Mouse430_2 GMP (Old mice) CLP 93 64 86 24 61 14 57 95 80 95 71 86 42 75 47 73 99 86 CLP (Old mice) BLP 96 80 85 45 66 45 71 96 89 95 70 87 57 77 35 72 99 85 BLP (Old mice) pre-ProB 95 80 86 59 72 39 72 96 90 96 74 88 69 67 22 75 99 88 95 78 86 67 74 24 71 95 89 Mouse430_2 pre-ProB (Old mice) B-Cell 98 90 89 92 85 50 82 96 87 Mouse430_2 B-Cell 95 81 90 89 96 86 98 91 Mouse430_2 B-Cell 97 86 91 95 92 10 86 98 89 MoGene1 91 95 83 92 92 46 71 96 81 92 90 75 97 85 48 78 97 84 89 74 81 50 56 54 73 70 79 MG_U74Av2 Resting B-Cell Activated B-Cell Whole Thymus cTEC 98 NA 70 68 67 76 47 91 92 MG_U74Av2 mTEC 97 NA 64 97 80 95 65 93 94 Mouse430_2 Mouse430_2 Mouse430_2 Mouse430_2 Mouse430_2 Mouse430_2 Mouse430_2 Mouse430_2 MoGene1 Mouse430_2 75 9 9 Table A1 (Cont’d) Kawazu et al 2007 Egawa et al 2011 Vigano et al 2013 Egawa et al 2011 Vigano et al 2013 Egawa et al 2011 Egawa et al 2011 Holwerda et al 2013 Holwerda et al 2013 Dudziak et al 2007 Dudziak et al 2007 Robbins et al 2008 Chambers et al 2007 Wei et al 2009 Chambers et al 2007 Chambers et al 2007 Chambers et al 2007 Rodriguez et al 2007 Wei et al 2009 Lin et al 2014 Rodriguez et al 2007 Wei et al 2009 Lin et al 2014 Mouse430_2 DN thymocytes DN thymocytes DN thymocytes DP thymocytes DP thymocytes CD4SP thymocytes CD8SP thymocytes Resting T-Cell Activated T-Cell Splenic T-helper Splenic T-cytotoxic Splenic T-cytotxic Naïve T-helper Naïve T-helper Naïve T-cytotoxic Activated T-helper Activated T-cytotoxic Th1 98 64 82 38 54 16 76 70 76 91 51 81 40 58 23 67 70 76 91 93 78 57 75 59 74 78 85 87 58 84 35 46 51 63 44 69 85 93 83 53 55 64 76 76 88 97 69 85 70 45 73 94 49 95 97 72 85 49 49 71 94 49 93 95 92 84 65 65 57 81 81 79 93 87 76 95 58 62 59 82 76 97 68 90 61 58 54 91 61 92 98 65 90 52 58 38 93 59 91 98 84 86 51 63 56 87 71 89 99 79 85 64 56 93 85 87 88 98 82 91 69 56 70 90 70 93 99 81 86 60 61 83 88 87 88 98 72 83 97 62 82 69 94 84 98 64 84 97 61 72 73 95 81 92 78 80 96 73 31 64 89 80 Mouse430_2 Th1 97 80 80 77 72 26 81 82 90 MoGene1 Mouse430_2 Th1 Th2 68 93 88 79 79 96 66 63 37 74 96 82 69 58 90 86 61 80 Mouse430_2 Th2 92 72 78 83 63 71 66 73 80 MoGene1 Th2 74 88 82 95 65 62 37 77 60 Mouse430_2 Mouse430_2 MoGene1 Mouse430_2 MoGene1 Mouse430_2 Mouse430_2 MoGene1 MoGene1 Mouse430_2 Mouse430_2 Mouse430_2 Mouse430_2 Mouse430_2 Mouse430_2 Mouse430_2 Mouse430_2 76 Table A1 (Cont’d) Wei et al 2009 Lin et al 2014 Wei et al 2009 Wei et al 2009 Chambers et al 2007 Robbins et al 2008 Dudziak et al 2007 Dudziak et al 2007 Robbins et al 2008 Robbins et al 2008 Robbins et al 2008 Derbinski et al 2005 Wong et al 2014 Wong et al 2014 Weischenfeldt et al 2008 Derbinski et al 2005 Chambers et al 2007 Chambers et al 2007 Wong et al 2014 Chambers et al 2007 Haddon et al 2009 Haddon et al 2009 Mouse430_2 Th17 93 79 83 86 64 71 72 79 86 MoGene1 Mouse430_2 Th17 iTreg 66 95 78 81 82 97 66 63 37 85 97 83 65 71 76 99 60 84 Mouse430_2 nTreg 98 87 89 83 72 79 86 77 94 Mouse430_2 NK 99 83 84 53 55 45 84 96 84 Mouse430_2 NK 99 83 89 47 70 21 93 99 83 CD11c+ 96 CD8-DCs Mouse430_2 CD11c+ 95 CD8+DCs Mouse430_2 CD11b+DCs 98 74 87 86 96 14 92 60 92 73 85 51 98 23 93 100 91 82 88 87 95 31 94 Mouse430_2 CD8+DCs 96 83 87 57 96 53 94 100 91 Mouse430_2 pDCs 99 78 88 85 92 34 98 100 91 MG_U74Av2 tDC 96 NA 71 92 96 39 90 99 96 MoGene1 84 77 76 29 68 44 49 96 72 MoGene1 Promyelocytes Myelocytes 89 91 74 38 66 45 82 71 84 Mouse430_2 BM.MPh 98 81 74 50 93 21 95 95 86 MG_U74Av2 tMPh 91 NA 77 57 83 42 67 97 92 Mouse430_2 Monocyte 100 91 86 50 79 46 93 75 91 Mouse430_2 Granulocytes 98 83 81 61 65 48 87 82 89 95 77 28 80 45 92 62 91 Mouse430_2 MoGene1 Granulocytes 94 92 91 Mouse430_2 Nu.RBCs 88 79 79 61 59 49 91 69 91 MOE430A mast.cells 82 5 74 42 49 23 49 30 70 MOE430A premast.cells 82 5 73 46 51 25 47 43 71 77 REFERENCES 78 REFERENCES Gene Expression Omnibus. http://www.ncbi.nlm.nih.gov/geo/. Gene Expression Atlas. http://www.ebi.ac.uk/gxa/. Acquaviva, J., Chen, X. & Ren, R. (2008). IRF-4 functions as a tumor suppressor in early B-cell development. Blood 112, 3798-3806. Becker, A. M., Michael, D. G., Satpathy, A. T., Sciammas, R., Singh, H. & Bhattacharya, D. (2012). IRF-8 extinguishes neutrophil production and promotes dendritic cell lineage commitment in both myeloid and lymphoid mouse progenitors. Blood 119, 2003-2012. Beerman, I., Seita, J., Inlay, M. A., Weissman, I. L. & Rossi, D. J. (2014). Quiescent Hematopoietic Stem Cells Accumulate DNA Damage during Aging that Is Repaired upon Entry into Cell Cycle. Cell Stem Cell. Blanco, J. C., Contursi, C., Salkowski, C. A., DeWitt, D. L., Ozato, K. & Vogel, S. N. (2000). Interferon regulatory factor (IRF)-1 and IRF-2 regulate interferon gammadependent cyclooxygenase 2 expression. J Exp Med 191, 2131-2144. Brien, J. D., Daffis, S., Lazear, H. M., Cho, H., Suthar, M. S., Gale, M., Jr. & Diamond, M. S. (2011). Interferon regulatory factor-1 (IRF-1) shapes both innate and CD8(+) T cell immune responses against West Nile virus infection. PLoS Pathog 7, e1002230. Brustle, A., Heink, S., Huber, M. & other authors (2007). The development of inflammatory T(H)-17 cells requires interferon-regulatory factor 4. Nat Immunol 8, 958966. Chambers, S. M., Boles, N. C., Lin, K. Y. & other authors (2007). Hematopoietic fingerprints: an expression database of stem cells and their progeny. Cell Stem Cell 1, 578-591. Chen, C., Grennan, K., Badner, J., Zhang, D., Gershon, E., Jin, L. & Liu, C. (2011). Removing batch effects in analysis of expression microarray data: an evaluation of six batch adjustment methods. PLoS One 6, e17238. Chen, Q., Yang, W., Gupta, S., Biswas, P., Smith, P., Bhagat, G. & Pernis, A. B. (2008). IRF-4-binding protein inhibits interleukin-17 and interleukin-21 production by controlling the activity of IRF-4 transcription factor. Immunity 29, 899-911. 79 Couzinet, A., Tamura, K., Chen, H. M. & other authors (2008). A cell-type-specific requirement for IFN regulatory factor 5 (IRF5) in Fas-induced apoptosis. Proc Natl Acad Sci U S A 105, 2556-2561. Cuesta, N., Nhu, Q. M., Zudaire, E., Polumuri, S., Cuttitta, F. & Vogel, S. N. (2007). IFN regulatory factor-2 regulates macrophage apoptosis through a STAT1/3- and caspase-1-dependent mechanism. J Immunol 178, 3602-3611. Derbinski, J., Gabler, J., Brors, B., Tierling, S., Jonnakuty, S., Hergenhahn, M., Peltonen, L., Walter, J. & Kyewski, B. (2005). Promiscuous gene expression in thymic epithelial cells is regulated at multiple levels. J Exp Med 202, 33-45. Dudziak, D., Kamphorst, A. O., Heidkamp, G. F. & other authors (2007). Differential antigen processing by dendritic cell subsets in vivo. Science 315, 107-111. Duncan, G. S., Mittrucker, H. W., Kagi, D., Matsuyama, T. & Mak, T. W. (1996). The transcription factor interferon regulatory factor-1 is essential for natural killer cell function in vivo. J Exp Med 184, 2043-2048. Egawa, T. & Littman, D. R. (2011). Transcription factor AP4 modulates reversible and epigenetic silencing of the Cd4 gene. Proc Natl Acad Sci U S A 108, 14873-14878. Elser, B., Lohoff, M., Kock, S., Giaisi, M., Kirchhoff, S., Krammer, P. H. & Li-Weber, M. (2002). IFN-gamma represses IL-4 expression via IRF-1 and IRF-2. Immunity 17, 703-712. Endo, Y., Blinova, K., Romantseva, T., Golding, H. & Zaitseva, M. (2014). Differences in PGE2 production between primary human monocytes and differentiated macrophages: role of IL-1beta and TRIF/IRF3. PLoS One 9, e98517. Farlik, M., Rapp, B., Marie, I., Levy, D. E., Jamieson, A. M. & Decker, T. (2012). Contribution of a TANK-binding kinase 1-interferon (IFN) regulatory factor 7 pathway to IFN-gamma-induced gene expression. Mol Cell Biol 32, 1032-1043. Ficara, F., Murphy, M. J., Lin, M. & Cleary, M. L. (2008). Pbx1 regulates self-renewal of long-term hematopoietic stem cells by maintaining their quiescence. Cell Stem Cell 2, 484-496. Fragale, A., Gabriele, L., Stellacci, E. & other authors (2008). IFN regulatory factor-1 negatively regulates CD4+ CD25+ regulatory T cell differentiation by repressing Foxp3 expression. J Immunol 181, 1673-1682. Gabriele, L., Fragale, A., Borghi, P. & other authors (2006). IRF-1 deficiency skews the differentiation of dendritic cells toward plasmacytoid and tolerogenic features. J Leukoc Biol 80, 1500-1511. 80 Gracias, D. T., Stelekati, E., Hope, J. L. & other authors (2013). The microRNA miR155 controls CD8(+) T cell responses by regulating interferon signaling. Nat Immunol 14, 593-602. Haddon, D. J., Hughes, M. R., Antignano, F., Westaway, D., Cashman, N. R. & McNagny, K. M. (2009). Prion protein expression and release by mast cells after activation. J Infect Dis 200, 827-831. Hida, S., Ogasawara, K., Sato, K. & other authors (2000). CD8(+) T cell-mediated skin disease in mice lacking IRF-2, the transcriptional attenuator of interferon-alpha/beta signaling. Immunity 13, 643-655. Hida, S., Tadachi, M., Saito, T. & Taki, S. (2005). Negative control of basophil expansion by IRF-2 critical for the regulation of Th1/Th2 balance. Blood 106, 20112017. Holtschke, T., Lohler, J., Kanno, Y. & other authors (1996). Immunodeficiency and chronic myelogenous leukemia-like syndrome in mice with a targeted mutation of the ICSBP gene. Cell 87, 307-317. Holwerda, S. J., van de Werken, H. J., Ribeiro de Almeida, C. & other authors (2013). Allelic exclusion of the immunoglobulin heavy chain locus is independent of its nuclear localization in mature B cells. Nucleic Acids Res 41, 6905-6916. Honda, K., Yanai, H., Negishi, H. & other authors (2005). IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature 434, 772-777. Hsieh, M. Y., Chang, M. Y., Chen, Y. J. & other authors (2014). The inducible nitricoxide synthase (iNOS)/Src axis mediates Toll-like receptor 3 tyrosine 759 phosphorylation and enhances its signal transduction, leading to interferon-beta synthesis in macrophages. J Biol Chem 289, 9208-9220. Hsu, L. C., Park, J. M., Zhang, K., Luo, J. L., Maeda, S., Kaufman, R. J., Eckmann, L., Guiney, D. G. & Karin, M. (2004). The protein kinase PKR is required for macrophage apoptosis after activation of Toll-like receptor 4. Nature 428, 341-345. Hu, C. M., Jang, S. Y., Fanzo, J. C. & Pernis, A. B. (2002). Modulation of T cell cytokine production by interferon regulatory factor-4. J Biol Chem 277, 49238-49246. Huang, B., Qi, Z. T., Xu, Z. & Nie, P. (2010). Global characterization of interferon regulatory factor (IRF) genes in vertebrates: glimpse of the diversification in evolution. BMC Immunol 11, 22. Ichikawa, E., Hida, S., Omatsu, Y., Shimoyama, S., Takahara, K., Miyagawa, S., Inaba, K. & Taki, S. (2004). Defective development of splenic and epidermal CD4+ 81 dendritic cells in mice deficient for IFN regulatory factor-2. Proc Natl Acad Sci U S A 101, 3909-3914. Iida, S., Rao, P. H., Butler, M., Corradini, P., Boccadoro, M., Klein, B., Chaganti, R. S. & Dalla-Favera, R. (1997). Deregulation of MUM1/IRF4 by chromosomal translocation in multiple myeloma. Nat Genet 17, 226-230. Ingraham, C. R., Kinoshita, A., Kondo, S. & other authors (2006). Abnormal skin, limb and craniofacial morphogenesis in mice deficient for interferon regulatory factor 6 (Irf6). Nat Genet 38, 1335-1340. Irizarry, R. A., Hobbs, B., Collin, F., Beazer-Barclay, Y. D., Antonellis, K. J., Scherf, U. & Speed, T. P. (2003). Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4, 249-264. Ito, M., Iida, S., Inagaki, H. & other authors (2002). MUM1/IRF4 expression is an unfavorable prognostic factor in B-cell chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL). Jpn J Cancer Res 93, 685-694. Janeway, C. A., Travers, P., Walport, M. & Shlomchik, M. J. (2001). Immunobiology, 5th edition edn. New York: Garland Science Jo, S. H. & Ren, R. (2011). IRF-4 suppresses BCR/ABL transformation of myeloid cells in a DNA binding-independent manner. J Biol Chem 287, 1770-1778. Kano, S., Sato, K., Morishita, Y., Vollstedt, S., Kim, S., Bishop, K., Honda, K., Kubo, M. & Taniguchi, T. (2008). The contribution of transcription factor IRF1 to the interferon-gamma-interleukin 12 signaling axis and TH1 versus TH-17 differentiation of CD4+ T cells. Nat Immunol 9, 34-41. Kasprzyk, A. (2011). BioMart: driving a paradigm change in biological data management. Database (Oxford), bar049. Kawazu, M., Yamamoto, G., Yoshimi, M. & other authors (2007). Expression profiling of immature thymocytes revealed a novel homeobox gene that regulates doublenegative thymocyte development. J Immunol 179, 5335-5345. Kim, N., Kukkonen, S., Martinez-Viedma Mdel, P., Gupta, S. & Aldovini, A. (2013). Tat engagement of p38 MAP kinase and IRF7 pathways leads to activation of interferon-stimulated genes in antigen-presenting cells. Blood 121, 4090-4100. Kim, S. S., Lee, K. G., Chin, C. S., Ng, S. K., Pereira, N. A., Xu, S. & Lam, K. P. (2014). DOK3 Is Required for IFN-beta Production by Enabling TRAF3/TBK1 Complex Formation and IRF3 Activation. J Immunol 193, 840-848. 82 Klein, U., Casola, S., Cattoretti, G., Shen, Q., Lia, M., Mo, T., Ludwig, T., Rajewsky, K. & Dalla-Favera, R. (2006). Transcription factor IRF4 controls plasma cell differentiation and class-switch recombination. Nat Immunol 7, 773-782. Kondo, S., Schutte, B. C., Richardson, R. J. & other authors (2002). Mutations in IRF6 cause Van der Woude and popliteal pterygium syndromes. Nat Genet 32, 285289. Lee, C. H., Melchers, M., Wang, H. & other authors (2006). Regulation of the germinal center gene program by interferon (IFN) regulatory factor 8/IFN consensus sequence-binding protein. J Exp Med 203, 63-72. Leek, J. T., Scharpf, R. B., Bravo, H. C., Simcha, D., Langmead, B., Johnson, W. E., Geman, D., Baggerly, K. & Irizarry, R. A. (2010). Tackling the widespread and critical impact of batch effects in high-throughput data. Nat Rev Genet 11, 733-739. Li, S., Wang, L., Berman, M., Kong, Y. Y. & Dorf, M. E. (2011). Mapping a dynamic innate immunity protein interaction network regulating type I interferon production. Immunity 35, 426-440. Li, W., Hofer, M. J., Nocon, A. L., Manders, P. & Campbell, I. L. (2013). Interferon regulatory factor 7 (IRF7) is required for the optimal initial control but not subsequent clearance of lymphocytic choriomeningitis virus infection in mice. Virology 439, 152-162. Lin, C. C., Bradstreet, T. R., Schwarzkopf, E. A. & other authors (2014). Bhlhe40 controls cytokine production by T cells and is essential for pathogenicity in autoimmune neuroinflammation. Nat Commun 5, 3551. Liu, J., Cao, S., Herman, L. M. & Ma, X. (2003). Differential regulation of interleukin (IL)-12 p35 and p40 gene expression and interferon (IFN)-gamma-primed IL-12 production by IFN regulatory factor 1. J Exp Med 198, 1265-1276. Lohoff, M., Ferrick, D., Mittrucker, H. W., Duncan, G. S., Bischof, S., Rollinghoff, M. & Mak, T. W. (1997). Interferon regulatory factor-1 is required for a T helper 1 immune response in vivo. Immunity 6, 681-689. Lohoff, M., Duncan, G. S., Ferrick, D. & other authors (2000). Deficiency in the transcription factor interferon regulatory factor (IRF)-2 leads to severely compromised development of natural killer and T helper type 1 cells. J Exp Med 192, 325-336. Lohoff, M., Mittrucker, H. W., Prechtl, S. & other authors (2002). Dysregulated T helper cell differentiation in the absence of interferon regulatory factor 4. Proc Natl Acad Sci U S A 99, 11808-11812. Lohoff, M. & Mak, T. W. (2005). Roles of interferon-regulatory factors in T-helper-cell differentiation. Nat Rev Immunol 5, 125-135. 83 Lu, R. & Pitha, P. M. (2001). Monocyte differentiation to macrophage requires interferon regulatory factor 7. J Biol Chem 276, 45491-45496. Lu, R., Medina, K. L., Lancki, D. W. & Singh, H. (2003). IRF-4,8 orchestrate the pre-Bto-B transition in lymphocyte development. Genes Dev 17, 1703-1708. Lucas, B. & Germain, R. N. (1996). Unexpectedly complex regulation of CD4/CD8 coreceptor expression supports a revised model for CD4+CD8+ thymocyte differentiation. Immunity 5, 461-477. Ma, S., Turetsky, A., Trinh, L. & Lu, R. (2006). IFN regulatory factor 4 and 8 promote Ig light chain kappa locus activation in pre-B cell development. J Immunol 177, 78987904. Mansson, R., Hultquist, A., Luc, S. & other authors (2007). Molecular evidence for hierarchical transcriptional lineage priming in fetal and adult stem cells and multipotent progenitors. Immunity 26, 407-419. Miranda-Saavedra, D., De, S., Trotter, M. W., Teichmann, S. A. & Gottgens, B. (2009). BloodExpress: a database of gene expression in mouse haematopoiesis. Nucleic Acids Res 37, D873-879. Nehyba, J., Hrdlickova, R. & Bose, H. R. (2009). Dynamic evolution of immune system regulators: the history of the interferon regulatory factor family. Mol Biol Evol 26, 2539-2550. Ouyang, X., Zhang, R., Yang, J. & other authors (2011). Transcription factor IRF8 directs a silencing programme for TH17 cell differentiation. Nat Commun 2, 314. Passioura, T., Shen, S., Symonds, G. & Dolnikov, A. (2005). A retroviral library genetic screen identifies IRF-2 as an inhibitor of N-ras-induced growth suppression in leukemic cells. Oncogene 24, 7327-7336. Pathak, S., Ma, S., Trinh, L., Eudy, J., Wagner, K. U., Joshi, S. S. & Lu, R. (2011). IRF4 is a suppressor of c-Myc induced B cell leukemia. PLoS One 6, e22628. Paul, W. E. (2008 ). Fundamental Immunology, sixth edition edn: Lippincott Williams & Wilkins. Penninger, J. M., Sirard, C., Mittrucker, H. W. & other authors (1997). The interferon regulatory transcription factor IRF-1 controls positive and negative selection of CD8+ thymocytes. Immunity 7, 243-254. 84 Rengarajan, J., Mowen, K. A., McBride, K. D., Smith, E. D., Singh, H. & Glimcher, L. H. (2002). Interferon regulatory factor 4 (IRF4) interacts with NFATc2 to modulate interleukin 4 gene expression. J Exp Med 195, 1003-1012. Richardson, R. J., Dixon, J., Malhotra, S., Hardman, M. J., Knowles, L., BootHandford, R. P., Shore, P., Whitmarsh, A. & Dixon, M. J. (2006). Irf6 is a key determinant of the keratinocyte proliferation-differentiation switch. Nat Genet 38, 13291334. Richardson, R. J., Dixon, J., Jiang, R. & Dixon, M. J. (2009). Integration of IRF6 and Jagged2 signalling is essential for controlling palatal adhesion and fusion competence. Hum Mol Genet 18, 2632-2642. Robbins, S. H., Walzer, T., Dembele, D. & other authors (2008). Novel insights into the relationships between dendritic cell subsets in human and mouse revealed by genome-wide expression profiling. Genome Biol 9, R17. Rodriguez, A., Vigorito, E., Clare, S. & other authors (2007). Requirement of bic/microRNA-155 for normal immune function. Science 316, 608-611. Sakaguchi, S., Negishi, H., Asagiri, M., Nakajima, C., Mizutani, T., Takaoka, A., Honda, K. & Taniguchi, T. (2003). Essential role of IRF-3 in lipopolysaccharideinduced interferon-beta gene expression and endotoxin shock. Biochem Biophys Res Commun 306, 860-866. Salkowski, C. A., Kopydlowski, K., Blanco, J., Cody, M. J., McNally, R. & Vogel, S. N. (1999). IL-12 is dysregulated in macrophages from IRF-1 and IRF-2 knockout mice. J Immunol 163, 1529-1536. Sato, M., Suemori, H., Hata, N. & other authors (2000). Distinct and essential roles of transcription factors IRF-3 and IRF-7 in response to viruses for IFN-alpha/beta gene induction. Immunity 13, 539-548. Sato, T., Onai, N., Yoshihara, H., Arai, F., Suda, T. & Ohteki, T. (2009). Interferon regulatory factor-2 protects quiescent hematopoietic stem cells from type I interferondependent exhaustion. Nat Med 15, 696-700. Savitsky, D. A., Yanai, H., Tamura, T., Taniguchi, T. & Honda, K. (2010). Contribution of IRF5 in B cells to the development of murine SLE-like disease through its transcriptional control of the IgG2a locus. Proc Natl Acad Sci U S A 107, 1015410159. Schiavoni, G., Mattei, F., Sestili, P., Borghi, P., Venditti, M., Morse, H. C., 3rd, Belardelli, F. & Gabriele, L. (2002). ICSBP is essential for the development of mouse type I interferon-producing cells and for the generation and activation of CD8alpha(+) dendritic cells. J Exp Med 196, 1415-1425. 85 Schiavoni, G., Mattei, F., Borghi, P., Sestili, P., Venditti, M., Morse, H. C., 3rd, Belardelli, F. & Gabriele, L. (2004). ICSBP is critically involved in the normal development and trafficking of Langerhans cells and dermal dendritic cells. Blood 103, 2221-2228. Schuettpelz, L. G. & Link, D. C. (2013). Regulation of hematopoietic stem cell activity by inflammation. Front Immunol 4, 204. Sciammas, R., Shaffer, A. L., Schatz, J. H., Zhao, H., Staudt, L. M. & Singh, H. (2006). Graded expression of interferon regulatory factor-4 coordinates isotype switching with plasma cell differentiation. Immunity 25, 225-236. Shaffer, A. L., Emre, N. C., Lamy, L. & other authors (2008). IRF4 addiction in multiple myeloma. Nature 454, 226-231. Simon, A. K., Desrois, M. & Schmitt-Verhulst, A. M. (1997). Interferon-regulatory factors during development of CD4 and CD8 thymocytes. Immunology 91, 340-345. Stellacci, E., Testa, U., Petrucci, E. & other authors (2004). Interferon regulatory factor-2 drives megakaryocytic differentiation. Biochem J 377, 367-378. Stone, R. C., Feng, D., Deng, J., Singh, S., Yang, L., Fitzgerald-Bocarsly, P., Eloranta, M. L., Ronnblom, L. & Barnes, B. J. (2012). Interferon regulatory factor 5 activation in monocytes of systemic lupus erythematosus patients is triggered by circulating autoantigens independent of type I interferons. Arthritis Rheum 64, 788-798. Sun, L., Zhu, Z., Cheng, N., Yan, Q. & Ye, R. D. (2014). Serum amyloid A induces interleukin-33 expression through an IRF7-dependent pathway. Eur J Immunol. Suzuki, S., Honma, K., Matsuyama, T. & other authors (2004). Critical roles of interferon regulatory factor 4 in CD11bhighCD8alpha- dendritic cell development. Proc Natl Acad Sci U S A 101, 8981-8986. Taki, S., Sato, T., Ogasawara, K. & other authors (1997). Multistage regulation of Th1-type immune responses by the transcription factor IRF-1. Immunity 6, 673-679. Taki, S., Nakajima, S., Ichikawa, E., Saito, T. & Hida, S. (2005). IFN regulatory factor2 deficiency revealed a novel checkpoint critical for the generation of peripheral NK cells. J Immunol 174, 6005-6012. Tamura, T., Tailor, P., Yamaoka, K., Kong, H. J., Tsujimura, H., O'Shea, J. J., Singh, H. & Ozato, K. (2005a). IFN regulatory factor-4 and -8 govern dendritic cell subset development and their functional diversity. J Immunol 174, 2573-2581. 86 Tamura, T., Thotakura, P., Tanaka, T. S., Ko, M. S. & Ozato, K. (2005b). Identification of target genes and a unique cis element regulated by IRF-8 in developing macrophages. Blood 106, 1938-1947. Tamura, T., Yanai, H., Savitsky, D. & Taniguchi, T. (2008). The IRF family transcription factors in immunity and oncogenesis. Annu Rev Immunol 26, 535-584. Testa, U., Stellacci, E., Pelosi, E. & other authors (2004). Impaired myelopoiesis in mice devoid of interferon regulatory factor 1. Leukemia 18, 1864-1871. Thibault, D. L., Chu, A. D., Graham, K. L. & other authors (2008). IRF9 and STAT1 are required for IgG autoantibody production and B cell expression of TLR7 in mice. J Clin Invest 118, 1417-1426. Thomason, H. A., Zhou, H., Kouwenhoven, E. N. & other authors (2010). Cooperation between the transcription factors p63 and IRF6 is essential to prevent cleft palate in mice. J Clin Invest 120, 1561-1569. Tian, Y., Wang, C., Ye, Z., Xiao, X., Kijlstra, A. & Yang, P. (2012). Effect of 1,25dihydroxyvitamin D3 on Th17 and Th1 response in patients with Behcet's disease. Invest Ophthalmol Vis Sci 53, 6434-6441. Tothova, Z., Kollipara, R., Huntly, B. J. & other authors (2007). FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128, 325-339. Tsuboi, K., Iida, S., Inagaki, H. & other authors (2000). MUM1/IRF4 expression as a frequent event in mature lymphoid malignancies. Leukemia 14, 449-456. Venkatraman, A., He, X. C., Thorvaldsen, J. L. & other authors (2013). Maternal imprinting at the H19-Igf2 locus maintains adult haematopoietic stem cell quiescence. Nature 500, 345-349. Vigano, M. A., Ivanek, R., Balwierz, P., Berninger, P., van Nimwegen, E., Karjalainen, K. & Rolink, A. (2013). An epigenetic profile of early T-cell development from multipotent progenitors to committed T-cell descendants. Eur J Immunol 44, 11811193. Wang, Y., Krivtsov, A. V., Sinha, A. U., North, T. E., Goessling, W., Feng, Z., Zon, L. I. & Armstrong, S. A. (2010). The Wnt/beta-catenin pathway is required for the development of leukemia stem cells in AML. Science 327, 1650-1653. Wei, G., Wei, L., Zhu, J. & other authors (2009). Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells. Immunity 30, 155-167. 87 Weiden, M., Tanaka, N., Qiao, Y. & other authors (2000). Differentiation of monocytes to macrophages switches the Mycobacterium tuberculosis effect on HIV-1 replication from stimulation to inhibition: modulation of interferon response and CCAAT/enhancer binding protein beta expression. J Immunol 165, 2028-2039. Weischenfeldt, J., Damgaard, I., Bryder, D., Theilgaard-Monch, K., Thoren, L. A., Nielsen, F. C., Jacobsen, S. E., Nerlov, C. & Porse, B. T. (2008). NMD is essential for hematopoietic stem and progenitor cells and for eliminating by-products of programmed DNA rearrangements. Genes Dev 22, 1381-1396. Wong, J. J., Ritchie, W., Gao, D., Lau, K. A., Gonzalez, M., Choudhary, A., Taft, R. J., Rasko, J. E. & Holst, J. (2014). Identification of nuclear-enriched miRNAs during mouse granulopoiesis. J Hematol Oncol 7, 42. Xi, Y., Shao, F., Bai, X. Y., Cai, G., Lv, Y. & Chen, X. (2014). Changes in the expression of the toll-like receptor system in the aging rat kidneys. PLoS One 9, e96351. Yamamoto, M., Kato, T., Hotta, C. & other authors (2011). Shared and distinct functions of the transcription factors IRF4 and IRF8 in myeloid cell development. PLoS One 6, e25812. Yanai, H., Chen, H. M., Inuzuka, T., Kondo, S., Mak, T. W., Takaoka, A., Honda, K. & Taniguchi, T. (2007). Role of IFN regulatory factor 5 transcription factor in antiviral immunity and tumor suppression. Proc Natl Acad Sci U S A 104, 3402-3407. Yang, L., Feng, D., Bi, X., Stone, R. C. & Barnes, B. J. (2012). Monocytes from Irf5-/mice have an intrinsic defect in their response to pristane-induced lupus. J Immunol 189, 3741-3750. Yasuda, K., Richez, C., Maciaszek, J. W., Agrawal, N., Akira, S., MarshakRothstein, A. & Rifkin, I. R. (2007). Murine dendritic cell type I IFN production induced by human IgG-RNA immune complexes is IFN regulatory factor (IRF)5 and IRF7 dependent and is required for IL-6 production. J Immunol 178, 6876-6885. Zhang, X., Tao, Y., Troiani, L. & Markovic-Plese, S. (2011). Simvastatin inhibits IFN regulatory factor 4 expression and Th17 cell differentiation in CD4+ T cells derived from patients with multiple sclerosis. J Immunol 187, 3431-3437. Zheng, Y., Chaudhry, A., Kas, A. & other authors (2009). Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T(H)2 responses. Nature 458, 351-356. Zhou, S., Cerny, A. M., Fitzgerald, K. A., Kurt-Jones, E. A. & Finberg, R. W. (2013). Role of interferon regulatory factor 7 in T cell responses during acute lymphocytic choriomeningitis virus infection. J Virol 86, 11254-11265. 88 CHAPTER THREE The role of Irf6 in T-cell development in the thymus 89 Abstract Rare variants in IRF6 cause Van der Woude and Popliteal Pterygium Syndromes, two autosomal dominant orofacial clefting disorders. Common IRF6 variants contribute risk toward non-syndromic orofacial clefting. In addition, rare somatic mutations in IRF6 are associated with squamous cell carcinoma. However, unlike the other eight members of the IRF family, a role for IRF6 in haematopoietic development has not been described. Previously, we used publically available data to discover dynamic IRF6 expression in developing thymocytes. Here, we utilized a mouse model to show that Irf6 was required for the regulation of thymocyte development. We found that Irf6 was expressed in the subcapsular region and medulla of the thymus. We further found that Irf6 regulated the distribution and proliferation of developing thymocytes. In addition, loss of Irf6 led to an increase in double negative cells with a concomitant increase in TCRγδ. Loss of Irf6 also led to a reduction in double positive cells with no corresponding reduction in single positive cell maturation. Finally, we found that Irf6 dose is critical in development of both CD4+ and CD8+ cells in an age-dependent manner. While perinatal lethality has limited investigation of Irf6 in hematopoiesis, we report here a novel gene function for Irf6 in thymocyte development. These data suggest that IRF6 variants may increase risk toward autoimmune disease and that individuals with VWS and PPS may require more rigorous immunological screening. With this work, all Irf family members have an important role in immunity. 90 Introduction The thymus gland is a specialized organ necessary for T-cell development. It is composed of an inner medulla and a peripheral cortex surrounded by an outer capsule (Rodewald, 2008; Singer et al., 1986). Thymus tissue is composed of lymphoid cells (CD45+CD7+) and stromal cells with a ratio of 50 lymphoid cells for each stromal cell (Rodewald, 2008; Singer et al., 1986). Non-hematopoietic stromal cells can be further classified into thymic epithelial cells (TEC, Keratin+) and mesenchymal cells (Keratin−) (Anderson et al., 1993). Dendritic cells and macrophages are CD45+ thymic stromal cells, thus they constitute the hematopoietic component of the stromal mesh (Rodewald, 2008). T-cell precursors seed the thymus at the medullary cortical junction. Recent thymic immigrants are called double negative (DN) thymocytes because they lack the expression of both CD4 and CD8 (Godfrey et al., 1993; Pearse et al., 1989). CD44 and CD25 are two surface markers which mark 4 major developmental sub-populations of DN thymocytes (DN1, CD44+CD25-; DN2, CD44+CD25+; DN3, CD44-CD25+; and DN4, CD44-CD25-) (Godfrey et al., 1993). DN3 is an obligatory check point where expression of pre-TCR or γδTCR identifies transition from DN3a to DN3b (Michie & ZunigaPflucker, 2002). Pre-TCR signaling drives expression of CD4 and CD8 producing double-positive (DP) thymocytes (Hoffman et al., 1996). Developing thymocytes have to migrate through the cortex toward the capsule then back to the medullary space (Takahama, 2006). Transition from DN4 (also called pre-DP) to DP cells occurs in the subcapsular region. Transforming growth factor β (TGFβ) signaling suppresses the proliferation of pre-DP thymocytes to regulate the production of DP cells (Benz et al., 2004). DP cells migrate back through the cortex where positive and negative selections 91 occur allowing only 3-5% of cells to survive and reach the thymic medulla. DP cells lose either CD4 or CD8 to reach the single positive (SP) stage. SP thymocytes, either CD4+ (T helper) cells or CD8+ (T cytotoxic) cells, continue their maturation and central tolerance in the medulla before being shuttled out of the thymus (Blackburn & Manley, 2004; Germain, 2002; Hoffmann et al., 2003; Lind et al., 2001; Plotkin et al., 2003; Prockop & Petrie, 2000). Regulation of peripheral CD4:CD8 cell ratios originate in the thymus. Species and strain differences contributed to TCR selection-dependent mechanisms or thymic lineage commitment signaling (Damoiseaux et al., 1999; Rocha et al., 1989; Sim et al., 1998; van Meerwijk et al., 1998). Genetic variations of TCRα loci and MHC haplotypes represent the most important TCR selection-dependent factors (Damoiseaux et al., 1999; Sim et al., 1998). Intrinsic activity of Notch is an example of other factors that influence the thymic lineage commitment and affect the CD4:CD8 T cell ratio (Fowlkes & Robey, 2002; Huang et al., 2003; Robey et al., 1996). Also the interaction of Notch ligand Jagged1 on thymic stroma and DP thymocytes controls the CD4:CD8 ratio (Jimenez et al., 2001). Thymic involution was shown to be induced by Jagged1 expression in thymocytes (Beverly et al., 2006) and is associated with increased CD4:CD8 ratio (Kozlowska et al., 2007). Although several Irfs are expressed in thymocytes (Colantonio et al., 2011; Hrdlickova et al., 2001; Nordang et al., 2011; Simon et al., 1997), Irf1 is the only family member with functional studies supporting a role in T-cell development. Irf1 reduces expression of the major histocompatibility complex related genes in the thymic microenvironment (Lee et al., 1999; White et al., 1996). However it is the Irf1 intrinsic 92 activity in T-cells that is required for development and thymic selection of naïve CD8 Tcells (Matsuyama et al., 1993; Penninger et al., 1997; Penninger & Mak, 1998). In Irf4 knockout mice, while cell count changes appear to be unaffected, the proliferative capacity, antiviral cytotoxicity, allogenic graft rejection, tumor surveillance and cytokine production of CD8 cells are markedly impaired (Mittrucker et al., 1997). Recently, I performed a meta-analysis of microarray experiments on hematopoietic lineages and found that the steady-state level of Irf6 is induced in the DP and SP thymocytes as well as medullary thymic epithelium. Previous studies in the palatal and skin development show Irf6 as a downstream target to Tgf-β and notch signaling (Le et al., 2012; Restivo et al., 2011; Xu et al., 2006). However, these interactions have not been tested in T cells. In this study, we used a mouse model to test for the necessity of Irf6 in thymocyte development. We found that Irf6 regulated the distribution and proliferation of developing thymocytes. Also, we found that Irf6 dose is critical in development of both CD4+ and CD8+ cells in an age-dependent manner. These data suggest that Irf6 like all other family members is involved in regulation of immune system. 93 Figure 3.1: Stages of T cell development in the thymus. Figure modified from Ciofani and Zuniga-Pflucker, Nat Rev Immunol 2010. 94 Materials and methods Mice: C57BL/6 mice were obtained from The Jackson Laboratory (Bar Harbor, ME) or raised in house. Mice with Irf6 gene trap allele (Irf6gt/+) were described before (Ingraham et al., 2006). Four week-old wild type C57BL/6 mice were used for initial assessment of Irf6 expression in the thymus. To compare wild-type and Irf6-knockout thymi, Irf6gt/+ mice were mated to produce wild-type and Irf6knockout embryos. Pregnant females were sacrificed, embryos were dissected and thymi were collected at E17.5. To study the effect of Irf6 heterozygosity on postnatal thymic proliferation, Irf6gt/+ and wildtype mice were allowed to mate and heterozygous mice were compared to their wild type littermates at 6-7, 12-13, or 18-20 weeks of age. Morphological assessment of Irf6 knockout thymi: Relative size of thymi was determined as previously described (Candi et al., 2007). Briefly, thymic gland and heart were dissected using a dissecting microscope. Thymus/heart ratio was defined using the longest dimension of each. Western blot analysis: Whole cell extracts were prepared by mechanical disruption of thymi in RIPA buffer (150 mM NaCl, 1% Triton X-100, 0.5% sodium deoxycholate, 0.1% SDS, 50 mM Tris, pH 8.0) supplemented by protease inhibitors (Complete Protease Inhibitor Cocktail Tablets, Roche). Samples were denatured by incubating at 95°C for 10 minutes. Protein extracts were separated by 10% SDS-PAGE Electrophoresis System and transferred onto PVDF membranes with 1X Tris-Glycine Transfer Buffer. Membranes were blocked with Odyssey blocking solution and probed with polyclonal rabbit antibody against Irf6 95 (1:250) (Ingraham et al., 2006) and mouse antibody against GAPDH (sc-365062, 1:15000, Santa Cruz Biotechnology, Santa Cruz, CA). Proteins of interest were detected with Infrared IRDye-labeled secondary Donkey anti-Rabbit and goat anti-mouse IgG (H + L) antibodies (1:15000, Thermo Scientific, Rockford, IL) imaged with the Odyssey Infrared Imaging System using the 700 and 800 channels (Thermo Scientific, Rockford, IL), according to the provided protocol. Immunohistochemistry: Thymi were fixed in 10% formaldehyde, embedded in paraffin and cut into 3 µm-thick transverse sections. To deparaffinize and rehydrate sections, slides were passed through three changes of Xylene, followed by reducing concentrations of ethanol. Antigen retrieval was conducted by boiling in 10mM Sodium Citrate pH6.0 for 10 minutes. Sections were permeabilized in 0.5% Triton X-100 and blocked for one hour in blocking solution (10% normal goat serum, 0.1% Bovine Serum Albumin in 1X Phosphate Buffered Saline) at room temperature. Sections were then incubated in primary antibodies diluted in blocking solution overnight at 4°C. Following incubation, slides were washed three times in 1X PBS and incubated in fluorescent labeled secondary antibodies for 45 min at room temperature. To detect nuclei, slides were incubated in DAPI (Invitrogen, D3571) diluted 1:10,000 in distilled water for 10 minutes. Slides were imaged using the Nikon i90 upright fluorescent microscope. Primary antibodies included rabbit polyclonal anti-Irf6 antibody (Ingraham et al., 2006) and rat monoclonal anti-BrdU (Abcam, Ab6326).. Secondary antibodies included Oregon Green 488 goat anti-rabbit (Molecular Probes, O-6381) and Alexa Fluor 555 Goat Anti-Rat (Molecular Probes, A-21434). 96 Total cell count and Flow cytometric analysis: Single cell suspension was prepared from dissociated thymi by filtering through a 48-µm nylon mesh. Total thymocyte count was calculated using hemocytometer. Cell count was adjusted to 1x106 followed by FcR blocking with anti-FcR mAb 2.4G2. Subsequent flowcytometric analysis included one of the following: a) Analysis of the thymocyte subpopulations: To cover most of the developing T-cell populations, cells were co-stained for 6 surface markers; against CD4 (APC, Catalog# 100516, Biolegend), CD8a (FITC, Catalog # 140404, Biolegend), TCRβ (PE-cy7, Catalog# 109222, Biolegend), TCRγδ (PE-cy5.5, Catalog# 118118, Biolegend), CD25 (PE, Catalog# 100610, Biolegend), CD44 (APC-cy7, Catalog# 103028, Biolegend) (figure 3.2). Natural Treg was assessed separately by co-surface staining of CD4 (APC, Catalog# 100516, Biolegend) and CD8a (FITC, Catalog # 140404, Biolegend) with intracellular staining for Foxp3 (PE, Catalog# 12-5773-82, eBioscience) according to the recommended protocol of the company. 97 Figure 3.2: Flowcytometric analysis of the thymocyte subpopulations. Left scatter diagram plots CD4 and CD8 expression to classify the developing thymocytes into four major groups (DN; black, DP; blue, CD4-SP; red, and CD8-SP; green). Middle scatter diagram plots CD25 and CD44 expression to tease apart the sub-populations of DN thymocytes. Right scatter plots TCRβ and TCRγδ expression. 98 b) Bromodeoxyuridine (BrdU) incorporation analysis: For embryonic studies, pregnant dams were injected intraperitoneally with BrdU (100ug/gm body weight) one hour before euthanasia. For postnatal studies, each animal was injected intraperitoneally with BrdU (100ug/gm body weight) two hours before euthanasia. After blocking of non-specific binding of Fc receptors, surface staining was done using monoclonal antibodies against CD4 (Alexafluor 700, Catalog# 100536, Biolegend) and CD8a (FITC, Catalog # 140404, Biolegend). Cells were fixed and permeabilized with BD Cytofix/Cytoperm Buffer (Catalog# 554722, BD) for 20 min on ice. For nuclear permeabilization, cells were kept overnight at -80oC in a freezing medium. On the next day, cells were thawed and washed with FACS buffer prior to refixation in BD Cytofix/Cytoperm Buffer for 5min on ice. DNA was partially digested by incubating the cells with 30 µg of DNase/106 cells (Catalog# D-4513, Sigma) for 45min at 37oC. Cells were stained with Anti-BrdU (PerCP-Cy 5.5, Catalog# 560809, BD). c) Detection of apoptosis was performed using combinations of Annexin V Apoptosis Detection Kit PerCP-eFluor® 710 and Fixable Viability Dye eFluor-780 (eBioscience) 99 Results Thymic expression of Irf6: Whole-cell protein extracts were prepared from thymi of 4 week-old mice (n=3). Western blot analysis showed a protein band at the expected size (~53kD) (figure 3.3). To determine in situ localization of Irf6 expression, immunofluorescent staining was done on paraffin embedded tissue (n=3). Irf6 expression was mainly confined to the thymic medulla and subcapsular compartments (Fig.3.4). The thymic structure did allow delineating the exact cell type expressing Irf6 without co-staining for additional markers. Unlike expected for transcriptions factors, Irf6 expression was confined to the cytoplasmic spaces. However this pattern of Irf6 expression is consistent with previous studies of in other tissues (Bailey et al., 2005; Ingraham et al., 2006). Irf6 Gapdh Figure 3.3: Two-color western blot for Irf6 and Gapdh. Irf6 is Red (~53kD) and Gapdh is green (~37kD). Lanes from left to right represent protein standard, TNT (IRF6 protein synthesized by invitro transcription and translation system) and whole-cell protein extract from thymus from a 4 week old mouse (n=3). 100 A D B E C Figure 3.4: Irf6 expression in the thymus. (A) Schematic representation of the thymus. (B-E) Immunofluorescent staining of thymic sections obtained from 4 week-old mice. Nuclear DAPI staining (Blue), Irf6 staining (green). The primary Irf6 antibody was pre-incubated with (B) or without (C) blocking peptide and images were taken under 4x objective magnification. 40x magnification is shown for the sub-capsular region (D) and thymic medulla (E) (n=3). 101 Characterization of thymic changes in Irf6 knock-out embryos: Thymi of E17.5 littermates were compared. No obvious changes of the position, shape or size were detected (figure 3.5). Total thymocyte count showed insignificant changes from wild type littermates by paired t-test analysis. However further analysis of developing thymocyte sub-populations showed significant increase of DN population (p = 0.017) and significant decrease of DP population (p < 0.001) with insignificant change of single positive populations. We used BrdU staining as a marker of DNA synthesis and cellular proliferation. While the total uptake of BrdU incorporation was not different, we found that DN cells were more proliferative (p = 0.007) while DP cells were less proliferative (p = 0.018) (figure 3.6 and 3.7). Immunostaining for BrdU in paraffin embedded thymi showed a different distribution of proliferating thymocytes through the cortical space compared to the typical localization in the subcapsular space (Figure 3.8). Surface staining for TCR showed significant increase of TCRγδ in knockout thymi by a paired t-test analysis between littermates (p = 0.01). We did not find a significant difference in TCRβ (Figure 3.9). Testing nuclear expression to mark nTreg showed that embryonic thymi did not start producing Foxp3 +ve T cells at E17.5. We also analyzed Annexin V as a marker of cellular apoptosis and the fixable viability dye to quantify dead cells. Co-staining for both markers showed no change in the total rate of thymocyte apoptosis or cell death in Irf6 knockout thymocytes (Figure 3.10). 102 A. Wild type T H B. Irf6gt/gt T H Figure 3.5: Thymic-cardiac ratio in A) wild type and B) Irf6gt/gt mice. (T) refers to the thymus and (H) refers to the heart. There was no obvious change in the thymic-cardiac ratio (n=3). 103 Wild type Irf6gt/gt Figure 3.6: BrdU incorporation in developing thymocytes. BrdU incorporation in thymocyte populations in wild type (upper panel) and Irf6gt/gt (lower panel) using flowcytometry. Left column scatter plots show the size scatter on x-axes and forward scatter on y-axes. Right side scatter plots show the surface expression of CD4 on the x-axes and CD8 on the y-axes. Red color identifies the thymocytes from black colored debris. Blue color overlay the red color to identify the cells incorporating BrdU. There is no significant difference in total BrdU incorporation. However, we observed a shift of BrdU incorporation from the DP compartment to the DN compartment which compensated the overall proliferation in Irf6 knockout mice. 104 Figure 3.7: Irf6 regulates proliferation and cell count of thymocytes. Statistical analysis of BrdU incorporation and cell counts of total thymocytes and the developing sub-population shows underline the importance of Irf6. In spite of insignificant changes in total proliferation and cell count, there is a significant increase in the count and proliferation of DN cells with a corresponding reduction in DP cells. 105 Irf6gt/gt Wild Type Figure 3.8: Loss of Irf6 leads to an abnormal distribution of proliferating cells. Immunofluorescent staining of thymic sections from E17.5 embryos. Nuclear DAPI staining (Blue), BrdU staining (red). Compared to wildtype embryos, Irf6 knockout embryos have more cellular proliferation in the thymic medulla (n=3). % 3 2 2.8 1 2.6 2 2.4 2.2 2 2 4 2 1.8 1.6 3 2 1.4 Irf6-WT Irf6-KO Figure 3.9: Frequency of TCRγδ. The figure shows the average frequency of TCRγδ for each genotype per littermates. Each point is labeled by the numbers of embryos. Paired t-test is significant with p = 0.01 106 Figure 3.10: Loss of Irf6 does not alter total cell death. Analysis of apoptosis and cell death. Left scatter diagram shows the plotting of the fixable viability dye on the x-axes versus Annexin V on the y-axes where (Q1) square the apoptotic cells and (Q2) square represents the dead cells. Par plot on the right shows insignificant changes in the total count of apoptotic or dead cells in Irf6 knock-outs (n=11 wildtype/heterozygous and 7 knockouts from 4 litters). Effect of Irf6 heterozygosity in postnatal thymic proliferation: To study the effect of Irf6 dosage on thymic proliferation over time, we tested mice at postnatal week 6-7, 12-13, or 18-20. At each time point, three heterozygous mice were compared to their matching wild type littermates. BrdU incorporation study showed an insignificant change of total BrdU uptake but the ratio CD4-SP subpopulation positive for BrdU shows significant increase (p = 0.04) versus a significant reduction in the CD8-SP compartment (p = 0.03) at 18-20 weeks of age (figure 3.11). 107 A B Figure 3.11: Effect of Irf6 dosage on BrdU incorporation in thymocyte populations. 108 Figure 3.11 (Cont’d) (A) Flowcytometric analysis of thymi from Irf6 wild type (Upper panel) and Irf6gt/+ (lower panel). Scatter plots shows the surface expression of CD4 on x-axes and CD8 on y-axes at 6-7, 12-13, or 18-20 weeks of age (from left to right). Red color identifies the thymocytes incorporating BrdU. (B) Bar diagram represents the changes of total thymocytes proliferation and the four major sub-populations (mean and SD) at 6, 12 and 20 weeks of age in Irf6+/+ and Irf6gt/+ thymi (n=3 for each time point and genotype). There is a significant increase of CD4 population with a significant reduction of CD8 thymocytes after 20 weeks of age. 109 Discussion The IRF family of transcription factors has been widely studied and regarded in hematopoiesis as master regulators. While IRF6 is a paralog, sharing a highly conserved DNA and protein binding domains, a role in immunity has not been reported. Instead, previous and ongoing IRF6 studies have focused on other important roles in cutaneous, limb and craniofacial development. In humans, common variants in IRF6 lead to common, complex diseases, including cleft lip and palate and squamous cell carcinoma. Furthermore, rare variants in IRF6 lead to Van der Woude (VWS) and Popliteal Pterygium Syndromes (PPS). Most proximally, these data suggest that individuals with common and rare IRF6 variants are at increased risk for immunological diseases. Like cutaneous development, we found that Irf6 regulates proliferation in thymocytes. In skin and thymus, loss of Irf6 leads to an expansion of the progenitor cells; germinative and double negative cells respectively. Also a concomitant reduction in downstream daughter cells (keratinizing and double positive cells). However, in contrast to skin which loses the cornified cells, DP cells are not lost. Also we did not see changes in the terminally differentiated single positive thymocytes. Therefore, while some parallels exist, a more complex relationship in the thymus plausible. Importantly, we found that counts of single positive cells were not altered despite a significant reduction in double positive cells. Our analysis did not show a change in the total number of apoptotic or dead cells. However, further analysis for the thymic sub-population is mandatory. These data can be 110 explained by either enhanced survival of single positive cells or thymic retention of naïve lymphocytes. The enhancement of survival of single positive cells may occur at the expense of negative selection and predisposition for autoimmune responses. We further observed that Irf6 was expressed in the subcapsular cortex and medulla. However, we have not delineated if this expression was in the thymic epithelium or thymocytes or both. Considering that negative selection is a product of the interaction between the thymic epithelium and double positive thymocytes, enhanced survival could be a cell-autonomous mechanism or results from a milieu of factors. Retention of thymic naïve lymphocytes can be presented as reduced peripheral lymphocyte count. Neonatal lymphopenia itself was shown to predispose for autoimmune disease (Gleeson et al., 1996; Sakaguchi & Sakaguchi, 1989). While an association between VWS/PPS and autoimmune diseases or neonatal lymphopenia has not been described in the literature, multiple factors are likely involved. For example, VWS/PPS are rare congenital anomalies and subclinical manifestations of immunological diseases can be missed. Also considering the role of the Irf family members in T cell development, redundant function is possible. Interactions between Irf6 and other Irf family members seem plausible, if not likely. Pursuing this gene regulatory network may provide novel gene functions for Irf6 in T cell subsets. Thymic involution is a very controlled process which starts early in life and considered as a central driver of T cell aging (Goronzy & Weyand, 2013) (Aspinall et al., 2010). Aging of thymus is associated with altered distribution of thymocyte populations. Whereas the DN cells start to accumulate, the DP cell 111 counts decrease with a significant increase of CD4:CD8 ratio (Kozlowska et al., 2007). Embryonic absence of Irf6 is associated with similar changes of DN and DP populations but without significant changes of CD4:CD8 ratio. However, by the age of 6 month, animals with reduced dose of Irf6 start to show significant increase of CD4:CD8 ratio. . Two transgenic lines with constitutively active form of Notch in DP cells showed a decrease in CD4 SP thymocytes and a corresponding increase in CD8 SP thymocytes (Fowlkes & Robey, 2002). IRF6 is a known mediator of Notch in keratinocytes (Restivo et al., 2011). Irf6 heterozygosity in thymocytes might be associated with decreased downstream signals of Notch pathway causing increased CD4:CD8 ratio. Also Expression of the Notch ligand Jagged1 in thymocytes results in thymic involution by inducing apoptosis of thymic stromal epithelial cells (Beverly et al., 2006). This might explain the altered CD4:CD8 observed in mice heterozygous for Irf6 as a sign of early thymic involution. In summery, we describe a novel gene function for Irf6 in hematopoietic development. To our knowledge, this is the first study to ascribe immunological function to Irf6, completing the family so that all nine paralogs now have a role in haematopoiesis. 112 REFERENCES 113 REFERENCES Anderson, G., Jenkinson, E. J., Moore, N. C. & Owen, J. J. (1993). MHC class II-positive epithelium and mesenchyme cells are both required for T-cell development in the thymus. Nature 362, 70-73. Aspinall, R., Pitts, D., Lapenna, A. & Mitchell, W. (2010). Immunity in the elderly: the role of the thymus. J Comp Pathol 142 Suppl 1, S111-115. Bailey, C. M., Khalkhali-Ellis, Z., Kondo, S. & other authors (2005). Mammary serine protease inhibitor (Maspin) binds directly to interferon regulatory factor 6: identification of a novel serpin partnership. J Biol Chem 280, 34210-34217. Benz, C., Heinzel, K. & Bleul, C. C. (2004). Homing of immature thymocytes to the subcapsular microenvironment within the thymus is not an absolute requirement for T cell development. Eur J Immunol 34, 3652-3663. Beverly, L. J., Ascano, J. M. & Capobianco, A. J. (2006). Expression of JAGGED1 in T-lymphocytes results in thymic involution by inducing apoptosis of thymic stromal epithelial cells. Genes Immun 7, 476-486. Blackburn, C. C. & Manley, N. R. (2004). Developing a new paradigm for thymus organogenesis. Nat Rev Immunol 4, 278-289. Candi, E., Rufini, A., Terrinoni, A., Giamboi-Miraglia, A., Lena, A. M., Mantovani, R., Knight, R. & Melino, G. (2007). DeltaNp63 regulates thymic development through enhanced expression of FgfR2 and Jag2. Proc Natl Acad Sci U S A 104, 11999-12004. Colantonio, A. D., Epeldegui, M., Jesiak, M., Jachimowski, L., Blom, B. & Uittenbogaart, C. H. (2011). IFN-alpha is constitutively expressed in the human thymus, but not in peripheral lymphoid organs. PLoS One 6, e24252. Damoiseaux, J. G., Cautain, B., Bernard, I., Mas, M., van Breda Vriesman, P. J., Druet, P., Fournie, G. & Saoudi, A. (1999). A dominant role for the thymus and MHC genes in determining the peripheral CD4/CD8 T cell ratio in the rat. J Immunol 163, 2983-2989. Fowlkes, B. J. & Robey, E. A. (2002). A reassessment of the effect of activated Notch1 on CD4 and CD8 T cell development. J Immunol 169, 1817-1821. Germain, R. N. (2002). T-cell development and the CD4-CD8 lineage decision. Nat Rev Immunol 2, 309-322. 114 Gleeson, P. A., Toh, B. H. & van Driel, I. R. (1996). Organ-specific autoimmunity induced by lymphopenia. Immunol Rev 149, 97-125. Godfrey, D. I., Kennedy, J., Suda, T. & Zlotnik, A. (1993). A developmental pathway involving four phenotypically and functionally distinct subsets of CD3CD4-CD8- triple-negative adult mouse thymocytes defined by CD44 and CD25 expression. J Immunol 150, 4244-4252. Goronzy, J. J. & Weyand, C. M. (2013). Understanding immunosenescence to improve responses to vaccines. Nat Immunol 14, 428-436. Hoffman, E. S., Passoni, L., Crompton, T., Leu, T. M., Schatz, D. G., Koff, A., Owen, M. J. & Hayday, A. C. (1996). Productive T-cell receptor beta-chain gene rearrangement: coincident regulation of cell cycle and clonality during development in vivo. Genes Dev 10, 948-962. Hoffmann, R., Bruno, L., Seidl, T., Rolink, A. & Melchers, F. (2003). Rules for gene usage inferred from a comparison of large-scale gene expression profiles of T and B lymphocyte development. J Immunol 170, 1339-1353. Hrdlickova, R., Nehyba, J. & Bose, H. R., Jr. (2001). Interferon regulatory factor 4 contributes to transformation of v-Rel-expressing fibroblasts. Mol Cell Biol 21, 6369-6386. Huang, E. Y., Gallegos, A. M., Richards, S. M., Lehar, S. M. & Bevan, M. J. (2003). Surface expression of Notch1 on thymocytes: correlation with the doublenegative to double-positive transition. J Immunol 171, 2296-2304. Ingraham, C. R., Kinoshita, A., Kondo, S. & other authors (2006). Abnormal skin, limb and craniofacial morphogenesis in mice deficient for interferon regulatory factor 6 (Irf6). Nat Genet 38, 1335-1340. Jimenez, E., Vicente, A., Sacedon, R., Munoz, J. J., Weinmaster, G., Zapata, A. G. & Varas, A. (2001). Distinct mechanisms contribute to generate and change the CD4:CD8 cell ratio during thymus development: a role for the Notch ligand, Jagged1. J Immunol 166, 5898-5908. Kozlowska, E., Biernacka, M., Ciechomska, M. & Drela, N. (2007). Agerelated changes in the occurrence and characteristics of thymic CD4(+) CD25(+) T cells in mice. Immunology 122, 445-453. Le, M., Naridze, R., Morrison, J., Biggs, L. C., Rhea, L., Schutte, B. C., Kaartinen, V. & Dunnwald, M. (2012). Transforming growth factor Beta 3 is required for excisional wound repair in vivo. PLoS One 7, e48040. 115 Lee, C. K., Gimeno, R. & Levy, D. E. (1999). Differential regulation of constitutive major histocompatibility complex class I expression in T and B lymphocytes. J Exp Med 190, 1451-1464. Lind, E. F., Prockop, S. E., Porritt, H. E. & Petrie, H. T. (2001). Mapping precursor movement through the postnatal thymus reveals specific microenvironments supporting defined stages of early lymphoid development. J Exp Med 194, 127-134. Matsuyama, T., Kimura, T., Kitagawa, M. & other authors (1993). Targeted disruption of IRF-1 or IRF-2 results in abnormal type I IFN gene induction and aberrant lymphocyte development. Cell 75, 83-97. Michie, A. M. & Zuniga-Pflucker, J. C. (2002). Regulation of thymocyte differentiation: pre-TCR signals and beta-selection. Semin Immunol 14, 311-323. Mittrucker, H. W., Matsuyama, T., Grossman, A. & other authors (1997). Requirement for the transcription factor LSIRF/IRF4 for mature B and T lymphocyte function. Science 275, 540-543. Nordang, G. B., Viken, M. K., Amundsen, S. S., Sanchez, E. S., Flato, B., Forre, O. T., Martin, J., Kvien, T. K. & Lie, B. A. (2011). Interferon regulatory factor 5 gene polymorphism confers risk to several rheumatic diseases and correlates with expression of alternative thymic transcripts. Rheumatology (Oxford) 51, 619-626. Pearse, M., Wu, L., Egerton, M., Wilson, A., Shortman, K. & Scollay, R. (1989). A murine early thymocyte developmental sequence is marked by transient expression of the interleukin 2 receptor. Proc Natl Acad Sci U S A 86, 1614-1618. Penninger, J. M., Sirard, C., Mittrucker, H. W. & other authors (1997). The interferon regulatory transcription factor IRF-1 controls positive and negative selection of CD8+ thymocytes. Immunity 7, 243-254. Penninger, J. M. & Mak, T. W. (1998). Thymocyte selection in Vav and IRF-1 gene-deficient mice. Immunol Rev 165, 149-166. Plotkin, J., Prockop, S. E., Lepique, A. & Petrie, H. T. (2003). Critical role for CXCR4 signaling in progenitor localization and T cell differentiation in the postnatal thymus. J Immunol 171, 4521-4527. Prockop, S. & Petrie, H. T. (2000). Cell migration and the anatomic control of thymocyte precursor differentiation. Semin Immunol 12, 435-444. 116 Restivo, G., Nguyen, B. C., Dziunycz, P. & other authors (2011). IRF6 is a mediator of Notch pro-differentiation and tumour suppressive function in keratinocytes. EMBO J 30, 4571-4585. Robey, E., Chang, D., Itano, A., Cado, D., Alexander, H., Lans, D., Weinmaster, G. & Salmon, P. (1996). An activated form of Notch influences the choice between CD4 and CD8 T cell lineages. Cell 87, 483-492. Rocha, B., Dautigny, N. & Pereira, P. (1989). Peripheral T lymphocytes: expansion potential and homeostatic regulation of pool sizes and CD4/CD8 ratios in vivo. Eur J Immunol 19, 905-911. Rodewald, H. R. (2008). Thymus organogenesis. Annu Rev Immunol 26, 355388. Sakaguchi, S. & Sakaguchi, N. (1989). Organ-specific autoimmune disease induced in mice by elimination of T cell subsets. V. Neonatal administration of cyclosporin A causes autoimmune disease. J Immunol 142, 471-480. Sim, B. C., Aftahi, N., Reilly, C., Bogen, B., Schwartz, R. H., Gascoigne, N. R. & Lo, D. (1998). Thymic skewing of the CD4/CD8 ratio maps with the T-cell receptor alpha-chain locus. Curr Biol 8, 701-704. Simon, A. K., Desrois, M. & Schmitt-Verhulst, A. M. (1997). Interferonregulatory factors during development of CD4 and CD8 thymocytes. Immunology 91, 340-345. Singer, K. H., Wolf, L. S., Lobach, D. F., Denning, S. M., Tuck, D. T., Robertson, A. L. & Haynes, B. F. (1986). Human thymocytes bind to autologous and allogeneic thymic epithelial cells in vitro. Proc Natl Acad Sci U S A 83, 6588-6592. Takahama, Y. (2006). Journey through the thymus: stromal guides for T-cell development and selection. Nat Rev Immunol 6, 127-135. van Meerwijk, J. P., Bianchi, T., Marguerat, S. & MacDonald, H. R. (1998). Thymic lineage commitment rather than selection causes genetic variations in size of CD4 and CD8 compartments. J Immunol 160, 3649-3654. White, L. C., Wright, K. L., Felix, N. J., Ruffner, H., Reis, L. F., Pine, R. & Ting, J. P. (1996). Regulation of LMP2 and TAP1 genes by IRF-1 explains the paucity of CD8+ T cells in IRF-1-/- mice. Immunity 5, 365-376. Xu, X., Han, J., Ito, Y., Bringas, P., Jr., Urata, M. M. & Chai, Y. (2006). Cell autonomous requirement for Tgfbr2 in the disappearance of medial edge epithelium during palatal fusion. Dev Biol 297, 238-248. 117 CHAPTER FOUR The role of Irf6 in functional commitment of T-cell subsets 118 Abstract The IRF family of transcription factors is essential in the differentiation of T helper subsets. In contrast, IRF6 is canonically known for critical roles in craniofacial, limb and cutaneous development. In the mouse, loss of Irf6 leads to perinatal lethality. Recently, we showed that Irf6 is also involved in the regulation of thymic proliferation. However, little is known about the necessity of Irf6 in the development of functionally committed T helper subsets. Here, we used in silico, in vivo and in vitro assays to determine the role of Irf6 in T cell differentiation. Using in silico analysis, we found and propose a model for Irf6 function in Th17/Treg balance. To test our hypothesis in vivo and overcome perinatal lethality, we employed an adaptive transfer of Irf6 knockout cells into lethally irradiated mice. We observed a 100% survival of chimeric mice receiving Irf6 knockout fetal liver, and mice receiving Irf6 knockout cells had no deficit in restoration of lymphocyte production. In addition, we used two in vitro models to assess the necessity of Irf6 in the commitment of T helper cells. Using a stromal-free culture we found that naive T cells lacking Irf6 could be differentiated into Th1, Th2, Th17 and Treg using a specific cytokine cocktail. We found no differences in cell frequency and mean fluorescence intensity of intracellular cytokines between wild type and Irf6 knockout cells. In vitro differentiation of dendritic cells showed significant increase of MHC-II expression after three days of culture. Irf6 might be involved in post-translational regulation of MHC-II. In conclusion, we found that intrinsic Irf6 expression was not essential for T helper subset differentiation. However, a non-cell autonomous role for Irf6 in T cell differentiation through dendritic cells remains plausible. 119 Introduction T helper (Th) lymphocytes function as the conductors of the adaptive immune orchestra. Upon antigen exposure, T helper cells differentiate into specialized subsets. Each T helper subset differentiates under a unique signaling pathway and lineagespecific transcription factors to produce a characteristic cytokine milieu (Fietta & Delsante, 2009; Hirahara et al., 2011). T helper subsets include Th1, Th2, Th17, regulatory T-cells (Treg), T follicular helper cells, Th9 and Th22 cells (Bluestone et al., 2009; Shevach, 2010). The balance between different T helper cells is most typically defined by mutually exclusive expression of lineage-specific transcription factors. In vitro activation of T cells can be done using specific antibodies against the T-cell receptors. The combination of anti-CD3 and anti-CD28 is used to mimic antigenic stimulation (Bjorndahl et al., 1989; Verwilghen et al., 1991). Cytokine cocktails can be formulated to simulate the physiological signaling pathways and drive the differentiation of naïve CD4 T cells into one of several lineages of T helper cells (Constant & Bottomly, 1997; Zhu et al., 2010). Differentiation of naïve cells can be analyzed by measuring their secreted cytokine profile. As such, stromal free models of in-vitro differentiation enable identification of cell autonomous mutant T cell effects. Normal differentiation of T cells requires antigen presentation and cytokine guidance, two essential functions of Dendritic cells (DCs). DCs are a heterogeneous population of hematopoietic cells known as professional antigen presenting cells (APCs) that display different anatomical localizations, cell surface phenotypes and functions. They all come from CD34 bone marrow stem cells and express CD11c (Merad et al., 2013; Miller et al., 2012). In their immature state they have the ability to respond to 120 danger signals, engulf antigen, mature and migrate to lymphoid organs. Once the antigen, foreign or self, is internalized they are degraded and presented in the surface of the DCs in the context of major histocompatibility complex class I (MHC-I) and class II (MHC-II). DC maturation involves an increase in surface expression of MHC-II and costimulatory molecules like CD86 (Banchereau et al., 2000). There are four major categories of DCs: Conventional DC, Langerhan cells, plasmacytoid DCs and monocyte-derived DCs. All except for the Langerhan cells are derived from bone marrow cells. Conventional DCs are specialized for antigen presentation and have two further subdivisions, CD11b+ and CD103+ DCs (Belz & Nutt, 2012). DCs can differentiate in vitro from bone marrow and blood using a combination of growth factors like granulocyte macrophage-colony stimulating factors (GM-CSF), IL-4, Flt3 ligand and tumor necrosis factor-α (Lipscomb & Masten, 2002). We have shown abnormal thymic development in Irf6 deficient mice (Chapter 3). With abnormal thymic development, we can predict abnormal counts of mature T cell subsets in the peripheral blood and/or biased T cell immune response (Tanigaki et al., 2004). Irf1, 2, 4, 8 are indispensable for normal T helper differentiation (Kano et al., 2008; Lohoff et al., 2000; Lohoff & Mak, 2005; Tamura et al., 2008; Tominaga et al., 2003). Also, variants in IRF5, IRF7 and IRF8 are associated with psoriasis, multiple sclerosis and systemic lupus erythematosus (SLE) (De Jager et al., 2009; Demirci et al., 2007; Gateva et al., 2009; Graham et al., 2006; Harley et al., 2008; Leppa et al., 2011; Patel, 2011; Sanchez et al., 2008). Conservation of IRF family members (Lohoff & Mak, 2005) and documented protein-protein interactions between IRF6 and both IRF5 and IRF8 (Li et al., 2011) suggest a role of IRF6 in T helper cell commitment. My meta- 121 analysis of microarray studies in T helper subsets reveals a significant reduction of Irf6 expression in Th1 compared to Th2, Th17 and Treg suggesting an intrinsic role for Irf6 in development of T helper subsets (Chapter 1). Materials and methods Bioinformatic analysis: Publically available microarray studies were meta-analyzed using R computing environment (http://www.r-project.org/) to predict the possible roles of Irf6 in T-helper commitment. We examined two microarray studies altering Irf6 expression; 1) Irf6 knockout mouse skin (Ingraham et al., 2006) and 2) IRF6 knockdown in human keratinocytes (Botti et al., 2011). We also analyzed two microarray studies that altered Foxp3 expression; 1) knockout (Williams & Rudensky, 2007) and 2) overexpression (Fontenot et al., 2005) of Foxp3 in CD4+CD25+ cells. Data were integrated with Chip-seq analysis of IRF6 binding sites in human keratinocytes (Botti et al., 2011). Mice and adoptive transfer: Mice heterozygous for the Irf6 gene trap allele (Irf6gt/+) were mated to produce wild-type (Irf6+/+) and Irf6-knockout (Irf6gt/gt) embryos. Fetal livers were harvested from E12.5 embryos and suspended in Iscove’s modified Dulbecco’s medium supplemented by 2% fetal calf serum. A single-cell suspension was prepared by passage through a 26-gauge needle. Crude DNA extraction was done and rapid PCR-based genotyping was performed. Recipient mice were congenic strain of C57BL/6 mice that carry the differential B cell antigen designated CD45.1 (NCI, Washington, DC. NY). Ten-week old recipient mice were lethally irradiated using XRAD320 Irradiation System (PXi, North Branford, CT). Recipient mice received two doses of irradiation (5.5 Gy each) with a three hour interval. Irradiated mice were 122 injected retro-orbitally with the liver cell suspensions after 6 hours from the first dose of irradiation. Host mice were maintained on autoclaved water containing trimethoprimsulfamethoxazole (0.65-1.6 mg/ml). All mice were maintained at the Michigan State University pathogen-free facility. Th1, Th2, Th17 and Treg differentiation in vitro: Single cell suspensions of the spleens were prepared under sterile conditions. Naïve T-cells were purified by negative selection using magnetic beads from EasyStep Mouse Naïve CD4+ T Cell Isolation Kit (STEMCELL Technologies Inc, Vancouver, BC, Canada). Unwanted cells were targeted for removal with biotinylated antibodies directed against non-naïve CD4+ T cells (CD8, CD11b, CD11c, CD19, CD24, CD25, CD44, CD45R, CD49b, TCRγ/δ, TER119) then captured by streptavidin-coated magnetic particles. Naïve T-cells were cultured in Xvivo medium supplemented by 1 mM sodium pyruvate, nonessential amino acids, and L-glutamine. Culture plates were coated with anti-CD3ε antibody (clone 145-2C11, BD, Cat# 553058) 5µg/mL. Soluble anti-CD28 antibody (clone 37.51, BD, Cat# 553295) 2µg/ml, 2-mercaptoethanol 50 µM and 1X Penicillin-Streptomycin were added to the culture medium. Medium was enriched by the differentiation cocktail of Th1 [IL2 (20ng/ml, R&D, cat# 202-IL-010) + IL12 (20ng/ml, R&D, cat# 419-ML-010) + anti- IL4 (10µg/ml, R&D, cat# AB-404-NA)], Th2 [IL2 (20ng/ml, R&D, cat# 202-IL-010) + IL4 (100ng/ml, R&D, cat# 404-ML-010) + anti-IFN-γ (10µg/ml, BD, cat# 554408) + anti-IL12 (10µg/ml, Biolegend, cat# 505304)], Th17 [TGFβ1 (1ng/ml, R&D, cat# 7666-MB-005) + IL6 (100ng/ml, R&D, cat# 406-ML-005) + anti- IL4 (10µg/ml, R&D, cat# AB-404-NA) + anti-IFN-γ (10µg/ml, BD, cat# 554408) + FICZ (300 nM, Santa cruz, cat# sc-300019A)], or iTreg [IL2 (20ng/ml, R&D, cat# 202-IL-010) + TGFβ1 (5ng/ml, R&D, cat# 7666-MB- 123 005) ] or under non-skewing conditions [only IL2 (20ng/ml, R&D, cat# 202-IL-010)]. Cells were cultured for four days, washed, and re-suspended in new wells at 106 cells/ml in the presence of 10ng/ml PMA and 1µg/ml ionomycin (extra supplementation with 2-mercaptoethanol and IL2 or IL6 enhance the cells’ viability). After 1 hour, 1x Monensin was added for another four hours to block the cytokine secretion. Ice-cold EDTA was added to a final concentration 2 mM and incubated for 15 min at room temperature to decrease the cellular clumping. All cells were collected, washed and stained for viability with the fixable viability dye eFluor 780 as recommended (eBioscience, cat# 65-0865). Each cell type was intracellularly stained for CD4 and the appropriate differentiation marker (INF-γ for Th1, IL4 for Th2, IL17 for Th17 and Foxp3 for iTreg) (Bettelli et al., 2006; Ghoreschi et al., 2010; Huh et al., 2011; Maruyama et al., 2011; McKenzie et al., 1999; Nurieva et al., 2009; Rodriguez et al., 2007; Zhou et al., 2003). In vitro generation of bone marrow–derived DC: Femurs and tibias of adoptively transferred mice were flushed for bone marrow, and 5 x 106 cells were grown in a 6-well plate (BD Falcon Franklin Lakes, NJ). We used 4 ml of RPMI media supplemented with 10% serum containing 100 units/ml penicillin and 100 µg/ml streptomycin (Gibco, Invitrogen). This media was applied for 9 days in the presence of granulocytemacrophage colony stimulating factor (20 ng/ml; Peprotech, Rocky Hill, NJ). In total, 50% of the media was replaced on days 3, 6, and 8. After 9 days, bone marrow–derived DC (BM-DC) were left unstimulated (naive) or stimulated with LPS (1 µg/ml) in 2% fetal bovine serum RPMI. After 24 h, BM-DC were stained for CD8 (53–6.7), CD103 (2E7), CD11b (M1/70), CD11c (N418), CD86 (GL-1), Gr-1 (RB6-8C5), MHC-I (H-2Kb AF6- 124 88.5), and MHC-II (I-A/I-E, M5/114.15.2) and analyzed by flow cytometry for analysis of DC populations. Total CD11c+ were counted to represent the total DCs. We attempted to identify three subsets of DCs, including CD8 DCs, CD103 DCs and CD11b DCs (for gating scheme, see figure 4.1) 125 Figure 4.1: Gating scheme for identification of DC populations in BM. Living cells were either gated for CD8+CD11c DCs (A) or gated to identify CD103+ and CD11b+ cell populations (B). CD103+ cell population were further gated to identify CD103+CD11c+CD8- DCs (C). CD11b+ cell population were further gated to identify CD1b+CD11c+CD8- DCs (D) 126 Results Bioinformatic prediction of possible roles of Irf6 in T-helper commitment: Analysis of microarray data from Irf6-knockout skin (Ingraham et al., 2006) showed a 13-fold reduction of Il18. However, no significant change of IL18 was found when IRF6 was knocked down in keratinocytes (Botti et al., 20xx). Microarray analysis of the Foxp3 knockout cells showed a 2-fold reduction of Irf6, Foxp3 overexpression increases Irf6 transcription (citation). Therefore, Foxp3 regulates Irf6 expression in CD4+ CD25+ Tcells. Integration of gene expression profiling and Chip-seq analysis of IRF6 in keratinocytes showed significant effects on several T cell transcription factors. There was significant up-regulation of Id3 (p=0.000584) and down-regulation of GATA3 (p=0.015595) and Klf4 (p=0.006814). ChIP sequencing for IRF6 binding sites showed six candidate binding sites in a gene desert downstream to GATA3. In addition, IRF6 bound to and was required for the regulation of Klf4 (Botti et al., 20xx). Thus, bioinformatic analyses of publicly-available data suggest that Irf6 is involved in a gene regulatory network during T-helper commitment. Efficiency of adoptive transfer: As expected, 100% mortality was observed in untreated control animals 11-14 days after irradiation. Post-mortem examination showed severe pallor of the internal organs and gastrointestinal bleeding. On the other hand, all irradiated mice treated with adoptive transfer survived beyond this time point. To further confirm the efficiency of adoptive transfer, peripheral blood lymphocytes of recipient mice were stained for CD45.1 (the native marker of the recipient mice) and CD45.2 (marker of donor mice). 127 Flowcytometric analysis of living lymphocytes showed a predominance of the donor marker in recipient mice (Figure 4.2). Figure 4.2: Efficiency of adoptive transfer. The histograms show the level of CD45.1 expression (marker of the recipient mice) and CD45.2 expression (marker of donor mice) in peripheral blood lymphocytes of adoptively transferred mice. Th1, Th2, Th17 and Treg differentiation in vitro: We cultured naïve CD4 cells isolated from the spleens of recipient mice after adoptive transfer from either wild type or Irf6 knockout fetal livers. Cultured CD4 cells originating from wild type or Irf6 knockout cells were not different in both non-skewing (Il2 only) or differentiation conditions specific for Th1, Th2, Th17 or Treg (Figure 4.3). 128 A Figure 4.3: Th1, Th2, Th17 and Treg differentiation in vitro. 129 Figure 4.3 (Cont’d) % of activated cells B 90 80 70 60 50 40 30 20 10 0 WT/Het (n=7) Irf6-ko (n=6) INFg IL4 IL17 Foxp3 Naïve T-cells were cultured with plate-bound anti-CD3 and soluble anti-CD28 in X-vivo medium enriched by differentiation cocktails for Th1, Th2, Th17, Treg or under non-skewing conditions for 4 days. Cultured cells were reactivated with PMA and ionomycin for 5 hours and cytokine expression was blocked by Monensin. Living cells were determined by the viability dye assay. Differentiated cells were identified by co-expression of intracellular CD4 and either INF-γ, IL4, IL17, or Foxp3 as markers of Th1, Th2, Th17 and iTreg populations. Thresholds were identified by comparison to matching populations grown under non-skewing conditions (Panel A from top to bottom respectively shows a representative sample). Mean and standard error of Irf6 expression in non-mutants and Irf6 knockouts were blotted in the bar graph (B). 130 Changes of bone marrow DCs in Irf6 adoptively transferred mice: Bioinformatic analysis suggested a possible role for Irf6 in regulating the functions of DCs. Flowcytometric analysis of living cells in bone marrow (BM) from adoptively transferred animals showed ~ 3.5 % CD11c+ cells in both wild type and Irf6 knockout samples. CD8+CD11c+ cells were barely detected (< 0.2%). The two main DCs subpopulations identified were CD103+CD8- and CD11b+CD8- cells. Subpopulation size did not allow for a statistical analysis (Figure 4.4). Figure 4.4: Frequency of total DCs and their sub-populations in bone marrow. About 3.5 % are positive for CD11c+. CD8+CD11c+ cells were barely detected (< 0.2%). The two main DCs sub-populations identified were CD103+CD8- and CD11b+CD8- cells (n= 3 for each genotype). 131 In GM-CSF supplemented culture, the cells soon became adherent to the plates. Cells were sampled for flowcytometric analysis at days 3, 6, and 9 of culture. The cultures were sampled after another 24 hours under normal culturing conditions or under stimulation with LPS. The frequency of CD11c+ cells increased gradually with selective differentiation into CD11b+CD8- sub-population. We also found a CD11bhi population between days 3 to 6 day that was lost by the ninth day. We saw a relatively faster increase in the frequency of CD11c+ and CD11b+ at day 9 of culture in Irf6 knockout mice but this was not statistically significant (Figure 4.5). 132 A C B Total DCs cells (CD11c+) CD11b+CD11c+CD8- DCs D Figure 4.5: In vitro differentiation model of DCs. 133 Figure 4.5 (Cont’d): Cells were cultured with GM-CSF and sampled at culture days 0, 3, 6 and 9. Cells were than cultured for another 24 hours either unstimulated or under stimulation with LPS. (A) A representative sample showing the expression of CD11b and CD103 on living cells. The cells gradually progressed towards CD11b positive phenotypes. There is a CD11bhi population seen between days 3 to 6 that was than lost by the end of day 9. (B) The expression of CD11c and CD8 on CD11b+ cells from panel A. There is gradual increase in CD11c+ cells through the days of cultures until reaching almost 100%. (C,D) Bar diagrams showing the changes in the frequency of total DCs (All CD11c+ cells) (C) and changes in the CD11b+CD11c+CD8- DCs sub-population (D) in wild type and Irf6 knockout cells. GM-CSF skewed the differentiation of the bone marrow cells into CD11c+ CD11b+DC. By day 3, almost all cells that are CD11c+ are CD11b+ as well. There is no significant difference between the cell frequencies of wild type and Irf6 knockout mice (n= 3 for each genotype).. 134 MHC-II and the co-stimulatory molecule CD86 are surface markers of DCs associated with maturity and functional commitment (Banchereau et al., 2000). We followed up the expression of maturation markers at the time of BM sampling and throughout the in vitro differentiation course. We saw an initial increase of both MHC-II and CD86 in knockout Irf6 DCs at the time of BM collection, however it was not statistically significant. Cultured cells showed a marked increase of maturation markers at day 3 followed by gradual decrease until day 9. Interestingly, the level of MHC-II expression was significantly higher in knockout Irf6 DCs at day 3 (p value = 0.016). As expected, there was a marked increase in the expression of MHC-II and CD86 after 24 hours of LPS stimulation but there was no significant difference between wild type and knockout Irf6 cells (Figure 4.6). 135 A B Figure 4.6: Expression of maturation markers on DCs in culture. Mean fluorescence intensity of MHC-II (A) and CD86 (B) expression from DCs following in vitro differentiation cultures of wild type and Irf6 knockout mice. Cells were sampled at days 0, 3, 6 and 9. Then further sampled 24 hours after culturing either unstimulated or under stimulation with LPS. A significant increase in MHC-II expression was observed at day 3 between Irf6 wild type and knockout mice (n= 3 for each genotype). 136 Discussion Irf family members are either indispensable for normal T helper differentiation or have known variants associated with autoimmune disorders of T cells (Lohoff & Mak, 2005; Patel, 2011). Foxp3, Id3, Gata3 and Klf4 are a group of transcription factors responsible for developmental regulation and balance between Th17 and Treg. Microarray analysis of Foxp3 altered expression in CD4+ CD25+ suggested that Irf6 is a downstream target of Foxp3. Botti et al 2011 performed gene expression profiling in primary human keratinocytes after siRNA-mediated IRF6 depletion. Expression analysis showed a significant increase of ID3 and significant reduction of GATA3 and KLF4. In the same study, genome-wide analysis of IRF6 binding sites suggested direct transcriptional regulation with both GATA3 and KLF4. These data favor an intrinsic role of Irf6 in regulating the balance of Th17 and Treg with less Th17 differentiation relative to Tregs in the Irf6 knockout (Figure 4.7) Figure 4.7: Proposed model of Irf6 in Th17/Treg balance. Bold black lines represent the well-known pathway of Th17/Treg. The red lines represent the predicted genetic interactions with Irf6 137 Adoptive transfer is a successful tool to overcome the problem of perinatal lethality. One major advantage compared to the tissue specific knockout approaches is the ability to study the effect of gene deficiency in the whole haematopoietic system with the known extensive cell-cell interactions. However, adoptive transfer experiments generate animal chimeras which reduce but do not eliminate the effect of the target gene. Using adoptive transfer, we found that Irf6 knockout fetal livers were sufficient to replenish the bone marrow of lethally irradiated mice. Lymphocytes in the peripheral blood of chimeric mice were shown to be mostly derived from donor stem cells indicating functional competency of Irf6-deficient stem cells. We successfully completed in vitro culture to yield different T helper subsets using conditioned medium. The rate and efficiency of differentiated T helper subsets seen here was highly comparable to prior work. We compared the frequency and mean fluorescence intensity of cytokine production with and without Irf6. Remarkably, in vitro differentiation of Irf6 knockout cells successfully produced all T helper subsets targeted in this work. In fact, the production of these T helper subsets was not different from wild type cells. Therefore, we conclude that intrinsic Irf6 expression is not essential for the differentiation of Th1, Th2, Th17 or Treg. Bioinformatic analysis showed a 13-fold reduction of Il18 in Irf6-deficient skin but no change after knocking down IRF6 in human keratinocytes. Because keratinocytes and APCs are the predominate source of Il18 in skin (Huising et al., 2004), these findings favor APCs as the primary driver for changes in Il18 expression in the Irf6deficient skin (Nicklin et al., 1994; Suttles et al., 1990). Il18 is a powerful Th1-polarizing 138 cytokine (Fietta & Delsante, 2009) so that systemic deficiency of APC-derived Il18 in Irf6 knockout would be expected to prevent the appropriate polarization of Th1. To test this hypothesis we studied the Irf6-deficient bone marrow DCs from adoptively transferred mice. We tested for any changes in counts and adopted an in vitro approach to test their development and functional commitment Direct analysis of bone marrow from adoptively transferred animals showed that 3.5% of bone marrow cells stained positive for the DC marker CD11c independent of the genotype. Most of CD11c+ cells were almost equally divided between CD103 and CD11b sub-populations. The paucity of this subpopulation did not allow for a valid statistical analysis. For in vitro differentiation with GM-CSF, we saw a gradual increase in CD11c+ cells until reaching almost 100%. GMCSF skewed the differentiation of the bone marrow cells into CD11c+ CD11b+ DC. By day 3, CD103 DCs were lost and almost all cells that are CD11c+ became CD11b+ as well. It is interesting to note the appearance of the CD11b-high population between days 3 to 6. However, the magnitude of CD11b expression decreased by day 9. CD11b, also known as Integrin alpha M and macrophage-1 antigen (Mac-1), is a member of the β2-integrin family of adhesion molecules. It plays a role in cell adhesion, phagocytosis and extravasation (Springer, 1990). Initially, in our in vitro model, the seeded bone marrow cells that were non-adherent become adherent upon persistent GM-CSF addition. This observation might explain the requirement of such high levels of integrin expression. High confluence in the plates at the later time points of the culture could trigger a reduction in expression. 139 As markers of maturation, we assessed the expression of MHC-II antigens and the co-stimulatory molecule CD86 throughout the days of culture. Significantly, we found a trend toward increased MHC-II expression in knockout CD11b+ DCs that became statistically significant at culture day 3. MHC-II expression in APCs is tightly regulated (Pai et al., 2002). In mouse immature dendritic cells, MHC-II beta-chain cytoplasmic tail is ubiquitinated which is partly required for the sequestration of MHC-II (Shin et al., 2006; Tze et al., 2011). Interestingly, Irf6 is known to negatively regulate transcriptional factor P63 (Tp63) by targeting it for proteasome-mediated degradation (Thomason et al., 2010). Proteasomal degradation of Tp63 is also ubiquitin-mediated (Li et al., 2008; Westfall et al., 2005). Irf6 might be required at least partially for ubiquitinmediated regulation of MHC-II in immature DCs. This finding supports our bioinformatic suggestion for a non-cell autonomous role of Irf6 in T cell differentiation through dendritic cells. Normal counts and overall developmental pattern of Irf6-deficient bone marrow DCs suggests functional redundancy of Irf6. However, our bioinformatic proposal is mainly pointing to Langerhans cells, a DC population that predominate the skin. Considering that Irf6 function has most clearly been delineated in skin, specific analysis of Langerhans cells might be a valuable determination. 140 REFERENCES 141 REFERENCES Banchereau, J., Briere, F., Caux, C., Davoust, J., Lebecque, S., Liu, Y. J., Pulendran, B. & Palucka, K. (2000). Immunobiology of dendritic cells. Annu Rev Immunol 18, 767-811. Belz, G. T. & Nutt, S. L. (2012). Transcriptional programming of the dendritic cell network. Nat Rev Immunol 12, 101-113. Bettelli, E., Carrier, Y., Gao, W., Korn, T., Strom, T. B., Oukka, M., Weiner, H. L. & Kuchroo, V. K. (2006). Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441, 235-238. Bjorndahl, J. M., Sung, S. S., Hansen, J. A. & Fu, S. M. (1989). Human T cell activation: differential response to anti-CD28 as compared to anti-CD3 monoclonal antibodies. Eur J Immunol 19, 881-887. Bluestone, J. A., Mackay, C. R., O'Shea, J. J. & Stockinger, B. (2009). The functional plasticity of T cell subsets. Nat Rev Immunol 9, 811-816. Botti, E., Spallone, G., Moretti, F. & other authors (2011). Developmental factor IRF6 exhibits tumor suppressor activity in squamous cell carcinomas. Proc Natl Acad Sci U S A 108, 13710-13715. Constant, S. L. & Bottomly, K. (1997). Induction of Th1 and Th2 CD4+ T cell responses: the alternative approaches. Annu Rev Immunol 15, 297-322. De Jager, P. L., Jia, X., Wang, J. & other authors (2009). Meta-analysis of genome scans and replication identify CD6, IRF8 and TNFRSF1A as new multiple sclerosis susceptibility loci. Nat Genet 41, 776-782. Demirci, F. Y., Manzi, S., Ramsey-Goldman, R. & other authors (2007). Association of a common interferon regulatory factor 5 (IRF5) variant with increased risk of systemic lupus erythematosus (SLE). Ann Hum Genet 71, 308-311. Fietta, P. & Delsante, G. (2009). The effector T helper cell triade. Riv Biol 102, 61-74. Fontenot, J. D., Rasmussen, J. P., Williams, L. M., Dooley, J. L., Farr, A. G. & Rudensky, A. Y. (2005). Regulatory T cell lineage specification by the forkhead transcription factor foxp3. Immunity 22, 329-341. Gateva, V., Sandling, J. K., Hom, G. & other authors (2009). A large-scale replication study identifies TNIP1, PRDM1, JAZF1, UHRF1BP1 and IL10 as risk loci for systemic lupus erythematosus. Nat Genet 41, 1228-1233. 142 Ghoreschi, K., Laurence, A., Yang, X. P. & other authors (2010). Generation of pathogenic T(H)17 cells in the absence of TGF-beta signalling. Nature 467, 967-971. Graham, R. R., Kozyrev, S. V., Baechler, E. C. & other authors (2006). A common haplotype of interferon regulatory factor 5 (IRF5) regulates splicing and expression and is associated with increased risk of systemic lupus erythematosus. Nat Genet 38, 550555. Harley, J. B., Alarcon-Riquelme, M. E., Criswell, L. A. & other authors (2008). Genome-wide association scan in women with systemic lupus erythematosus identifies susceptibility variants in ITGAM, PXK, KIAA1542 and other loci. Nat Genet 40, 204-210. Hirahara, K., Vahedi, G., Ghoreschi, K., Yang, X. P., Nakayamada, S., Kanno, Y., O'Shea, J. J. & Laurence, A. (2011). Helper T-cell differentiation and plasticity: insights from epigenetics. Immunology 134, 235-245. Huh, J. R., Leung, M. W., Huang, P. & other authors (2011). Digoxin and its derivatives suppress TH17 cell differentiation by antagonizing RORgammat activity. Nature 472, 486-490. Huising, M. O., Stet, R. J., Savelkoul, H. F. & Verburg-van Kemenade, B. M. (2004). The molecular evolution of the interleukin-1 family of cytokines; IL-18 in teleost fish. Dev Comp Immunol 28, 395-413. Ingraham, C. R., Kinoshita, A., Kondo, S. & other authors (2006). Abnormal skin, limb and craniofacial morphogenesis in mice deficient for interferon regulatory factor 6 (Irf6). Nat Genet 38, 1335-1340. Kano, S., Sato, K., Morishita, Y., Vollstedt, S., Kim, S., Bishop, K., Honda, K., Kubo, M. & Taniguchi, T. (2008). The contribution of transcription factor IRF1 to the interferon-gamma-interleukin 12 signaling axis and TH1 versus TH-17 differentiation of CD4+ T cells. Nat Immunol 9, 34-41. Leppa, V., Surakka, I., Tienari, P. & other authors (2011). The genetic association of variants in CD6, TNFRSF1A and IRF8 to multiple sclerosis: a multicenter case-control study. PLoS One 6, e18813. Li, S., Wang, L., Berman, M., Kong, Y. Y. & Dorf, M. E. (2011). Mapping a dynamic innate immunity protein interaction network regulating type I interferon production. Immunity 35, 426-440. Li, Y., Zhou, Z. & Chen, C. (2008). WW domain-containing E3 ubiquitin protein ligase 1 targets p63 transcription factor for ubiquitin-mediated proteasomal degradation and regulates apoptosis. Cell Death Differ 15, 1941-1951. 143 Lipscomb, M. F. & Masten, B. J. (2002). Dendritic cells: immune regulators in health and disease. Physiol Rev 82, 97-130. Lohoff, M., Duncan, G. S., Ferrick, D. & other authors (2000). Deficiency in the transcription factor interferon regulatory factor (IRF)-2 leads to severely compromised development of natural killer and T helper type 1 cells. J Exp Med 192, 325-336. Lohoff, M. & Mak, T. W. (2005). Roles of interferon-regulatory factors in T-helper-cell differentiation. Nat Rev Immunol 5, 125-135. Maruyama, T., Li, J., Vaque, J. P. & other authors (2011). Control of the differentiation of regulatory T cells and T(H)17 cells by the DNA-binding inhibitor Id3. Nat Immunol 12, 86-95. McKenzie, G. J., Fallon, P. G., Emson, C. L., Grencis, R. K. & McKenzie, A. N. (1999). Simultaneous disruption of interleukin (IL)-4 and IL-13 defines individual roles in T helper cell type 2-mediated responses. J Exp Med 189, 1565-1572. Merad, M., Sathe, P., Helft, J., Miller, J. & Mortha, A. (2013). The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol 31, 563-604. Miller, J. C., Brown, B. D., Shay, T. & other authors (2012). Deciphering the transcriptional network of the dendritic cell lineage. Nat Immunol 13, 888-899. Nicklin, M. J., Weith, A. & Duff, G. W. (1994). A physical map of the region encompassing the human interleukin-1 alpha, interleukin-1 beta, and interleukin-1 receptor antagonist genes. Genomics 19, 382-384. Nurieva, R., Yang, X. O., Chung, Y. & Dong, C. (2009). Cutting edge: in vitro generated Th17 cells maintain their cytokine expression program in normal but not lymphopenic hosts. J Immunol 182, 2565-2568. Pai, R. K., Askew, D., Boom, W. H. & Harding, C. V. (2002). Regulation of class II MHC expression in APCs: roles of types I, III, and IV class II transactivator. J Immunol 169, 1326-1333. Patel, D. R. (2011). Linking interferon regulatory factors to the pathogenesis of human lupus. Transl Res 157, 323-325. Rodriguez, A., Vigorito, E., Clare, S. & other authors (2007). Requirement of bic/microRNA-155 for normal immune function. Science 316, 608-611. Sanchez, F. O., Linga Reddy, M. V., Mallbris, L., Sakuraba, K., Stahle, M. & Alarcon-Riquelme, M. E. (2008). IFN-regulatory factor 5 gene variants interact with the 144 class I MHC locus in the Swedish psoriasis population. J Invest Dermatol 128, 17041709. Shevach, E. D., T. (2010). Regulatory T cells. Nature reviews of Immunology 10. Shin, J. S., Ebersold, M., Pypaert, M., Delamarre, L., Hartley, A. & Mellman, I. (2006). Surface expression of MHC class II in dendritic cells is controlled by regulated ubiquitination. Nature 444, 115-118. Springer, T. A. (1990). Adhesion receptors of the immune system. Nature 346, 425434. Suttles, J., Giri, J. G. & Mizel, S. B. (1990). IL-1 secretion by macrophages. Enhancement of IL-1 secretion and processing by calcium ionophores. J Immunol 144, 175-182. Tamura, T., Yanai, H., Savitsky, D. & Taniguchi, T. (2008). The IRF family transcription factors in immunity and oncogenesis. Annu Rev Immunol 26, 535-584. Tanigaki, K., Tsuji, M., Yamamoto, N., Han, H., Tsukada, J., Inoue, H., Kubo, M. & Honjo, T. (2004). Regulation of alphabeta/gammadelta T cell lineage commitment and peripheral T cell responses by Notch/RBP-J signaling. Immunity 20, 611-622. Thomason, H. A., Zhou, H., Kouwenhoven, E. N. & other authors (2010). Cooperation between the transcription factors p63 and IRF6 is essential to prevent cleft palate in mice. J Clin Invest 120, 1561-1569. Tominaga, N., Ohkusu-Tsukada, K., Udono, H., Abe, R., Matsuyama, T. & Yui, K. (2003). Development of Th1 and not Th2 immune responses in mice lacking IFNregulatory factor-4. Int Immunol 15, 1-10. Tze, L. E., Horikawa, K., Domaschenz, H. & other authors (2011). CD83 increases MHC II and CD86 on dendritic cells by opposing IL-10-driven MARCH1-mediated ubiquitination and degradation. J Exp Med 208, 149-165. Verwilghen, J., Baroja, M. L., Van Vaeck, F., Van Damme, J. & Ceuppens, J. L. (1991). Differences in the stimulating capacity of immobilized anti-CD3 monoclonal antibodies: variable dependence on interleukin-1 as a helper signal for T-cell activation. Immunology 72, 269-276. Westfall, M. D., Joyner, A. S., Barbieri, C. E., Livingstone, M. & Pietenpol, J. A. (2005). Ultraviolet radiation induces phosphorylation and ubiquitin-mediated degradation of DeltaNp63alpha. Cell Cycle 4, 710-716. 145 Williams, L. M. & Rudensky, A. Y. (2007). Maintenance of the Foxp3-dependent developmental program in mature regulatory T cells requires continued expression of Foxp3. Nat Immunol 8, 277-284. Zhou, W., Zhang, F. & Aune, T. M. (2003). Either IL-2 or IL-12 is sufficient to direct Th1 differentiation by nonobese diabetic T cells. J Immunol 170, 735-740. Zhu, J., Yamane, H. & Paul, W. E. (2010). Differentiation of effector CD4 T cell populations (*). Annu Rev Immunol 28, 445-489. 146 CHAPTER FIVE Summary and Future Directions 147 Summary and Future Directions IRF protein family has nine members in mammals (Huang et al., 2010; Nehyba et al., 2009). All family members share a highly conserved N-terminus DBD and a less conserved protein-binding domain at the C-terminus (Kondo et al., 2002). The IRF family is known to orchestrate homeostasis of host defense. They are mandatory for regulation of type I interferon responses, development and function of a cadre of immune cell types, and cell cycle control and oncogenic pathogenesis (Tamura et al., 2008; Taniguchi et al., 2001). Irf6 is the only IRF known to be involved in skin and craniofacial development. Haploinsufficiency of human IRF6 cause two Mendelian clefting disorders (Kondo et al., 2002). Furthermore, a common DNA variant at the IRF6 locus contributes risk for isolated cleft lip and palate (Rahimov et al., 2008). Mice deficient for Irf6 display severe skin, limb, and craniofacial defects while heterozygosity for Irf6 null allele is associated with oral adhesions (Ingraham et al., 2006; Richardson et al., 2006). However, IRF6 shares some native functions of its family. IRF6 is involved in regulating the cell cycle with an anti-proliferative function in keratinocytes and mammary epithelial cell (Bailey et al., 2008; Ingraham et al., 2006; Richardson et al., 2006). Also, mutations in IRF6 have also been associated with oncogenesis (Bailey et al., 2009; Botti et al., 2011; Stransky et al., 2011). Regulation of the immune system, a major function of all family member, is yet undetermined for IRF6. Proteomic studies showed IRF6/IRF5 and IRF6/IRF8 as candidate protein complexes involved in regulating interferon type I. Despite DNA conservation of the DBD, and structural homology and interactions between IRF6 and more canonically described immune IRF family members, expression data of IRF6 in haematopoietic system is completely lacking. Cost and labor required for assessment of expression for a given gene through 148 out the whole haematopoietic system is devastating. We decided to use a bioinformatic approach to examine the expression of Irf6 thought out the developing stages of haematopoietic cells using the publically available whole transcriptomic data for almost every cell type. We found that Irf6 is expressed early in haematopoiesis specially in long term hematopoietic stem cells with abrupt attenuation of Irf6 expression in hematopoietic lineage committed progenitors. Common myeloid progenitor and myeloid erythroid progenitor isolated from old mice showed relative increase of Irf6. Also we identified Irf6 expression in T cell lineage, including developing and functionally committed stages. Future studies should confirm the expression of Irf6 in these predicted populations. Flowcytometric sorting of targeted populations from bone marrow, thymus and splenic suspensions followed by RT-PCR and western plot for Irf6 would be ideal. In our bioinformatic analysis, Variability of expression of in the same cell type among different experiments was sometimes striking. Unavoidable biological variability and using different probes in different microarray chips are valid reasons for variability. However non-linear noise signature of microarray experiments (also called batch effect) is another important factor that always hinders similar meta-analysis studies. Batch effect can be defined as the systematic error introduced when samples are processed in multiple batches. Several approaches have been developed for removing batch effects from microarray data (Scherer, 2009). However, having different cell types running in separate batches is the worst possible experimental design. Chen et al said “No way to correct for poor experimental design. If cases and controls are run in separate batches, genuine biological variation can be entirely confounded by batch effects. No method 149 was able to reduce the batch effects sufficiently without also removing the variation caused by case-control differences” (Chen et al., 2011). I am proposing a new approach to deal with similar conditions. If we can collect enough number of microarray profiles done in multiple batches for a given cell type, we can assume that only true biological data is shared among all profiles. Simple data decomposition approaches can discriminate between batch effects and biological variance. Once we identify the batch effect for every experiment, we can correct the profiles of other cell types tested in these experiments. Working on development of such technique that might allow us to correct for batch effects in cases of poor experimental design would open a new horizon for utilizing the tremendous resources of publically available data. Selective Expression of Irf6 in HSC and T cell lineage suggested its functional requirement. However perinatal mortality of Irf6 knockout mouse hindered our ability to study the role of Irf6 in the haematopoietic system (Ingraham et al., 2006). To test if expression of Irf6 expression is essentially required for development of these cells, Irf6 knockout fetal livers were adoptively transferred to lethally irradiated adult mice. Chimeric mice were able to recover and lymphocytes in their peripheral blood were mostly derived from donor stem cells. This indicates the ability of Irf6 knockout stem cells to replenish the bone marrow and develop into mature lymphocytes. Chimeric mice are particularly useful because they enable us to test the effect of Irf6 deficiency in adult mice. Furthermore, it enables the study of the gene effect in the whole haematopoietic system with the known extensive cell-cell interactions. Future studies should employ the Chimeric mice to explore the function of Irf6 in HSC. LTHSC should be able to repopulate the bone marrow for life while ST-HSC has a 150 repopulation potential not exceeding 8-12 weeks (Passegue et al., 2005). High level of Irf6 expression in LT-HSC suggests a possible role in long term engraftment studies. Chimeric mice should be followed up for more than 3 moths to detect later complete or partial failure of engraftment. One alternative approach to test for this is serial transplantation, in which donor HSCs are engrafted into a primary host then subsequently isolated and engrafted into secondary hosts (Weissman, 2000). Serial transplantation of HSC can replenish the recipient bone marrow in successive but limited transplants. The number of transplants correlates with the efficiency of HSC. Furthermore, the stem cell exhaustion in serial transplantations mimics an accelerated aging process, thus we can use this to test if deprivation of exhausted progenitors of Irf6 would accelerate their aging process (Ramkumar et al., 2013). Competitive repopulation is another assay to detect the minimal defects by mixing Irf6 deficient HSC with wild type HSC. Blood cells produced by each genotype should be proportional to the mixing ratio unless the Irf6 deficient progenitor is sub-optimally efficient for one or more lineage commitment (Harrison, 1980). To test for the effect of Irf6 expression in developing lymphocytes, we characterized the thymi in Irf6 knockout embryos at E17.5 in comparison to their wild type littermates. We found that Irf6 regulates proliferation in thymocytes where loss of Irf6 leads to an expansion of DN thymocytes and reduction of DP thymocytes. This effect is parallel to some extend to what Irf6 does in the skin. In contrast to the proapoptotic effect of Irf6 in the skin, we did not see change in the total no of apoptotic or dead cells. Further analysis for the no of apoptotic cells in the thymic sub-popualtion is mandatory to identify any hidden balanced disturbances. Our rate of apoptosis of 151 embryonic thymocytes is about 2% which comparable to those observed by others in adult mice (Ismael et al., 1998; Jung et al., 2004). However this rate of spontaneous apoptosis might not be able to detect saddle effect. Apoptosis induction experiments (e.g. Anti-CD3 inducted apoptosis) should be done to verify the effect of Irf6 deficiency on thymic apoptosis (Chrest et al., 1995). In vitro apoptosis assays can be used as well to test for activation-induced cell death by culturing thymocytes with anti-Fas, platebound anti-CD3, or dexamethasone for 24 h to induce apoptosis (Shui et al., 2007). We observed sustained counts of single positive thymocytes in spite of the reduction of their immediate precursors. This was not an effect of increased proliferation as shown by normal level of BrdU incorporation. We still have 2 possible explanations; enhanced survival of single positive cells and thymic retention of naïve lymphocytes. We need to test for the efficiency of negative selection to see how if it is reduced to compensate for lower rate of proliferation in DP cells. We can breed The Irf6 heterozygous mice onto a background expressing alloreactive TCR (e.g. BM3.6 transgenic mice). Efficient negative selection would eliminate all the DP thymocytes (Sponaas et al., 1994). To test the hypothesis of thymic retention, we can mate the Irf6gt/+ with Rag2p-GFP mice. Boursalian et al created these mice to identify recent emigrants of the thymus. GFP is expressed at a high level in DP thymocytes under the Rag2 promoter. Because GFP is a relatively stable protein, cells recently emigrating from the thymus can be identified by a “shoulder” of GFP expression (Boursalian et al., 2004). McCaughtry et al calculated the half-life of GFP protein in RAG2p-GFP transgenic mice to calculate the thymic retention time (McCaughtry et al., 2007). 152 We showed that Irf6 is expressed in the subcapsular cortex and medulla. However, we did not delineate if this expression is in the thymic epithelium or thymocytes or both. One of the important future directions is to determine the exact site of expression underlying the reported altered proliferation. One of the best genetic approaches would be the conditional knockouts. Floxed Irf6 strain can be crossed with different tissue specific Cre recombinase strains to induce Irf6 excision only in these target tissues. Lck-Cre and CD2-Cre transgenic mice are typically used to generate Tcell-specific conditional knockout mice. Regulatory sequences of the two stains drive the Cre recombinase enzyme expression very early in DN thymocytes, however CD2Cre target the B cells as well (Garvin et al., 1988; Wildin et al., 1991; Zhumabekov et al., 1995). CD4-Cre mouse is another transgenic strain which expresses the Cre recombinase under the control of a CD4 minigene. The CD4 minigene is composed of the proximal enhancer, the promoter and the silencer of CD4 gene. These regulatory sequences start the Cre expression in DN3 (CD44-CD25+) stage (Wolfer et al., 2001). To target developing thymocytes starting from the DP stage, Rorc-Cre strain would drive the expression of Cre recombinase in DP thymocytes and their single positive progeny whereas DN precursors would be untouched (Eberl & Littman, 2004). By the age of 6 month, we saw significant increase of CD4:CD8 ratio in Irf6 heterozygous mice. A more prolonged study is required to test the effect of Irf6 heterozygocity in older mice. We proposed impaired notch signaling in developing thymocytes mediating the altered CD4:CD8 ratio. Notch suppression can be assessed by measuring its canonical target gene HES1 transcription. 153 Adoptively transferred mice is another approach to discriminate between haematopoietic form non-haematopoietic mechanisms. In chimeric mice, the haematopoietic elements of the thymus should be Irf6 deficient while thymic epithelium would be wild type. Careful analysis should be considered because of the possible left over recipient Irf6 wild type cells. Also we should consider the possible differences between embryonic developmental events we are trying to replicate and the adult thymic microenvironment we have in chimeric mice. Whereas the adoptively transferred mice is a very suitable model to study the Irf6 dosage effect on the postnatal thymic changes of CD4:CD8 ratio. Future studies should be designed to determine the underlying molecular changes in Irf6 knockout thymocytes. Previous transcriptomic studies tried to figure out transcriptional networks regulated by Irf6 but non of them was completely successful. There are 3 microarray studies on mammalian tissues aimed to profile the transcriptional signature of IRF6. The earliest study examined murine knockout skin. An important caveat is that skin includes both the epidermis and dermis. Considering that Irf6 is only expressed in the epidermis, the dermal tissue added unavoidable noise to the expression profile. The other studies were done by knocking down IRF6 in primary human keratinocytes and erythroid progenitors (Botti et al., 2011; Xu et al., 2012). Silencing efficiency limits the generalizability of these results. Microarray experiment utilizing a homogenous cell population and a knockout model would be the most sensitive assays at detecting transcriptional regulation by IRF6. We need to identify beyond doubt the primary cell type causing the altered proliferation of developing 154 thymocytes. The ease of sorting the thymic population would provide an almost homogenous population where transcriptional analysis would be very valuable. Common variants in IRF6 contribute risk toward orofacial clefting. Rare variants in IRF6 lead to Van der Woude and Popliteal Pterygium Syndromes. This data suggests that individuals with common and rare IRF6 variants are at increased risk for immunological diseases. Future studies should be designed to screen for the neonatal lymophocyte counts in those patients. Furthermore, old patients with Irf6 variants should be tested for early thymic involution. Irf family members are either indispensable for normal T helper differentiation or have known variants associated with autoimmune disorders of T cells (Lohoff & Mak, 2005; Patel, 2011). We tried to utilize the publically available microarray studies to suspect the possible roles of Irf6 in T-helper commitment. We predicted a possible role of intrinsic Irf6 expression in induction of Th17 differentiation on the expense of Treg differentiation. Also we expected that Irf6 regulates the DC functions to mediate a noncell autonomous polarization of Th1. To test our hypothesis about the necessity of intrinsic Irf6 expression for regulation of T helper differentiation, we adopted an in vitro differentiation model. Naive T cells lacking Irf6 could be differentiated into Th1, Th2, Th17 and Treg using a specific cytokine cocktail. This experiment proves for the first time that intrinsic Irf6 expression is not essential for T helper subset differentiation. There is no difference in cell frequency and mean fluorescence intensity between wild type and Irf6 knockout cells. However, the sensitivity of the in vitro model to detect saddle changes is questionable. The 155 concentrations of cytokines used in culture to induce differentiation are too high to saturate the signaling pathways. Also the signaling pathways essential for in vitro differentiation are not always the same those invoked in vivo. For example, IL-4 is essential for in vitro Th2 differentiation and naive T cells with mutant genes in the IL4/STAT6 signaling pathway can not produce Th2 cells. By contrast, in vivo Th2 differentiation can occur in mice that have deletions in IL-4, IL-4Rα or STAT6 (van Panhuys et al., 2008). In the future, in vivo differentiation analysis of Irf6 deficient T cells should complement our current work. Adoptively transferred mice can be used in these experiments; however it will not be able to differentiate between the autonomous T cell defects and non-autonomous defects mediated by other Irf6 deficient haematopoietic cells. The lineage specific conditional knockout would be the best model for such experiments. Irf6 floxed strain needs to be crossed with Cre transgenic mice expressing the recombinase enzyme under the regulatory sequences of lineage specific promoters. Tbet-Cre, Gata3-Cre, RORγt-Cre, and Foxp3-Cre have been published before (Eberl & Littman, 2004; Francius et al., 2013; Haddad et al., 2013; Rubtsov et al., 2008). For the analysis of the possible role of Irf6 in DC functions, we tested the bone marrow of adoptively transferred mice for DC populations. Irf6 deficiency has no effect total DC count in BM. In vitro differentiation of BM in GM-CSF enriched medium, showed the same differentiation pattern of DCs of both wild type and Irf6 knock chimeric mice. There is a trend toward increased MHC-II expression in knockout CD11b+ DCs that became statistically significant at culture day 3. This finding supports our bioinformatic suggestion for a non-cell autonomous role of Irf6 in T cell differentiation through dendritic cells. Future studies should cover detailed analysis of DC functions 156 under Irf6 deficiency. In vitro differentiated DCs should be tested for cytokine production including IL6, IL12p40 and IL10. Flowcytometric analysis if intracellular cytokines is on approach and direct measurement of cytokines in culture supernatants is another one (Said et al., 2014). Migration assay is another important functional determination. The assay is usually done in transwell plates where activated DCs are cultured in the upper wells and allowed to migrate to the lower wells under the guidance of chemotactic agents e.g. CCL5 and CCL21 (Gibbs et al., 2013). T cell co-culture is one of the most canonical experiments to assess the antigen presenting capacity of DCs. To standardize the response, MHC restricted T cells with antigen specific TCRs are used for co-culture. For example, MHC class I-restricted, ovalbumin-specific, CD8+ T cells can be co-cultured with the target DCs after being pulsed with OVA peptide. The Coculture should be harvested after 4 days and CD8 T cell proliferation and IFN gamma secretion would be measured (Clarke et al., 2000). Langerhans cells (LC) are distinguishable from other DC populations. LCs are characterized by lower MHC-II levels, and very high levels of the C-type lectin langerin (Merad et al., 2008). Irf8 controls the trafficking of LCs to the regional lymph nodes (Schiavoni et al., 2004) while Irf2 is required to maintain normal counts of LCs (Ichikawa et al., 2004). Our bioinformatic analysis suggests DCs as the main source of IL8 deficiency in Irf6 knockout skin. If this holds true, Irf6 would have an important non-cell autonomous role in T cell differentiation. Considering that Irf6 function has most clearly been delineated in skin, specific analysis LCs should be considered in future studies. Most LCs develop from fetal liver monocytes and self-renew throughout life independently from the BM (Hoeffel et al., 2012; Merad et al., 2002). Importantly, with 157 severe LC depletion, LCs are repopulated by blood-borne monocytes (Ginhoux et al., 2006). These data indicate that we can study Irf6 deficient LCs in our adoptively transfer mice. One technical problem that usually faces the analysis of LCs is the contamination of cell preparations with significant numbers of conventional dermal dendritic cells. Initial digestion of the skin with dispase enzyme allows mechanical separation of epidermis without any attached dermal component. Trypsinization of epidermal cells allows the formation of single cell suspension for subsequent sorting of LCs (Pena-Cruz et al., 2001). Culturing and functional studies can be done the same way as described above with conventional DCs. In conclusion, this research tried for the first time to connect Irf6 to the canonical roles of the other family members in the immune system. We made use of publically available microarray data to predict the possible roles of Irf6 throughout the haematopoietic system. Successfully, we were able to confirm a novel role of Irf6 in the proliferation of developing thymocytes. Unlike other family members, intrinsic Irf6 expression is not essential for T helper subset differentiation. However, a non-cell autonomous role for Irf6 in T cell differentiation through dendritic cells remains plausible. 158 REFERENCES 159 REFERENCES Bailey, C. M., Abbott, D. E., Margaryan, N. V., Khalkhali-Ellis, Z. & Hendrix, M. J. (2008). Interferon regulatory factor 6 promotes cell cycle arrest and is regulated by the proteasome in a cell cycle-dependent manner. Mol Cell Biol 28, 2235-2243. Bailey, C. M., Margaryan, N. V., Abbott, D. E., Schutte, B. C., Yang, B., KhalkhaliEllis, Z. & Hendrix, M. J. (2009). Temporal and spatial expression patterns for the tumor suppressor Maspin and its binding partner interferon regulatory factor 6 during breast development. Dev Growth Differ 51, 473-481. Botti, E., Spallone, G., Moretti, F. & other authors (2011). Developmental factor IRF6 exhibits tumor suppressor activity in squamous cell carcinomas. Proc Natl Acad Sci U S A 108, 13710-13715. Boursalian, T. E., Golob, J., Soper, D. M., Cooper, C. J. & Fink, P. J. (2004). Continued maturation of thymic emigrants in the periphery. Nat Immunol 5, 418-425. Chen, C., Grennan, K., Badner, J., Zhang, D., Gershon, E., Jin, L. & Liu, C. (2011). Removing batch effects in analysis of expression microarray data: an evaluation of six batch adjustment methods. PLoS One 6, e17238. Chrest, F. J., Buchholz, M. A., Kim, Y. H., Kwon, T. K. & Nordin, A. A. (1995). AntiCD3-induced apoptosis in T-cells from young and old mice. Cytometry 20, 33-42. Clarke, S. R., Barnden, M., Kurts, C., Carbone, F. R., Miller, J. F. & Heath, W. R. (2000). Characterization of the ovalbumin-specific TCR transgenic line OT-I: MHC elements for positive and negative selection. Immunol Cell Biol 78, 110-117. Eberl, G. & Littman, D. R. (2004). Thymic origin of intestinal alphabeta T cells revealed by fate mapping of RORgammat+ cells. Science 305, 248-251. Francius, C., Harris, A., Rucchin, V. & other authors (2013). Identification of multiple subsets of ventral interneurons and differential distribution along the rostrocaudal axis of the developing spinal cord. PLoS One 8, e70325. Garvin, A. M., Pawar, S., Marth, J. D. & Perlmutter, R. M. (1988). Structure of the murine lck gene and its rearrangement in a murine lymphoma cell line. Mol Cell Biol 8, 3058-3064. Gibbs, S., Spiekstra, S., Corsini, E., McLeod, J. & Reinders, J. (2013). Dendritic cell migration assay: a potential prediction model for identification of contact allergens. Toxicol In Vitro 27, 1170-1179. 160 Ginhoux, F., Tacke, F., Angeli, V., Bogunovic, M., Loubeau, M., Dai, X. M., Stanley, E. R., Randolph, G. J. & Merad, M. (2006). Langerhans cells arise from monocytes in vivo. Nat Immunol 7, 265-273. Haddad, R., Lanjuin, A., Madisen, L., Zeng, H., Murthy, V. N. & Uchida, N. (2013). Olfactory cortical neurons read out a relative time code in the olfactory bulb. Nat Neurosci 16, 949-957. Harrison, D. E. (1980). Competitive repopulation: a new assay for long-term stem cell functional capacity. Blood 55, 77-81. Hoeffel, G., Wang, Y., Greter, M. & other authors (2012). Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J Exp Med 209, 1167-1181. Huang, B., Qi, Z. T., Xu, Z. & Nie, P. (2010). Global characterization of interferon regulatory factor (IRF) genes in vertebrates: glimpse of the diversification in evolution. BMC Immunol 11, 22. Ichikawa, E., Hida, S., Omatsu, Y., Shimoyama, S., Takahara, K., Miyagawa, S., Inaba, K. & Taki, S. (2004). Defective development of splenic and epidermal CD4+ dendritic cells in mice deficient for IFN regulatory factor-2. Proc Natl Acad Sci U S A 101, 3909-3914. Ingraham, C. R., Kinoshita, A., Kondo, S. & other authors (2006). Abnormal skin, limb and craniofacial morphogenesis in mice deficient for interferon regulatory factor 6 (Irf6). Nat Genet 38, 1335-1340. Ismael, S. J., Callera, F., Garcia, A. B., Baffa, O. & Falcao, R. P. (1998). Increased dexamethasone-induced apoptosis of thymocytes from mice exposed to long-term extremely low frequency magnetic fields. Bioelectromagnetics 19, 131-135. Jung, K. C., Park, W. S., Kim, H. J., Choi, E. Y., Kook, M. C., Lee, H. W. & Bae, Y. (2004). TCR-independent and caspase-independent apoptosis of murine thymocytes by CD24 cross-linking. J Immunol 172, 795-802. Kondo, S., Schutte, B. C., Richardson, R. J. & other authors (2002). Mutations in IRF6 cause Van der Woude and popliteal pterygium syndromes. Nat Genet 32, 285289. Lohoff, M. & Mak, T. W. (2005). Roles of interferon-regulatory factors in T-helper-cell differentiation. Nat Rev Immunol 5, 125-135. McCaughtry, T. M., Wilken, M. S. & Hogquist, K. A. (2007). Thymic emigration revisited. J Exp Med 204, 2513-2520. 161 Merad, M., Manz, M. G., Karsunky, H., Wagers, A., Peters, W., Charo, I., Weissman, I. L., Cyster, J. G. & Engleman, E. G. (2002). Langerhans cells renew in the skin throughout life under steady-state conditions. Nat Immunol 3, 1135-1141. Merad, M., Ginhoux, F. & Collin, M. (2008). Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat Rev Immunol 8, 935-947. Nehyba, J., Hrdlickova, R. & Bose, H. R. (2009). Dynamic evolution of immune system regulators: the history of the interferon regulatory factor family. Mol Biol Evol 26, 2539-2550. Passegue, E., Wagers, A. J., Giuriato, S., Anderson, W. C. & Weissman, I. L. (2005). Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. J Exp Med 202, 1599-1611. Patel, D. R. (2011). Linking interferon regulatory factors to the pathogenesis of human lupus. Transl Res 157, 323-325. Pena-Cruz, V., Ito, S., Oukka, M., Yoneda, K., Dascher, C. C., Von Lichtenberg, F. & Sugita, M. (2001). Extraction of human Langerhans cells: a method for isolation of epidermis-resident dendritic cells. J Immunol Methods 255, 83-91. Rahimov, F., Marazita, M. L., Visel, A. & other authors (2008). Disruption of an AP2alpha binding site in an IRF6 enhancer is associated with cleft lip. Nat Genet 40, 13411347. Ramkumar, C., Gerstein, R. M. & Zhang, H. (2013). Serial transplantation of bone marrow to test self-renewal capacity of hematopoietic stem cells in vivo. Methods Mol Biol 976, 17-24. Richardson, R. J., Dixon, J., Malhotra, S., Hardman, M. J., Knowles, L., BootHandford, R. P., Shore, P., Whitmarsh, A. & Dixon, M. J. (2006). Irf6 is a key determinant of the keratinocyte proliferation-differentiation switch. Nat Genet 38, 13291334. Rubtsov, Y. P., Rasmussen, J. P., Chi, E. Y. & other authors (2008). Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28, 546-558. Said, A., Bock, S., Muller, G. & Weindl, G. (2014). Inflammatory conditions distinctively alter immunological functions of Langerhans-like cells and dendritic cells in vitro. Immunology. Scherer, A. (2009). Batch Effects and Noise in Microarray Experiments: Sources and Solutions: John Wiley and Sons. 162 Schiavoni, G., Mattei, F., Borghi, P., Sestili, P., Venditti, M., Morse, H. C., 3rd, Belardelli, F. & Gabriele, L. (2004). ICSBP is critically involved in the normal development and trafficking of Langerhans cells and dermal dendritic cells. Blood 103, 2221-2228. Shui, J. W., Hu, M. C. & Tan, T. H. (2007). Conditional knockout mice reveal an essential role of protein phosphatase 4 in thymocyte development and pre-T-cell receptor signaling. Mol Cell Biol 27, 79-91. Sponaas, A. M., Tomlinson, P. D., Antoniou, J., Auphan, N., Langlet, C., Malissen, B., Schmitt-Verhulst, A. M. & Mellor, A. L. (1994). Induction of tolerance to self MHC class I molecules expressed under the control of milk protein or beta-globin gene promoters. Int Immunol 6, 277-287. Stransky, N., Egloff, A. M., Tward, A. D. & other authors (2011). The mutational landscape of head and neck squamous cell carcinoma. Science 333, 1157-1160. Tamura, T., Yanai, H., Savitsky, D. & Taniguchi, T. (2008). The IRF family transcription factors in immunity and oncogenesis. Annu Rev Immunol 26, 535-584. Taniguchi, T., Ogasawara, K., Takaoka, A. & Tanaka, N. (2001). IRF family of transcription factors as regulators of host defense. Annu Rev Immunol 19, 623-655. van Panhuys, N., Tang, S. C., Prout, M., Camberis, M., Scarlett, D., Roberts, J., HuLi, J., Paul, W. E. & Le Gros, G. (2008). In vivo studies fail to reveal a role for IL-4 or STAT6 signaling in Th2 lymphocyte differentiation. Proc Natl Acad Sci U S A 105, 12423-12428. Weissman, I. L. (2000). Stem cells: units of development, units of regeneration, and units in evolution. Cell 100, 157-168. Wildin, R. S., Garvin, A. M., Pawar, S., Lewis, D. B., Abraham, K. M., Forbush, K. A., Ziegler, S. F., Allen, J. M. & Perlmutter, R. M. (1991). Developmental regulation of lck gene expression in T lymphocytes. J Exp Med 173, 383-393. Wolfer, A., Bakker, T., Wilson, A. & other authors (2001). Inactivation of Notch 1 in immature thymocytes does not perturb CD4 or CD8T cell development. Nat Immunol 2, 235-241. Xu, J., Shao, Z., Glass, K. & other authors (2012). Combinatorial Assembly of Developmental Stage-Specific Enhancers Controls Gene Expression Programs during Human Erythropoiesis. Dev Cell 23, 796-811. 163 Zhumabekov, T., Corbella, P., Tolaini, M. & Kioussis, D. (1995). Improved version of a human CD2 minigene based vector for T cell-specific expression in transgenic mice. J Immunol Methods 185, 133-140. 164 CHAPTER SIX Role of endogenous Avian Leukosis Virus in the pathophysiology of spontaneous ALV-like tumors in chickens 165 Abstract Lymphoid leukosis (LL) is a B cell tumor of the chicken bursa with a huge economic impact on the poultry industry. The tumors are induced by infection with Avian Leukosis Virus (ALV). However, endogenous ALVs is known to have little or no oncogenic potential. Marek’s disease (MD) is another viral neoplastic disorder with hogh mortality rate in chicken. Serotype 2 MD vaccine (MDV2) is an attenuated virus and naturally non-oncogenic but has been shown to enhance the development of both exogenous ALV-induced and spontaneous lymphoid leukosis. AF-227 is a new field strain of subgroup E endogenous ALV (ALV-E). AF-227 has been isolated from commercial chicken experiencing spontaneous ALV-like LL. Although ALV-E viruses are known to be non-oncogenic, the influence of ALV-E and its possible interaction with MDV2 on the enhancement of spontaneous ALV-like LL are still unclear. In this study we used RNA-Seq to generate an expression profile from spontaneous ALV-like LL obtaine from chickens inoculated with strain AF-227 of ALV-E in conjunction serotype 2 MD vaccine to uncover potential molecular oncogenic events.. We identified the absence of Tumor suppressor candidate 2 (TUSC2 ) in all tumor samples. TUSC2 is a tumor suppressor gene which is considered as a molecular link between inflammatory response and mitochondrial homeostasis (Hood et al., 2013; Uzhachenko et al., 2012). TUSC2 can influence and complement the PI3K/AKT and p53 pathways. Our pathway enrichment analysis showed significant dysregulation of both pathways. We also identified overexpression of another important proto-oncogene called Eukaryotic translation initiation factor 4E (EIF4E). While EIF4E overexpression could be a downstream effect of TUSC2 loss and abnormal Akt signaling, another possible 166 scenario is the convergence of the both genetic defects to induce the B cell transformation. In this work we identified candidate molecular targets of LL in chickens infected by a new endogenous ALV in conjunction with MDV2. 167 Introduction Lymphoid leukosis (LL) is a B-cell lymphoma of chickens. LL usually appears in chickens of about 4 months of age and older. Tumors typically involve the liver, spleen, and bursa of Fabricius (Fadly & Nair, 2008). Tumors are usually composed of aggregates of lymphoblasts of B-cell origin and characterized by monoclonal production of IgM (Payne & Rennie, 1975). The primary lesion usually presents as a well defined mass at the site of the bursa, but at a time the normal bursa tissue is usually gone (Fadly & Nair, 2008). LL can be induced by transmissible strains of retroviruses called Avian Leukosis Virus (ALV). These strains are defined as exogenous for being transmitted as infectious virus particles. Exogenous ALVs multiply in most tissues and organs of the body but the infection persists longer in bursal lymphocytes, the target cells of neoplastic transformation (Baba & Humphries, 1985). Exogenous non-defective ALVs do not harbor any oncogene; they have been shown to induce lymphoid leukosis by activation of cellular myc oncogene. Only Defective exogenous ALV harbor an oncogene such vmyc, v-src, v-myb, etc. and have been shown to induce acute tumors in susceptible host (Fadly & Nair, 2008). On the other hand, spontaneous ALV-like LL has been shown to develop in certain lines of chickens at one year of age or older. These spontaneous ALV-like LL tumors are detected in certain genetic lines of chickens in absence of infection with any of the subgroups of exogenous ALV, and hence the name spontaneous is used to describe such tumors (Crittenden et al., 1979) 168 Based on envelope glycoproteins, ALVs that occur in chickens are classified into six groups, A, B, C, D, E and J (Payne et al., 1991; Vogt, 1997; Weiss et al., 1982). Unlike exogenous ALVs, subgroup E viruses are avian retrovirus-like elements that are transmitted genetically in a Mendelian fashion and are termed endogenous viruses (Fadly & Nair, 2008). Domestic chicken genome carries at least 16 endogenous ALV proviral loci (ev-1 through ev-16) (Rovigatti & Astrin, 1983). Many endogenous viruses are genetically defective and incapable of giving rise to infectious virions (Crittenden & Astrin, 1981), whereas others are not and may be expressed in an infectious form (Crittenden et al., 1983). In this form, they then are transmitted similarly to exogenous viruses, although most chickens are genetically resistant to such exogenous infection (Fadly & Nair, 2008). Rous-associated virus type-0 (RAV-0), a subgroup E endogenous virus had little or no oncogenic potential (Motta et al., 1975). However RAV-60, subgroup E recombinants of endogenous and exogenous viruses enhanced the development of lymphoid leukosis (Crittenden et al., 1980; Robinson et al., 1980). Endogenous ALVs also influence the response of the bird to infection by exogenous ALV (Crittenden et al., 1982; Smith & Fadly, 1988). Recently, a field strain of endogenous ALV-E, termed AF-227 was isolated from blood from commercial chickens. The ability of serotype 2 Marek's disease virus serotype 2 (MDV2) to enhance the development of LL after ALV exposure was reported (Bacon et al., 1989; Fadly & Witter, 1993). Bursa cells co-infected with ALV and MDV2 are more likely to be transformed (Fynan et al., 1992). MDV2 was shown to increase ALV gene expression and virus production (Pulaski et al., 1992). This interaction presents a huge danger because it holds true with the marek’s vaccine attenuated viruses (Marsh et al., 1995). 169 Furthermore, vaccination of exogenous ALV-free chicken against Marek's disease increased the chance of developing LL (Crittenden et al., 1979). The interaction of MDV with endogenous ALVs in these birds is not well understood. In this study, in order to uncover the molecular oncogenic events of spontaneous ALV-like LL, we used RNASeq to generate an expression profile from tumor tissues collected from chickens embryonically inoculated with strain AF-227 of ALV-E and vaccinated with MDV2 at hatch. Materials and methods Experimental treatment of chicken: RFS is a strain of chicken that is free of endogenous virus but is susceptible to ALV infection (Zhang et al., 2005). Studied chicken were categorized into 4 groups; control group of 24 chickens injected with phosphate buffered saline (PBS), 36 chickens injected with AF227 alone, 24 chickens treated with MDV2 vaccine alone, and 36 chickens treated with AF227 plus MDV2 vaccination. The PBS and AF227 were given via yolk-sac inoculation at 7 days of embryonation. The MDV2 vaccination was given at hatch intraabdominally. Chickens were followed up closely for the appearance of the tumors at 46 month of age. After euthanasia, tumor samples were collected as well circumscribed masses from the anatomical site of the bursa. Next generation sequencing of RNA: Six tumor samples from the group treated simultaneously with AF-227 inoculation and MDV2 vaccination were used for the 170 analysis. As controls, we collected 3 whole bursa samples from 3-week old chickens, and we collected 3 B cell samples from spleens of age matched chicken. We extracted mRNA from tissue homogenates as recommended (Qiagen, RNeasy Kit, Valencia, CA). RNA-seq libraries were prepared using the Illumina TruSeq Stranded mRNA kit following the manufacturer's instructions (Illumina, San Diego, CA). Sequencing was performed on an Illumina HiSeq 2500, run in High Output mode in a paired end 2x100bp format using Illumina TruSeq PE Cluster Kit (v3) and TruSeq SBS Kit (v3). Samples were divided on 3 lanes (4 samples per lane). Raw reads were filtered to remove adaptors, ambiguous reads and low-quality reads. Approximately 661 million clean paired reads (132 Gbp) were obtained (Table 6.1). 171 Lane R1 PF % per clusters lane Sample R2 R1 Ave R2 Ave Insert Yield %≥ %≥ Q- Q- Size (Gbp) Q30 Q30 Score Score (bp) 1 Tumor 1 49,249,019 21.6 91.1 68.1 35.7 27.9 484 9.85 1 Tumor 2 37,972,904 16.6 91.0 69.3 35.6 28.4 521 7.59 1 Tumor 3 93,542,998 41.0 91.0 67.9 35.6 27.9 506 18.71 1 Tumor 4 43,807,926 19.2 91.0 68.8 35.6 28.2 530 8.76 2 Tumor 5 38,709,590 17.2 88.4 62.2 34.9 26.0 547 7.74 2 Tumor 6 90,399,233 40.1 87.9 62.1 34.7 25.9 503 18.08 2 B cells 1 52,096,499 23.1 87.7 62.3 34.7 26.0 495 10.42 2 B cells 2 40,697,155 18.1 86.2 58.1 34.2 24.5 540 8.14 3 B cells 3 72,903,292 33.2 86.5 62.2 34.3 26.1 542 14.58 3 Bursa 1 76,772,884 35.0 86.1 61.3 34.2 25.7 541 15.35 3 Bursa 2 34,194,285 15.6 87.0 64.0 34.5 26.7 524 6.84 3 Bursa 3 30,856,288 14.0 86.4 63.0 34.3 26.3 532 6.17 Total 661,202,073 132.2 Table 6.1: Yield and quality of NGS. PF clusters: Paired end clusters passing filter. R1: first strand read (the antisense or 3’ read). R2: The sense or the 5’ read. Q-scores: quality score (A property that is logarithmically related to the base call error probability P where Q=-10 Log10 P). Q30: quality score of 30 172 Bioinformatic analysis: Initial exploration of the data quality was done using the FASTX toolkit (FASTX Toolkit by Hannon lab). Quality checking showed abnormally rapid deterioration of the quality scores of the reverse reads (Figure 6.1a). Also there was abnormal distribution of the nucleotide content of the initial 13 bases of either Percentage Quality score forward or revere reads (Figure 6.1b). Position in read (bp) Position in read (bp) Figure 6.1: Snap shot from the FASTX toolkit analysis. (a) Quality score of the reverse reads samples. (b) Sequence content across all bases Trimmomatic software was used with its palindromic approach to remove readthrough adaptor sequences and isolate orphans from true paired reads (Bolger et al., 2014). Simultaneous head cropping of the first 13 bases was done. Three approaches of data processing were compared to identify the optimal gene model with minimal loss of data. The first two approaches used the Trimmomatic built-in functions. 1) The SlidingWindow method scans from the 5’ end and clips the read once the average quality within the window falls below a threshold. 2) The MaxInfo method is an adaptive 173 quality trimmer that balances read length and error rate to maximize the value of each read. Short reads (less than 36 bases) were excluded after both trimming approaches. 3) We used the pre-processing method (FASTX toolkit) which trims the last 20 bases then reads were filtered out if they fell below a threshold. TopHat v2.0.10 Software was used to map the reads back to the chicken genome (Kim et al., 2013). The most updated versions of the chicken genome assembly and annotation (Galgal4, release 74) were obtained from the ensemble Genome Browser (ftp.ensembl.org/pub/release-74). Ensemble annotation was preferred over the reference annotation of the Genebank because it covered more genes in our transcriptome (Figure 6.2). Figure 6.2: Snap shot of a genome browser (IGV) comparing the annotations of different assemblies. A representative stretch of the Genebank reference annotation (1st lane), ensemble annotation (2nd lane), and our assembly (3rd lane). The ensemble annotation covered our transcriptome more completely. 174 One of the known issues about the current chicken assembly is the possibility of replication of some genes between chromosomal and non-chromosomal sequences. To ensure appropriate alignment of the reads to their corresponding genes, we performed two rounds of mapping. For the first round, we mapped all reads against the chromosomal sequences only and limited the initial transcriptome indexing to the chromosomal annotation. The unmapped reads were used for another round of assembly using the non-chromosomal sequences and annotation. Transcriptome assembly and subsequent differential expression analysis was done using the Cufflinks package v2.1.1 as shown before (Figure 6.3) (Trapnell et al., 2012) Figure 6.3: Diagrammatic representation of the TopHat/Cufflinks analysis (Trapnell et al., 2012) 175 Downstream clustering analysis was done using R software. For pathway analysis, DAVID annotation tool was used (Figure 6.4 summarizes the whole pipeline). Figure 6.4: Work flow for the pipeline of NGS analysis 176 Results Tumor incidence in the studied groups: In the experiment, there are four experimental groups; PBS control group, a group injected with AF227 alone, a group treated with MDV2 vaccine alone, and a group treated with AF227 plus MDV2 vaccination. The incidence of LL-like tumors was 8% in the PBS control group, 14% with chicken injected with AF227 alone, 17% in the group of MDV2 vaccination alone, and 42% in the group treated with both AF227 injection and MDV2 vaccination. The performance of different preprocessing approaches: The MaxInfo approach caused the minimal loss of data during pre-processing. This allowed the preservation of the highest number of paired ended clusters. However most of these data were lost during the mapping with TopHat. The SlidingWindow and Tail trimming approaches were almost equal when we compared the no of dropped reads after quality filtering. However, SlidingWindow was more successful in keeping longer reads giving the highest average read length after pre-processing. Furthermore, SlidingWindow showed a slightly better sensitivity compared to the other two techniques. Interestingly, the SlidingWindow approach was the least no to map reads outside the annotated genome giving the smallest number of isoforms of new genes. This allowed the SlidingWindow approach to achieve the highest locus specificity (Table 6.2). Considering the quality of the assembly and the accompanying annotation, we recognize that this comparison is not fair and we do not have the appropriate control to judge the actual specificity of 177 these techniques. In this analysis, our original plan did not include identification of new genes so adding more non-annotated transcripts to our assembly would do nothing but decreasing the statistical power. Thus we selected the SlidingWindow technique to be our preprocessing approach. Future study to compare pre-processing approaches should consider using of a simulated data. 178 MaxInfo SlidingWindow Tail trimming Statistics after sample pre-processing paired end 75,377,436 59,487,520 57,131,448 singletons 280,950 7,676,613 8,536,476 75,658,386 67,164,133 65,667,924 5,583,750,709 5,493,544,077 4,596,335,379 74 82 70 Total reads # of bases Average read length no of alignments and unmapped reads after TopHat analysis no of alignments 44,900,716 42,669,372 41,734,076 unmapped-qc passed 31,102,767 25,340,970 25,375,868 31,536 5,115 11,067 76,035,019 68,015,457 67,121,011 properly paired 22,512,228 20,867,419 18,671,936 improperly paired 16,213,828 13,747,268 13,719,707 6,174,660 8,054,685 9,342,433 unmapped-qc failed Total Quality of mapping singletons Comparison to reference annotation (Cuffcompare stat) All assembled isoforms 48,353 48,609 51,395 Ref isoform: Complete match 12,393 12,465 12,438 7,174 5,852 7,539 Locus Sensitivity 78.7 79 78.8 Locus Specificity 51 57.6 52.2 Isoforms of new genes Table 6.2: Comparing the performance of different preprocessing approaches. Results shown represent the smallest sample in size (Tumor sample 2). 179 Expression profile of Tumor samples is closer to bursa tissue than mature splenic B cells: The normalized read counts mapped to cufflink genes were used for analysis. Testing the rate of gene expression across all the studied samples showed unequal depth of sequencing (Figure 6.5a). To overcome the sequencing depth effect, genes that showed up uniquely due to high sequencing depth were excluded. Ward Hierarchical Clustering was done using Euclidean distance to generate the distance matrix. The new clustering analysis showed low biological variability between samples of the same biological origin and also showed a clear distinction of tumor and bursa samples from splenic B cells (Figure 6.5b) 180 Figure 6.5a: Boxplot of expression profiles Figure 6.5b: Ward Hierarchical Clustering using Euclidean distance 181 Differential expression analysis: We performed pairwise comparisons for each of the three groups. Significant differential expression was considered with p value < 0.05 after correction for multiple analyses (Figure 6.6). To identify genes relevant to tumorigenesis, we chose genes that showed significant change in tumor tissues when compared to both bursa and splenic B cells (385 went up and 395 went down; supplementary table A2 and A3). Among the differentially expressed genes TUSC2 is a tumor suppressor gene that was completely lost in all tumor samples but was the highest expressed gene in both bursa and splenic B cells (Kondo et al., 2001; Meng et al., 2013). On the other hand, the oncogene EIF4E ranked 2nd among the up regulated genes in malignant samples compared to being undetected in the other tissues (Carroll & Borden, 2013; Culjkovic-Kraljacic et al., 2012; Mamane et al., 2004) 182 Figure 6.6: Differential expression Analysis: The differentially expressed genes between every two studied tissues are counted in a circle. (a) MvsB.sign: Significant differentially expressed genes between malignant and bursa tissue. (b) MvsS.sign: Significant differentially expressed genes between malignant and splenic B cells. (c) SvsB.sign: Significant differentially expressed genes between splenic B cells and bursa tissue. Intersections count the genes that appeared significant in more than one comparison 183 Pathway enrichment analysis: For pathway analysis, significant hits were further enriched by moving the p value threshold to 0.01 and excluding genes with low level of expression (20 FPKM was used as an arbitrary cut off). With these criteria we had 228 differentially expressed genes (144 went up and 84 went down). To overcome the poor annotation of chicken genome and low resources of their genetic pathways, we identified 218 human orthologs. KEGG pathway analysis showed significant enrichment of several cancer pathways (Table 6.3). PANTHER Pathway analysis ensured the involvement of p53 signaling pathway. KEGG Pathways PValue Genes Prostate cancer 0.013 E2F3, PIK3CD, PIK3R5, RB1,TCF7L2, CTNNB1 p53 signaling pathway 0.023 BID, CD82, FAS, CCNG1, THBS1 Chronic myeloid leukemia 0.031 E2F3, HDAC1, PIK3CD, PIK3R5, RB1 Small cell lung cancer 0.044 E2F3, ITGA6, PIK3CD, PIK3R5, RB1 Alzheimer's disease 0.045 BID, APP, PSEN1, SDHD, IL1B, FAS, ATP5A1 Apoptosis 0.049 BID, PIK3CD, IL1B, PIK3R5, FAS Endometrial cancer 0.049 PIK3CD, PIK3R5, TCF7L2, CTNNB1 Non-small cell lung cancer 0.054 E2F3, PIK3CD, PIK3R5, RB1 Cytosolic DNA-sensing 0.057 POLR1D, IL18, IL1B, IRF3 Cell adhesion molecules 0.058 ALCAM, SDC1, PTPRF, ITGA6, PVRL3, SDC4 Pathways in cancer 0.075 BID, E2F3, HDAC1, ITGA6, PIK3CD, PIK3R5, RB1, FAS, TCF7L2, CTNNB1 Table 6.3: Pathway enrichment analysis (DAVID Functional annotation tool 6.7) 184 Table 6.3 (Cont’d): Toll-like receptor signaling 0.077 MAP2K3, PIK3CD, IL1B, PIK3R5, IRF3 Glioma 0.078 E2F3, PIK3CD, PIK3R5, RB1 PANTHER Pathways PValue Genes E2F3, PIK3CD, PIK3R5, RB1, CCNG1, p53 feedback loops 2 0.000 TPTE, CTNNB1 E2F3, HDAC1, CD82, PIK3CD, PIK3R5, FAS, p53 pathway 0.000 CCNG1, THBS1, TPTE 185 Discussion Avian tumor viruses of economic importance include Marek’s disease virus and avian retroviruses, namely avian leukosis virus (ALV) and reticuloendotheliosis virus (Witter, 1997). Avian retroviruses are associated with neoplastic diseases that represent a serious burden in poultry industry. Lympohoid leukousis is a B cell tumor that usually arises from the bursa tissue and is known to be enhanced by ALV (Baba & Humphries, 1985). ALV mortality and morbidity has a huge economic burden estimated to be in millions of U.S. dollars each year (Fadly & Nair, 2008). Marek’s vaccine is routinely used in poultry industry; however the attenuated vaccine strains were shown to enhance the development of LL after ALV exposure (Fynan et al., 1992; Marsh et al., 1995). Understanding the molecular events of oncogenesis mediated by interactions of ALV and MDV2 is critical for development of better strategies of prevention and control. The development of the RFS, a strain of chicken which is endogenous virus free but still susceptible to endogenous AVL infection, represents a proper negative control to the virally infected chicken. However, the late onset of the disease was an obstacle for the study design. By the time the tumors develop, there was no visible bursa tissue in the non-infected controls. To have the appropriate control, we compared the tumor samples to bursa tissues from 3 month old chicken as well as B cells sorted from the spleens of the age matched chicken. Bursa of Fabricius is known as the primary site of LL (Fadly & Nair, 2008). In our analysis, the phylogenic convergence of tumor samples with bursa samples confirms the bursa as a tissue of origin for these tumors. The homogeneity of tumor samples 186 seen in the clustering analysis suggests a common underlying molecular pathogenesis in all samples. Exogenous ALV was shown to enhance the B cell transformation by activation of the c-myc oncogene by adjacent integration of ALV provirus (Kung & Liu, 1997). In our experiment, we did not find a change of Myc expression in any tumor samples. Instead, TUSC2 (FUS1) is a tumor suppressor gene that was completely lost in all tumor samples. Among the differentially expressed genes, TUSC2 was the highest expressed gene in both normal bursa and splenic B cells. The TUSC2 gene resides in the 3p21.3 human chromosomal region. Chromosomal abnormalities in the 3p21.3 region are observed in lung, breast, cervical, and other cancers (Lerman & Minna, 2000; Senchenko et al., 2003; Zabarovsky et al., 2002). The impaired expression of TUSC2 is a pathognomonic feature in most types of lung cancers (Ivanova et al., 2009; Prudkin et al., 2008) and an effective therapeutic target also in these tumors (Meng et al., 2013). Mice lacking one or both copies of the Tusc2 gene develop a chronic inflammatory autoimmune disorder and produce tumors at the sites of chronic inflammation (Ivanova et al., 2007). TUSC2 is considered as a molecular link between inflammatory response and mitochondrial homeostasis (Hood et al., 2013; Uzhachenko et al., 2012). TUSC2 was shown to influence and complement the PI3K/AKT and p53 pathways (Figure 6.7) (Ji & Roth, 2008; Meng et al., 2013). Our pathway enrichment analysis showed significant dysregulation of both pathways. 187 Figure 6.7: TUSC2 Molecular pathway (Ji & Roth, 2008) We also observed increased expression of EIF4E, a the proto-oncogene associated with many cancers including colon (Zimmer et al., 2000), head and neck (Franklin et al., 1999; Nathan et al., 1997; Nathan et al., 1999), and breast cancers (Larsson et al., 2007; Soni et al., 2008). Transgenic mice overexpressing Elf4e by the ubiquitous β-actin promoter show high incidence of tumors including B-cell lymphomas, angiosarcomas, lung adenocarcinomas and hepatocellular adenomas (Ruggero et al., 2004). Importantly, a cooperation between Elf4e and c-Myc in B-cell lymphomagenesis was shown. In this co-operation, Elf4e suppresses the c-Myc-induced apoptosis while c-Myc antagonized Elf4e-induced growth arrest (Ruggero et al., 2004). The Elf4e 188 expression is usually secondary to apparent phosphoinositide-3 kinase and Akt signaling (Culjkovic et al., 2008; Ruggero et al., 2004; Zimmer et al., 2000). EIF4E overexpression seen in our tumor sample could be a downstream effect of TUSC2 loss and abnormal Akt signaling. Another possible scenario is the convergence of the both genetic defects to induce the B cell transformation Future studies should confirm the transcriptional changes of TUSC2 and EIF4E in the tumor samples. We also need to confirm this association in more independent ALV-like tumors. Transgenic mice overexpressing EIF4E by the ubiquitous β-actin promoter have been published before (Ruggero et al., 2004). This mouse is good source to test the interaction between EIF4E and TUSC2. We can knock down TUSC2 in non-transformed bursa cells obtained from these mice and observe for transformation. Both TUSC2 and EIF4E are known therapeutic targets in cancer (Meng et al., 2013; Soni et al., 2008). Meng et al transfected the lung cancer cells with vectors expressing TUSC2 to increase the susceptibility of the cells to apoptotic agents. Similarly, Soni et al. knocked-down EIF4E by small interfering RNA to inhibit growth in different breast cancer cell subtypes. We should implement similar experiments on malignant lymphoblasts obtained from LL tumors to prove the necessity of these genes in the oncogenic changes of LL. 189 APPENDIX 190 Normalized expression q value Gene Malig vs Bursa Spleen vs Bursa Malig Bursa 2.47E+09 3.63E+08 0 0.006024 0.000508 0.000508 EIF4E 490456 0 0 0.009843 0.009843 1 FAM117A 235448 0 0 0.0281 0.0281 1 ENSGALG00000022685 82305.1 19629.8 7658.55 0.000859 0.000859 0.101893 RPS15 7755.37 5058.15 3563.76 0.028294 0.000508 0.137016 RPS25 5165.21 3229.19 2909.53 0.013846 0.003083 0.692084 RPL17L 5141.41 1722.61 993.123 0.015006 0.000859 0.434835 RPL7A 4936.85 3163.14 1930.87 0.042044 0.000508 0.040248 RPS4 3878.47 2553.37 2268.59 0.046946 0.010375 0.662805 ENSGALG00000027261 3439.99 600.394 1154.29 0.000859 0.035828 0.393214 ATP5A1W 2978.79 674.247 415.53 0.000859 0.000859 0.507745 GNG4 2542.9 769.284 15.0153 0.000508 0.002755 0.007117 RPS12 2093.8 1344.21 1231.73 0.026069 0.008392 0.758037 FAM13B 1786.25 292.282 48.545 0.000508 0.000508 0.000508 EEF1D 1724.06 1151.83 961.716 0.049562 0.002755 0.50061 CRIP2 1716.01 836.72 672.093 0.000508 0.000508 0.398886 BTF3 1572.08 252.772 293.264 0.000859 0.001511 0.857955 1525.8 1027.51 918.353 0.040248 0.007615 0.671937 POLN 1447.48 247.656 98.4941 0.000508 0.000508 0.018625 SLC39A14 1327.44 202.748 135.206 0.000859 0.000859 0.394509 SLC25A37 RPL35A Spleen Malig vs Spleen Table A2: Genes significantly up regulated in malignant samples over both bursa and spleen samples 191 Table A2 (Cont’d) HSPD1 874.058 551.08 215.93 0.021405 0.000508 0.000508 MYBL1 736.129 271.897 7.88936 0.002755 0.000508 0.000508 EIF2S3 692.205 441.462 270.228 0.032432 0.000508 0.038893 656.52 42.5198 233.911 0.000859 0.03681 0.005736 DMTN 649.783 97.4209 79.6317 0.000859 0.000859 0.764011 ENSGALG00000014584 614.533 122.83 128.524 0.003299 0.005347 0.958976 IL18 605.775 140.243 171.42 0.000508 0.000508 0.578448 LDHA 594.428 330.738 172.09 0.003402 0.000508 0.004024 ENSGALG00000000140 572.084 81.5445 127.872 0.000859 0.009905 0.610247 ENSGALG00000013312 569.394 51.6126 233.934 0.000859 0.03933 0.001511 SC4MOL 552.336 288.086 225.925 0.000939 0.000508 0.39884 ACTR3B 539.3 311.391 67.1201 0.004604 0.000508 0.000508 PIWIL2 509.391 9.13099 22.159 0.000859 0.000859 1 VMP1 483.036 231.505 80.8812 0.000508 0.000508 0.000508 PDIA6 477.388 275.772 153.555 0.004889 0.000508 0.004314 TPI1 418.364 280.348 217.931 0.036676 0.000508 0.317232 FKBP4 406.854 229.886 188.823 0.003707 0.000508 0.454642 WDR66 395.587 156.601 4.3107 0.000508 0.000508 0.000508 RSU1 392.048 245.256 99.3984 0.018835 0.000508 0.000508 CSDE1 367.353 233.261 234.746 0.019233 0.020418 0.980924 FAM60A 354.441 201.146 164.332 0.003707 0.000508 0.468064 TM9SF2 350.555 217.29 132.598 0.01606 0.000508 0.027709 341.85 198.31 179.967 0.006845 0.000508 0.733828 ENSGALG00000028307 SEPT6 192 Table A2 (Cont’d) ENSGALG00000020895 338.071 193.897 124.225 0.003707 0.000508 0.091393 BLNK 333.235 224.644 169.254 0.049562 0.000508 0.249674 SPINW 332.045 42.6419 101.209 0.000859 0.010258 0.177605 DHDH 319.533 145.858 35.3754 0.042556 0.000859 0.031897 ENSGALG00000019602 315.064 74.458 143.549 0.000508 0.002413 0.014947 SNORA16 312.335 171.836 142.492 0.024147 0.004024 0.639895 IPO7 308.907 199.58 94.4668 0.033302 0.000508 0.000508 LGALS1 306.282 92.8542 38.9417 0.000508 0.000508 0.05016 BID 298.275 162.666 46.1011 0.000508 0.000508 0.000508 ENSGALG00000022335 293.607 36.9159 29.3242 0.009485 0.005736 0.838977 gga-mir-762 291.055 148.323 85.3646 0.000508 0.000508 0.009893 GPI 284.194 190.053 81.8 0.039549 0.000508 0.000508 UBE2E1 283.805 159.514 116.691 0.002061 0.000508 0.217859 HSPA4L 278.009 169.702 97.8684 0.023951 0.000508 0.032086 BZW2 269.658 181.166 97.6183 0.041552 0.000508 0.006024 PNN 268.402 171.811 150.208 0.020792 0.003402 0.618302 CCNC 266.684 136.492 58.9468 0.000508 0.000508 0.000508 EIF3B 264.604 171.512 109.77 0.021596 0.000508 0.042792 GMPR 263.33 107.439 18.6393 0.000508 0.000508 0.000508 SDHD 260.804 110.21 64.9439 0.000508 0.000508 0.021 DUSP22 257.983 115.374 78.0294 0.010857 0.000508 0.326545 PRKCD 248.53 138.264 53.4495 0.004314 0.000508 0.000508 CCND3 247.497 166.089 121.128 0.042792 0.000508 0.203596 193 Table A2 (Cont’d) ODC1 239.001 114.771 63.6004 0.000508 0.000508 0.016719 PURH 229.909 150.336 111.574 0.032991 0.001337 0.258521 RBM38 229.787 151.976 7.25138 0.032086 0.000508 0.000508 FAM103A1 227.108 136.682 64.5182 0.006301 0.000508 0.002755 RAB9A 219.558 107.797 41.7997 0.000508 0.000508 0.000508 PKNOX1 214.933 29.8019 37.4982 0.000508 0.000508 0.501767 NUCB2 203.125 91.7936 63.5541 0.000508 0.000508 0.255855 C18orf25 199.863 23.698 23.4113 0.000859 0.001511 0.991141 PRKD3 198.314 98.7271 65.1583 0.001337 0.000508 0.162981 TPD52L2 195.664 99.5018 129.402 0.000508 0.035035 0.303313 CXCL13L2 191.823 109.278 18.7992 0.012919 0.000508 0.000508 NFI1 188.678 52.1044 72.4373 0.000508 0.000508 0.344425 HBS1L 188.626 117.347 78.164 0.046054 0.000508 0.180928 LRMP 187.84 49.8108 42.2991 0.000508 0.000508 0.56758 DNAJB6 182.228 101.845 96.1518 0.000939 0.000939 0.842609 CENPW 177.263 102.599 9.06266 0.007117 0.000508 0.000508 RCC2 176.647 85.701 114.035 0.000508 0.023544 0.2502 RCSD1 176.273 83.8259 81.6743 0.000508 0.000508 0.92688 SRD5A3 173.525 78.4872 49.0575 0.000508 0.000508 0.08419 STOM 172.502 55.8967 36.2095 0.000508 0.000508 0.089407 RASGRP3 171.546 73.8425 97.1626 0.000508 0.003707 0.30215 PAFAH1B2 171.149 102.004 64.456 0.033141 0.000508 0.165353 CCNG1 166.591 86.5334 42.4034 0.000508 0.000508 0.003707 194 Table A2 (Cont’d) HSPH1 165.808 58.8905 62.9261 0.000508 0.000508 0.839923 NMT2 160.332 87.4131 92.5816 0.001707 0.004604 0.850306 HDAC1 159.336 78.9645 80.4277 0.000508 0.000939 0.948897 WEE1 156.792 104.255 13.4923 0.042377 0.000508 0.000508 MND1 156.462 90.6355 70.8982 0.034519 0.002755 0.500816 ANXA5 152.907 90.9782 96.2058 0.019233 0.038529 0.859449 VGLL4 152.286 69.2409 59.312 0.000508 0.000508 0.566259 IMPAD1 148.816 80.0987 28.648 0.031558 0.000508 0.010375 HSPA4 147.318 81.99 84.4473 0.001337 0.003083 0.919397 USP10 146.244 96.6553 68.995 0.032259 0.000508 0.169499 POLR1D 145.753 67.6363 57.0256 0.003402 0.000508 0.665455 NCOA7 141.926 86.2869 61.679 0.013626 0.000508 0.172718 TEC 140.088 70.6503 9.15593 0.000508 0.000508 0.000508 SASS6 131.796 77.3956 38.1112 0.014731 0.000508 0.009636 C19ORF12 131.184 60.3329 72.1503 0.000508 0.008392 0.562318 GGA.31975 129.111 84.9572 40.5779 0.03415 0.000508 0.000939 LYAR 127.994 83.0236 30.6556 0.025695 0.000508 0.000508 R3HDM1 126.274 54.4965 38.6039 0.000508 0.000508 0.163572 METTL14 125.206 75.238 27.3061 0.009143 0.000508 0.000508 LRRC4 124.358 8.82336 33.1291 0.001511 0.007037 0.133254 SRPK1 123.492 74.8857 62.7456 0.016284 0.000508 0.540002 MED7 122.227 67.8779 41.24 0.008904 0.000508 0.093999 121.27 56.1096 59.1198 0.000508 0.000508 0.854879 MOSPD2 195 Table A2 (Cont’d) SEPT11 120.431 57.4969 11.085 0.004314 0.000508 0.000939 URI1 118.785 78.1492 52.5295 0.037726 0.000508 0.114861 PPIF 118.176 73.6934 41.4491 0.019233 0.000508 0.015839 ZNF644 118.023 53.6857 26.3529 0.029363 0.000508 0.023367 CD1A1 118.021 19.2832 61.4972 0.000508 0.004889 0.000939 FAM129A 117.796 55.668 46.4483 0.000508 0.000508 0.487016 USP15 117.323 67.2571 56.5604 0.000508 0.000508 0.49222 WAPAL 116.197 78.5842 75.9481 0.048049 0.027354 0.907135 SDR16C5 113.738 44.0725 52.0214 0.000508 0.000508 0.546921 MTR 109.581 61.3958 52.282 0.027354 0.001707 0.66036 ENSGALG00000006723 108.964 67.3431 41.6608 0.029707 0.000508 0.081169 P4HA2 108.706 42.9481 27.6161 0.000508 0.000508 0.117276 EXOC6 107.899 67.9605 61.3801 0.024758 0.004604 0.728121 TMEM248 107.301 68.3728 59.4079 0.035516 0.005742 0.638185 RNF139 106.856 57.1786 40.3825 0.003083 0.000508 0.222124 MAP2K3 106.519 48.9303 52.8649 0.001707 0.001707 0.828237 TMEM65 102.539 48.417 17.1679 0.001707 0.000508 0.003083 FAS 100.487 40.2208 15.6239 0.006024 0.000508 0.021791 STAMBPL1 98.5653 56.5554 14.698 0.010626 0.000508 0.000508 97.395 56.7945 32.6014 0.030653 0.000508 0.054967 ZEB1 92.5795 55.8862 60.1504 0.029363 0.045323 0.831729 DTNBP1 91.8084 43.069 36.0961 0.020792 0.003402 0.676834 EHBP1 90.6004 57.3104 20.4566 0.046498 0.000508 0.000508 MRPL3 196 Table A2 (Cont’d) TRIP12 87.9382 52.68 59.2868 0.004889 0.040878 0.65692 BEND7 87.3775 54.7902 44.5491 0.045615 0.004604 0.511324 MTSS1 86.1399 33.4998 49.5651 0.000508 0.049102 0.252313 IL20RA 85.2251 13.2447 13.7549 0.000508 0.000508 0.933827 HDAC11 85.0971 33.0563 25.9839 0.000508 0.000508 0.433117 BLOC1S4 85.0875 47.8756 38.9432 0.046792 0.004604 0.619534 SLC20A2 83.7792 36.4321 14.754 0.000508 0.000508 0.000508 INPP5K 83.4341 45.9041 34.41 0.004024 0.000508 0.293996 CHORDC1 82.6696 50.9036 36.955 0.008392 0.000508 0.180993 DDRGK1 82.5846 38.1112 48.2685 0.000508 0.018195 0.420128 SCN4B 82.3832 17.9699 4.2067 0.000508 0.000508 0.019045 PLS3 81.0464 39.4989 6.53773 0.000508 0.000508 0.000508 SCIN 80.5355 52.5698 24.6277 0.035705 0.000508 0.003083 LPCAT3 80.169 47.095 33.5928 0.013846 0.000508 0.264015 FABP4 78.3716 2.08767 1.12654 0.001337 0.003402 0.59548 NFYA 78.2558 50.3369 33.5311 0.024561 0.000508 0.096047 GGA.46369 76.9525 32.6288 19.9279 0.018409 0.000508 0.292991 DNAJC10 74.7917 46.3469 28.5099 0.035705 0.000508 0.081406 CA13 73.5144 15.4671 7.89494 0.015156 0.003707 0.508024 LIN7C 73.11 45.1682 34.8197 0.013626 0.000508 0.316899 SLC7A10 72.2439 4.85704 0.506395 0.000508 0.000508 0.014287 VLDLR 70.5922 19.8233 11.9741 0.000508 0.000508 0.171605 70.535 32.3002 32.5506 0.000508 0.000508 0.980794 C12ORF35 197 Table A2 (Cont’d) RIF1 70.3058 45.3766 14.7656 0.035035 0.000508 0.000508 SERHL2 68.804 22.1829 39.1919 0.000508 0.033141 0.115818 NUDCD1 68.3359 39.6472 18.4469 0.046498 0.000508 0.021596 RRN3 67.9998 35.1339 36.232 0.000939 0.001707 0.920274 PGAM5 65.5674 42.8371 39.9614 0.045615 0.021178 0.827901 HIVEP2 65.0191 33.9471 33.0092 0.000508 0.001337 0.924921 PSEN1 64.5392 16.969 21.6597 0.000508 0.000508 0.529285 ZCCHC10 62.9052 37.6818 23.9699 0.016936 0.000508 0.106616 SLC25A12 62.8992 38.8356 29.5155 0.029707 0.000508 0.364586 RECQL5 61.5121 14.4256 5.69456 0.000939 0.000508 0.068378 TPK1 61.4578 23.2101 16.919 0.010133 0.001337 0.555679 ENSGALG00000027836 61.2314 11.8349 12.4052 0.019632 0.004833 1 LYRM1 60.4049 25.6746 30.0922 0.010375 0.040248 0.758453 PIT54 59.1434 4.81754 2.2398 0.006625 0.043255 1 MAP2K2 58.011 32.9457 33.5946 0.030281 0.046645 0.960897 OSBPL3 56.6664 17.8905 13.8551 0.000508 0.000508 0.397079 KCNK17 56.5455 15.8311 20.4354 0.000508 0.000508 0.501887 GGA.31495 56.3451 19.7133 14.4842 0.000508 0.000508 0.357609 56.03 18.7015 21.4638 0.007615 0.010857 0.816414 FAM125B 55.8808 31.6062 24.6963 0.003083 0.000508 0.367547 NOC3L 55.1614 34.3804 22.0094 0.023367 0.000508 0.103494 COL6A1 54.2246 34.0331 0.340008 0.034691 0.000508 0.000508 GMPS 54.0928 32.0966 0.010133 0.000508 0.139349 CCDC34 22.1008 198 Table A2 (Cont’d) ACSL5 53.9811 33.1821 26.4646 0.023181 0.000508 0.454946 MOXD1 53.6435 14.8233 0.253233 0.000508 0.005742 0.012254 TOMM34 53.4712 24.6449 18.191 0.002755 0.000508 0.429937 MB21D1 53.3292 25.9723 12.8736 0.009387 0.000508 0.073075 KIAA1430 52.8945 27.9942 23.5556 0.005455 0.001337 0.616729 PGPEP1 52.5872 20.3683 18.9546 0.004314 0.002413 0.892303 E2F3 52.2599 24.161 6.26593 0.003083 0.000508 0.004889 FKBP7 52.1913 26.6998 5.84127 0.026647 0.000508 0.002061 TPTE 51.8425 13.1802 2.25835 0.000508 0.000508 0.000508 SLCO4A1 51.6363 23.5946 1.47743 0.000508 0.000508 0.000508 SNX10 51.4705 28.2791 31.1594 0.004604 0.020593 0.764273 ABCF2 50.9524 32.9192 24.0782 0.029178 0.000508 0.240839 ACSL3 50.9399 26.1913 11.2004 0.000508 0.000508 0.002755 ARID1B 50.6929 33.42 28.4761 0.032786 0.003083 0.549158 50.607 22.2764 1.78904 0.01756 0.000508 0.004604 GZMA 50.4736 7.8734 11.696 0.000508 0.000508 0.40661 MAMDC2 50.1389 12.7447 2.81786 0.000508 0.000508 0.001337 HSF2 49.8087 29.6242 18.9939 0.019233 0.000508 0.123308 RB1 49.5251 26.7261 22.8285 0.002755 0.000939 0.625532 ZNF598 49.3741 31.3705 32.6548 0.023737 0.04055 0.894454 ANKRD27 48.2475 28.6898 17.8925 0.012919 0.000508 0.076479 ENSGALG00000027080 47.4958 10.2974 10.7812 0.009485 0.028262 0.964009 LCA5L 47.4075 9.8274 2.95464 0.000508 0.000508 0.012919 AK1 199 Table A2 (Cont’d) RAG2 47.3762 26.5306 0.219988 0.025695 0.038204 0.065085 RFLB 46.9476 2.0684 3.19626 0.002061 0.000508 0.653075 LSS 46.0031 21.4339 24.1398 0.002755 0.029556 0.78243 NAA25 45.7087 30.3296 25.4397 0.036199 0.001337 0.517014 ENSGALG00000021862 45.6503 14.2771 6.47071 0.037877 0.005455 0.367625 LRRCC1 45.4312 25.1816 8.99862 0.005742 0.000508 0.000508 PITRM1 44.9262 22.7592 18.0313 0.001707 0.000508 0.440357 TNFRSF1B 44.0166 13.1469 27.0104 0.000508 0.04055 0.008648 FGF12 43.8994 18.8334 4.47353 0.015617 0.000508 0.007875 ZFP92 43.794 24.4881 7.88216 0.024147 0.000508 0.001337 DDX47 43.5566 24.7253 23.9979 0.010133 0.006301 0.930252 JMJD6 43.3195 20.7141 27.1286 0.000508 0.023181 0.335209 C3H2ORF43 43.2789 21.2175 19.7676 0.000508 0.000508 0.822123 DHX40 43.0061 20.6415 19.8732 0.006571 0.002755 0.927894 TADA1L 42.8508 24.6315 22.821 0.048785 0.022381 0.852897 G0S2 40.9724 1.22642 4.25318 0.002061 0.000939 0.199004 CD200 40.9574 20.6454 21.9917 0.001337 0.027037 0.869328 ANKRD60 40.7367 25.1179 23.0967 0.032596 0.014519 0.807934 TRABD 40.5668 25.6072 15.4532 0.029178 0.000508 0.044542 MTHFD1 40.2411 23.3022 10.1842 0.004889 0.000508 0.001707 PGS1 39.4513 17.8118 13.3978 0.000939 0.000508 0.475205 GLT1D1 39.3647 13.3234 7.87353 0.000508 0.000508 0.116698 BTBD11 39.3412 18.2841 7.07037 0.000508 0.000508 0.002061 200 Table A2 (Cont’d) NCAPD3 39.2565 24.5125 4.12427 0.023544 0.000508 0.000508 DENND3 38.9517 3.77331 10.8735 0.000508 0.000508 0.005162 PTPRF 38.931 18.485 1.1627 0.000508 0.000508 0.000508 FBXO38 38.848 22.2507 13.0786 0.008648 0.000508 0.046792 GFER 38.4681 20.0902 14.8312 0.012919 0.000939 0.432814 TNFSF4 38.2446 3.50097 1.72807 0.001337 0.006024 0.552373 WDR4 38.2083 22.1554 12.5673 0.024758 0.000508 0.07635 COL6A2 37.8591 24.4129 0.406533 0.034691 0.000508 0.000508 GUCY1B3 36.5347 16.6291 10.5002 0.003083 0.000508 0.228148 FAM160B1 36.1975 20.6031 18.1704 0.004889 0.000508 0.663588 FAF1 35.1842 21.1093 20.0421 0.019843 0.009893 0.877255 IFIH1 34.6964 12.5105 19.0742 0.000508 0.007357 0.129485 NEK4 33.7175 18.1395 5.38546 0.022381 0.000508 0.000508 WDHD1 33.4796 21.5096 3.32527 0.041371 0.000508 0.000508 TTPAL 33.0189 14.7241 8.14469 0.012919 0.028453 0.364262 MAPK11 32.9836 16.3064 1.62906 0.032432 0.000508 0.000508 RNF111 32.9516 19.5606 17.5657 0.032786 0.006571 0.758753 C4ORF21 32.9371 21.9377 0.87406 0.017772 0.000508 0.000508 8.46255 0.265238 0.000508 0.000508 0.000508 PVRL3 32.775 PCM1 32.7401 18.9889 18.9329 0.003707 0.004024 0.992666 ENSGALG00000006092 32.3991 13.9438 12.3499 0.000508 0.000508 0.748264 ARFGAP1 31.7584 19.4565 20.5426 0.021178 0.043084 0.865256 SYT1 31.0921 1.16967 0.238788 0.000508 0.000508 0.051824 201 Table A2 (Cont’d) PUS1 30.584 17.5488 14.9951 0.018195 0.003707 0.649421 GPHN 30.0662 9.25647 6.73085 0.000508 0.000508 0.40091 LRRK2 29.1594 11.5437 15.7136 0.000508 0.003707 0.285162 PAX3 28.1347 13.0398 0.32904 0.000508 0.000508 0.000508 FADS1 28.0537 7.39644 6.88725 0.000508 0.000508 0.884774 SH3GL3 27.9784 3.8012 2.63069 0.000508 0.000508 0.562974 SELO 27.9732 8.76824 8.94975 0.000508 0.000508 0.956393 MAD1L1 27.9508 15.3401 15.0505 0.012707 0.012482 0.960504 THBS1 27.4234 8.26698 0.58576 0.000508 0.000508 0.000508 ENSGALG00000006966 27.2309 15.2322 13.0741 0.007875 0.000939 0.620797 DNAJB9 26.9246 16.7054 12.0064 0.028632 0.000508 0.252139 TCF7L2 26.7849 8.60307 9.09137 0.000508 0.000508 0.894998 STK32C 26.3767 3.2101 0.453204 0.000508 0.000508 0.040086 MED13 25.6803 15.2209 15.6017 0.010133 0.015839 0.936589 CALB1 25.3831 3.99522 0.226361 0.000508 0.000508 0.000508 ENSGALG00000014126 25.2239 11.2387 0.440083 0.048785 0.000508 0.005455 24.762 13.2319 13.2114 0.009143 0.008392 0.996596 FNIP2 24.7337 5.55651 5.60829 0.000508 0.000508 0.983789 DSCAM 24.5632 1.38692 0.239229 0.000508 0.000508 0.007875 TUBE1 23.8774 12.0403 13.3382 0.016936 0.041552 0.812206 RNF157 23.849 12.4548 0.539129 0.043558 0.000508 0.000508 MYOM1 22.8251 1.02126 0.262186 0.000508 0.000508 0.046366 CIP1 22.6267 4.85745 0.000508 0.032432 0.244025 FAM114A1 9.41451 202 Table A2 (Cont’d) ORAI2 21.508 11.0767 9.72237 0.011088 0.003707 0.745265 CEP192 21.288 12.3918 2.61824 0.025695 0.000508 0.000508 ENSGALG00000021106 21.1327 2.37149 1.10897 0.026458 0.040406 0.580822 FAM217B 21.0359 9.85268 9.38811 0.000508 0.000508 0.889108 SNAP47 20.9185 12.3021 12.7205 0.027709 0.034867 0.925882 NLE1 20.1023 10.0715 6.48369 0.016284 0.001337 0.286941 GPSM1 19.9191 11.8862 14.4637 0.003402 0.043422 0.582877 19.697 11.2927 9.11708 0.012254 0.002413 0.519092 BAG3 19.5765 9.61523 8.35796 0.004889 0.006301 0.729573 DHX35 18.8492 9.35007 8.2523 0.010375 0.005162 0.765197 METTL4 18.735 11.2722 10.0986 0.048939 0.016508 0.776927 TVB 18.4069 7.27926 2.97432 0.004889 0.000508 0.103013 MOCOS 17.9925 0.933332 3.0796 0.000508 0.000508 0.035868 MYCN 17.5292 1.05757 1.79772 0.000508 0.000508 0.498167 ASPG 17.3655 9.10193 5.39033 0.018195 0.000508 0.139711 SLC25A33 17.1409 8.93389 3.0606 0.045917 0.000508 0.02791 gga-mir-1815 16.7175 1.26115 0.873254 0.003083 0.002413 0.732404 DPYSL4 16.5991 4.54763 0.247559 0.001337 0.001707 0.012021 NEK3 16.4941 6.51483 1.55727 0.018625 0.000508 0.017772 THNSL1 16.4157 7.04515 7.33576 0.004889 0.006024 0.933537 RGS6 16.3934 3.0426 0.126573 0.000508 0.005742 0.022785 RHOBTB1 16.1637 8.78056 1.72698 0.029556 0.000508 0.000508 ENSGALG00000006123 15.9964 6.82803 4.8052 0.000508 0.000508 0.265098 FN1 203 Table A2 (Cont’d) EPHA7 15.9705 0.870556 0.216215 0.000508 0.000508 0.073203 SULT4A1 15.9019 6.81603 0.036028 0.006301 0.611525 PRSS12 15.7126 0.706388 0.368459 0.000508 0.002413 0.583741 PGF 15.6582 0.011573 0.004604 0.033141 CLASP1 15.6231 9.1885 8.76924 0.025882 0.010857 0.89444 LATS2 15.5634 8.09807 9.69152 0.004314 0.046792 0.574291 ENSGALG00000014441 15.5108 6.24225 5.81998 0.01756 0.014947 0.911365 STAM 15.3471 8.41972 8.63155 0.006845 0.008392 0.94356 ICOS 15.1298 5.19889 3.15919 0.000508 0.000508 0.264839 RGS2 15.0883 3.47913 2.14216 0.000508 0.000939 0.454777 RTTN 14.9542 8.57434 5.56366 0.008648 0.000508 0.141326 SYBU 14.6854 3.02304 1.381 0.000508 0.000508 0.13819 FIG4 13.5863 6.75678 7.9271 0.009893 0.041024 0.674637 COL6A3 13.5388 2.04746 0.210838 0.000508 0.000508 0.000508 PARM1 13.1774 1.89581 1.66977 0.009387 0.028816 0.914599 SLC35F2 13.1249 6.45205 1.14313 0.046366 0.000508 0.006845 ERI2 12.941 7.84044 1.76924 0.043084 0.000508 0.000508 AEN 12.9359 4.91742 4.76232 0.01315 0.009893 0.960825 ZNF407 12.9052 6.47408 7.63422 0.002061 0.021 0.610378 ASIC2 12.6336 0.186888 3.14368 0.002413 0.003083 0.011573 6.61767 0.803416 0.025324 0.000508 0.000508 6.25508 6.39541 0.008648 0.012482 0.953462 3.19749 0.216423 0.000508 0.000508 0.000508 DIAPH3 KIAA1009 PLCH2 11.932 11.68 11.3226 8.53686 4.49558 0.409219 204 Table A2 (Cont’d) ALPL 11.3097 2.07703 0.229626 0.003707 0.025512 0.093999 BBS9 11.1582 5.31855 0.349019 0.017363 0.000508 0.001707 BDKRB1 11.1482 1.13409 0.085925 0.000939 0.036028 0.072279 SLMO1 11.0455 5.55538 4.23482 0.037726 0.012254 0.594906 DOC2B 10.7208 2.40364 0.237793 0.007117 0.000508 0.018625 SCNN1B 10.7128 1.79812 0.123963 0.000508 0.001337 0.005742 FAM171A1 10.2727 5.25634 0.515851 0.049562 0.000508 0.000508 FAM184B 10.202 2.98471 0.811926 0.001337 0.000508 0.017363 0.29021 0.035035 0.001707 1 CHRDL2 10.0199 0.110684 AFAP1 9.8054 4.43864 0.276595 0.001707 0.000508 0.000508 OLFM3 9.73988 3.42697 0.377979 0.005162 0.000508 0.003083 TPCN3 9.40088 3.21536 2.99359 0.010375 0.008139 0.91364 ANXA10 9.27805 0.772904 0.43548 0.009143 0.006571 0.656324 ANKRD50 9.2712 3.15949 0.512016 0.001707 0.000508 0.000508 RGS9BP 9.1278 4.34957 1.58088 0.009143 0.000508 0.018409 MMS22L 8.14548 4.89063 2.08418 0.032596 0.000508 0.009893 OSBPL6 7.84098 2.15542 0.124638 0.000508 0.000508 0.002755 CRYM 7.57204 2.50579 3.35413 0.008139 0.042516 0.624648 PTCHD1 7.39944 1.74242 0.429777 0.000508 0.000508 0.029707 GPR37 7.30112 3.28242 0.027359 0.003707 0.015384 0.029707 C10orf71 7.04963 1.12007 0.658535 0.000508 0.000508 0.368281 CNKSR2 6.83599 0.616383 2.98556 0.000508 0.045323 0.023367 TTC8 6.80802 3.78663 0.000939 0.034519 0.261578 2.47346 205 Table A2 (Cont’d) MAGI2 6.70258 0.465019 0.151336 0.000508 0.000508 0.141028 LAG3 6.39963 2.01941 0.006845 0.042921 0.423992 SDR42E2 6.22889 0.913805 0.726087 0.000508 0.000508 0.778251 C7 6.11809 2.60805 0.15911 0.020418 0.000508 0.000508 FHL5 6.11332 1.37099 0.033903 0.001707 0.03838 0.062552 PLCH1 6.01695 0.293417 1.57899 0.000508 0.000508 0.002061 ITGA8 5.80644 2.53176 0.205959 0.035868 0.000508 0.000508 RFWD3 5.57267 2.90438 1.54593 0.019233 0.000508 0.117474 MYO3A 5.37341 0.158732 0.030312 0.000508 0.013846 1 5.1624 0.067117 0.774846 0.000508 0.000508 0.011088 0.004024 0.000508 0.019233 0.6655 0.001337 0.001707 0.843259 ENSGALG00000005470 1.0365 ENSGALG00000002318 4.78461 1.7871 0.403331 NXPH2 4.69216 0.556137 TMEM136 4.56306 1.41621 0.008648 0.017363 0.836256 HTR4 4.21403 0.589149 0.814369 0.006571 0.009143 0.74288 SLC1A6 3.77089 1.27315 0.919667 0.041194 0.046199 0.716556 RUNX2 3.72993 1.42735 1.71663 0.009636 0.036839 0.732349 KIAA1107 3.62648 1.07242 0.332517 0.000939 0.000508 0.033623 GLRB 3.61473 0.140573 0.022559 0.000508 0.002755 1 ENSGALG00000019276 3.43791 1.00268 0.224975 0.007117 0.001707 0.065488 ATRNL1 3.25328 1.23281 0.271235 0.01315 0.000508 0.018625 RALY 3.07879 0.545744 0.463026 0.000939 0.000508 0.835098 ANGPTL4 2.94338 0.400201 0.085569 0.003707 0.019638 1 ENO2 2.91482 0.392882 0.734461 0.005162 0.014287 0.451768 1.21256 206 Table A2 (Cont’d) TLE1 2.8866 1.03826 0.928909 0.010133 0.004024 0.856261 COL28A1 2.67343 0.239752 0.018897 0.000508 0.003707 1 LRRC2 2.48313 0.123282 0.09928 0.000939 0.001337 1 CORIN 2.32654 0.089815 0.126155 0.000508 0.000508 1 CDKL1 1.93031 0.830798 0.713016 0.041024 0.027354 0.81552 SCML4 1.84969 0.089224 0.039091 0.002413 0.002061 1 SYCP2 1.84611 0.64382 0.366739 0.015617 0.000939 0.374654 BICD1 1.70598 0.23625 0.109174 0.000508 0.000508 1 PLXNA2 1.53473 0.471503 0.381628 0.006845 0.001337 0.741867 NPAS3 1.46558 0.28029 0.123925 0.004314 0.000939 1 CD109 1.44851 0.413856 0.051065 0.020225 0.001707 0.033141 THBS2 1.36734 0.451123 0.023163 0.037211 0.045761 0.086943 ATP7B 1.29929 0.059637 0.218104 0.006845 0.005162 1 RBP2 1.27597 0 0 0.000508 0.000508 1 NPFFR2 1.17549 0 0 0.000508 0.000508 1 CMYA5 1.13088 0.140566 0.205191 0.000508 0.002061 1 SYPL1 1.09834 0.248076 0.153447 0.010626 0.017989 1 0 0.020593 0.020593 1 GGA.46638 0.768885 0 GPR123 0.662402 0.16424 0.037028 0.031014 0.024758 1 GPR179 0.574987 0.060694 0.033828 0.006571 0.010133 1 207 Normalized expression Gene Malig Bursa Spleen q value Malig vs Bursa Malig vs Spleen Spleen vs Bursa TUSC2 0 99564600 28623200 0.000508 0.009143 0.366455 TMEM194B 0 343911 359023 0.005742 0.013846 0.976893 ENSGALG00000027490 0 11363 4.38036 0.000508 0.000508 0.000508 GGA.13329 0 59.6019 14.739 0.000508 0.000939 0.140236 GRP 0 1.37458 37.4668 0.000508 0.000508 0.007117 PTPRO 0.031932 0.636413 2.64164 0.037019 0.033808 0.007875 CPAMD8 0.067003 0.883522 0.898642 0.004604 0.004604 0.979558 TSPAN7 0.070848 3.17931 4.88668 0.03838 ENSGALG00000000584 0.091068 1.59459 0.829181 0.000508 0.000508 0.178746 CASZ1 0.091848 1.06893 0.708934 0.006845 0.017158 0.500045 PCSK5 0.096798 0.895686 0.387093 0.004314 0.045917 0.145658 0.10418 2.00911 2.55017 0.015156 0.012021 0.751984 NCAM1 0.108176 1.04421 2.52137 0.008904 0.000939 PAMR1 0.110381 0.878291 0.905252 0.032432 0.030281 0.971404 LAPTM4B 0.110602 5.93472 3.35264 0.001707 0.002061 0.185641 DTNA 0.113291 1.79288 0.771049 0.003083 0.022785 0.217859 GPR64 0.121855 2.28681 33.2029 0.001337 0.000508 0.000508 MB21D2 0.133389 0.838088 3.24251 0.022182 0.001337 0.012254 ZBTB42 0.145226 3.81675 4.88122 0.034324 0.033302 0.717333 SLCO6A1 0.146579 2.65498 1.46056 0.000939 0.001707 0.182957 RASGEF1C 0.038204 0.47944 0.05016 Table A3: Genes significantly down regulated in malignant samples over both bursa and spleen samples 208 Table A3 (Cont’d) SVEP1 0.156212 1.81515 0.501278 0.000508 0.036367 0.004604 SEMA5B 0.162812 0.880199 2.64042 0.016508 0.000508 0.041371 APCDD1 0.165404 5.56992 1.03581 0.002755 0.041876 0.028632 PCDH1 0.184728 1.03515 1.64086 0.005742 0.000508 0.363706 SLC16A7 0.187369 1.00218 7.13907 0.006845 0.000508 0.000508 NT5E 0.201322 15.8623 1.99502 0.011088 0.044686 0.000508 ILDR1 0.20429 2.74872 1.84065 0.000508 0.002755 0.357978 PLCB4 0.205287 1.58669 1.65557 0.001337 0.001337 0.939069 ZNF618 0.230157 1.53125 1.97447 0.000939 0.000508 0.66241 MISP 0.243856 1.48565 5.44347 0.041024 0.002755 0.10673 K60 0.244816 4.90364 12.8634 0.011573 0.006301 0.045323 AMOTL1 0.247764 2.55799 1.26129 0.003083 0.027183 0.241526 ZNF385B 0.260418 1.03713 2.33625 0.041552 0.004604 0.163572 CDS1 0.267813 3.5723 2.94819 0.000939 0.001707 0.685291 ENSGALG00000014946 0.275816 2.24047 2.70998 0.000939 0.000508 0.800608 SLC2A9 0.277643 13.3941 3.54365 0.000508 0.000508 0.000508 RAB11FIP1 0.284042 5.70401 1.7216 0.000508 0.007875 0.022381 DOCK5 0.288369 0.902696 4.29257 0.036367 0.000508 0.001337 ENSGALG00000020561 0.300321 3.05807 4.94227 0.000508 0.000508 0.216986 0.302022 0.968187 5.41723 0.03397 0.000508 0.000508 0.30577 7.29672 8.87752 0.014062 0.012707 0.758476 NFATC2 0.306909 1.13348 9.42593 0.040248 0.000508 0.000508 EPHB2 0.314025 2.64524 3.18688 0.002061 0.000939 0.734497 ADAM12 P2RY2 209 Table A3 (Cont’d) HUNK 0.32346 1.38466 1.51163 0.009387 0.007117 0.888702 TLR5 0.326069 1.93421 1.49421 0.009636 0.023544 0.677327 PTGS1 0.329113 2.77142 8.7778 0.019436 0.002061 0.072681 PKD1 0.33125 1.50511 2.80855 0.000939 0.000508 0.082469 PPL 0.333797 10.0143 1.4017 0.000508 0.015839 0.000508 ME3 0.336629 2.92409 2.07601 0.012021 0.02626 0.607226 KCNK5 0.353873 5.85075 6.0595 0.000508 0.000508 0.932756 SH3TC2 0.365139 3.61872 6.31777 0.002061 0.000508 0.283832 PKIB 0.365324 2.44848 2.53344 0.002755 0.002061 0.953462 ENC1 0.372791 1.8575 3.83025 0.037019 0.007615 0.214132 0.38573 5.91293 1.66328 0.000508 0.005455 0.001337 BEND5 0.391377 1.57968 2.04879 0.01315 0.005162 0.624454 CNFI-A4 0.392051 3.09856 1.57607 0.003083 0.031908 0.257037 ARHGEF17 0.407019 1.93121 1.26498 0.006024 0.045323 0.472769 ACOX2 0.413346 3.82857 2.90633 0.002755 0.006301 0.628671 DAB1 0.427216 1.43655 6.36677 0.032596 0.000508 0.000508 GATA3 0.43529 3.54972 3.10249 0.000508 0.000939 0.776628 ABI3BP 0.440246 2.00226 5.98381 0.006845 0.000508 0.012482 PCTP 0.446443 1.68886 2.06064 0.040878 0.021178 0.775497 KANK1 0.447502 1.33973 9.90361 0.009636 0.000508 0.000508 MAT1A 0.44817 2.66387 2.14173 0.006024 0.013626 0.708843 INADL 0.453643 3.67865 1.58889 0.000508 0.013386 0.035381 OSGIN1 0.456752 4.06742 4.14297 0.003402 0.003083 0.975903 SEMA3C 210 Table A3 (Cont’d) KLF4 0.457453 4.11843 4.44655 0.000508 0.000508 0.899002 GIPC2 0.466792 16.878 9.15438 0.001707 0.003402 0.201631 NIPAL1 0.470516 2.93558 5.2957 0.000939 0.000508 0.145118 SHROOM2 0.488595 9.49892 3.3803 0.000508 0.000508 0.013846 ENSGALG00000011717 0.527863 2.22384 1.913 0.008904 0.017989 0.807146 SLC6A8 0.545285 4.02394 4.25168 0.000939 0.000508 0.928389 ENSGALG00000006325 0.548728 3.3864 6.34173 0.005742 0.000939 0.214199 PTPN13 0.555312 3.00328 6.98901 0.000508 0.000508 0.008392 MAML3 0.559631 1.98999 4.44928 0.048939 0.003707 0.154647 EDN3 0.577767 3.03137 4.30061 0.002755 0.000508 ER81 0.583483 4.149 1.78656 0.000508 0.030653 0.041717 GOLPH3L 0.621384 39.7059 6.37631 0.000508 0.007117 0.001337 CHTL1A 0.625205 5.27514 3.06649 0.004024 0.027709 0.343902 OTUD7A 0.632015 4.35121 17.2045 0.003083 0.000508 0.000508 MAGI1 0.665257 2.73304 4.46245 0.007357 0.000508 0.335038 0.70358 3.98005 4.65347 0.000508 0.000508 0.709315 0.738464 3.02799 11.5467 0.017363 0.000508 0.007357 ENSGALG00000011930 0.740705 216.926 4.34613 0.000508 0.009893 0.000508 ENPP2 0.74349 2.80555 3.83038 0.043558 0.012482 0.665226 KALRN 0.745573 6.52853 10.1921 0.001337 0.000508 0.331392 IRK1 0.754172 5.58922 2.86682 0.001337 0.021791 0.184982 HIC2 0.767319 3.08165 6.69065 0.009387 0.000508 0.104579 ODZ3 0.800241 4.48102 18.6009 0.000508 0.000508 0.000508 ARHGAP6 HELIOS 211 0.42898 Table A3 (Cont’d) NFATC1 0.801019 2.40936 21.9287 0.042202 0.000508 0.000508 PLD1 0.820285 4.26886 4.49194 0.000508 0.000508 0.898116 0.8324 2.48963 2.21298 0.009893 0.020418 0.803342 TLR3 0.832801 7.75684 2.17355 0.000508 0.043902 0.000939 MAPK13 0.847589 5.66231 6.29015 0.009387 0.006571 0.871348 PLCD1 0.861296 9.24695 3.76277 0.000508 0.000939 0.004024 IL8 0.878176 6.55452 7.2487 0.000508 0.000508 0.831875 PPM1H 0.890929 12.4084 8.43123 0.003083 0.011088 0.654779 HOMER2 0.896948 3.86327 3.72386 0.038204 0.043558 0.960148 ARHGEF16 0.919259 4.193 4.40153 0.011088 0.008904 0.943239 STON2 0.923557 3.55556 8.60936 0.006301 0.000508 0.040248 SOCS2 0.928653 5.7905 11.0307 0.024561 0.004889 0.306597 0.94884 2.71065 4.45746 0.035705 0.000939 MLKL 0.953769 7.04383 3.69207 0.003083 0.023544 0.221607 EML1 0.965554 5.16482 3.64939 0.000508 0.003083 0.422559 SSBP2 0.969702 5.67496 4.77462 0.007357 0.014062 0.798599 MAPKAPK3 0.971674 12.6696 3.57895 0.000939 0.047893 0.006301 0.97844 3.04846 15.3345 0.043558 0.000508 0.000508 0.9896 4.01014 5.68699 0.000508 0.000508 0.300943 LRRC16A 0.996079 4.62987 5.59665 0.000508 0.000508 0.638122 TMEM116 1.02607 8.94275 3.22859 0.000939 0.046054 0.048181 RBM47 1.03047 9.2107 26.7843 0.000508 0.000508 0.004314 IL17REL 1.0513 27.0148 9.85616 0.000508 0.000508 0.007875 SEMA6D CABLES1 RXRA FAM135A 212 0.3128 Table A3 (Cont’d) ENSGALG00000019861 1.05626 10.9267 10.7654 0.000508 0.000508 0.973018 PLEKHA5 1.06136 3.77473 4.65813 0.001337 0.000508 0.569319 BACE2 1.06673 16.3356 9.0057 0.000508 0.000508 0.047893 BAAT 1.10777 5.504 5.16292 0.013846 0.019233 0.929253 EPHA3 1.11099 3.42788 4.95961 0.005162 0.000508 0.352751 MEIS1 1.11402 27.4519 5.38298 0.000508 0.010133 0.004889 SMAD6 1.12881 4.20661 19.2271 0.025134 0.000508 0.011803 UGT8 1.15209 31.7668 15.0632 0.000508 0.000508 0.012919 DOCK1 1.15404 4.09638 4.78731 0.000508 0.000508 0.730117 IQSEC1 1.15571 4.40534 6.71355 0.002413 0.000508 0.304986 BEAN1 1.17527 7.51839 7.33904 0.007615 0.007615 0.969601 HS1BP3 1.17598 3.32188 4.11694 0.030281 0.009387 0.673603 ARHGAP20 1.20295 3.18119 5.13516 0.020024 0.000939 0.250639 RGN 1.22335 6.0976 5.13665 0.029178 0.049102 0.791728 CSRP2 1.22801 21.5894 4.16787 0.000508 0.003707 0.000508 CAMKK1 1.24793 3.95076 5.2606 0.019045 0.004889 0.581677 FAM134B 1.26981 6.78977 5.36433 0.000508 0.000939 0.514154 IRF6 1.29576 20.0255 8.48929 0.000508 0.004314 0.024352 GNAL 1.29741 10.7448 7.62893 0.001337 0.007875 0.509265 CCBP2 1.30546 6.55851 4.69991 0.007357 0.024561 0.558356 1.3269 3.7982 6.80223 0.047893 0.001707 0.242418 ENSGALG00000002326 1.33972 6.4645 4.7818 0.000508 0.000508 0.395182 AASS 1.34746 13.0536 3.31588 0.000508 0.023367 0.000508 KIAA1598 213 Table A3 (Cont’d) EPHX2 1.35934 4.31111 8.24317 0.028294 0.000939 0.203465 ABCB1 1.3627 6.29988 19.4842 0.000508 0.000508 0.000508 MBOAT2 1.36666 4.80178 24.4073 0.012021 0.000508 0.000508 SRC 1.38908 5.73228 24.9429 0.011328 0.000508 0.002061 CDC42EP1 1.40803 7.30381 11.7118 0.020792 0.006301 0.511948 GAB1 1.42744 4.08284 5.57697 0.004024 0.000508 0.413481 SH3RF3 1.43413 9.60105 5.17429 0.000508 0.001337 0.052287 SLC41A3 1.44033 4.4492 5.19179 0.021596 0.009636 0.785341 ENSGALG00000012808 1.44155 3.29659 5.45497 0.028453 0.000939 0.160928 SRD5A2 1.44869 15.7273 7.11396 0.000508 0.006024 0.046366 TFEC 1.45138 5.14745 17.7414 0.000508 0.000508 0.000508 SATB1 1.47683 5.74643 7.94949 0.001707 0.000508 0.380986 1.4802 3.66993 12.5907 0.019233 0.000508 0.000508 CCR7 1.50697 3.83858 32.2082 0.023181 0.000508 0.000508 MICALL2 1.55982 6.74082 4.66559 0.001707 0.01606 0.452392 KLF9 1.56472 6.55789 7.65751 0.009387 0.005455 0.780665 PDE5A 1.59226 4.86121 4.56863 0.010857 0.014947 0.902645 DDAH1 1.60018 8.89949 12.4112 0.043084 0.020593 0.736545 FARP1 1.61098 5.96495 6.94414 0.000508 0.000508 0.686913 DYRK2 1.65168 7.3765 19.2492 0.001337 0.000508 0.003402 IRF2BPL 1.66892 5.8896 6.80751 0.043246 0.023181 0.846017 PCGF5 1.71267 7.82354 27.8981 0.014287 0.000508 0.004314 CPNE2 1.71884 9.4985 4.79694 0.000939 0.03738 0.151019 FAM117B 214 Table A3 (Cont’d) AFAP1L2 1.72013 6.95002 8.24132 0.000508 0.000508 KLF3 1.72173 7.14715 13.457 0.000508 0.000508 0.022588 CASS4 1.72638 6.18142 109.568 0.007615 0.000508 0.000508 TNRC18 1.74806 6.55669 6.06989 0.000508 0.000508 0.822839 ARRDC1 1.74908 5.0054 4.48179 0.014287 0.032786 0.832429 MGLL 1.75663 8.69318 27.0603 0.000508 0.000508 0.000508 LAMP5 1.76243 22.534 38.597 0.000508 0.000508 0.285486 PARD6G 1.78558 4.67688 24.6255 0.02791 0.000508 0.000508 WWTR1 1.84592 5.44693 6.83097 0.042792 0.017772 0.697029 1.847 6.50435 6.08477 0.032259 0.043422 0.924091 ARHGEF10L 1.87624 7.25919 16.5192 0.000939 0.000508 0.019233 SPIC 1.87633 24.1935 105.371 0.000508 0.000508 0.000508 LRP5 1.87986 5.38799 13.969 0.000508 0.000508 0.001337 TSPAN15 1.90083 24.7261 6.37061 0.000508 0.009893 0.001707 IGF2 1.91937 6.52536 49.4515 0.010375 0.000508 0.000508 PLK2 1.94176 5.44003 6.83741 0.004889 0.000508 0.551246 LAMA3 1.96362 9.81279 23.9708 0.000508 0.000508 0.000508 TIAM1 1.96809 5.03917 23.7046 0.030477 0.000508 0.000508 NOX1 1.98415 9.28745 9.17865 0.008648 0.009143 0.982039 KIAA1462 2.03437 4.99135 44.7799 0.007357 0.000508 0.000508 ABCA3 2.04775 4.21727 16.9987 0.036676 0.000508 0.000508 ANKRD6 2.07171 6.69452 14.0766 0.026069 0.000508 EMB 2.08889 6.70126 11.1727 0.009893 0.000508 0.098245 TMEM37 215 0.66036 0.12758 Table A3 (Cont’d) SULF2 2.08961 9.0529 46.7473 0.000508 0.000508 0.000508 TRIO 2.1088 5.19878 13.6031 0.014731 0.000508 0.006024 ENSGALG00000011008 2.1592 5.33329 5.90774 0.028816 0.016719 0.829355 HAAO 2.21228 9.76345 16.4075 0.010857 0.001337 0.363627 CRIM1 2.22432 6.98092 5.98069 0.004024 0.013626 0.730172 TMEM51 2.25335 9.94368 6.94165 0.003707 0.0281 0.498167 WDFY3 2.30125 5.42357 11.8498 0.000508 0.000508 0.000508 RNF144A 2.31835 4.87521 26.0959 0.019233 0.000508 0.000508 TSPAN9 2.32859 7.75289 7.96248 0.004314 0.003402 0.957816 TREM-B2V2 2.35699 8.34008 24.2048 0.023367 0.000508 0.023367 IGF2BP2 2.3577 8.21414 22.5546 0.002061 0.000508 0.000939 GNA11 2.37698 7.18044 17.4372 0.035516 0.000508 0.049562 SYT8 2.38611 13.1117 15.7214 0.000508 0.000508 0.664939 CHST9 2.40234 12.2818 17.7513 0.007615 0.001707 0.461785 TRAM2 2.40576 6.17187 25.4709 0.032086 0.000508 0.000508 FAM65B 2.45146 5.424 133.542 0.034691 0.000508 0.000508 CD36 2.56963 9.67458 117.539 0.004604 0.000508 0.000508 SERPINB1 2.57356 6.29345 6.67028 0.020225 0.012707 0.892507 C3H8ORF80 2.69404 20.8806 16.3182 0.000508 0.000508 0.446232 ALCAM 2.75823 127.507 20.4103 0.000508 0.000508 0.000508 TNFAIP2 2.75853 7.71761 102.071 0.011803 0.000508 0.000508 TAAR1 2.77091 26.0546 9.51468 0.000508 0.020792 0.019045 SLCO2B1 2.77915 14.05 26.7004 0.000508 0.000508 0.113746 216 Table A3 (Cont’d) C1orf198 2.78438 8.417 7.74482 0.002061 0.005742 0.840492 IL13RA2 2.79102 7.89643 59.5155 0.025695 0.000508 0.000508 ENSGALG00000026622 2.84781 6.31692 7.62765 0.020024 0.003707 0.651155 DCLK3 2.87625 8.71786 67.4573 0.004314 0.000508 0.000508 FNDC3B 2.90721 5.53627 13.5541 0.021 0.000508 0.001337 IDUA 2.94622 5.73965 10.1385 0.037877 0.000508 0.063686 TOM1 2.96555 6.65738 11.872 0.041717 0.000939 0.137252 TCN2 2.99622 7.7554 17.166 0.043558 0.000508 0.087339 GPR126 3.00488 14.0896 12.9421 0.000508 0.000508 0.793093 GSTA3 3.00787 61.8514 34.6994 0.000508 0.000508 0.086553 CMPK2 3.01488 10.1194 6.68449 0.000508 0.016284 0.209266 XDH 3.01609 14.616 10.2905 0.000508 0.000508 0.215215 ADIPOQ 3.02201 7.79763 31.0603 0.044819 0.000508 0.001707 gga-mir-1723 3.04244 8.47628 11.7999 0.005455 0.000508 0.368638 TMEM55A 3.08481 5.79507 15.8879 0.048472 0.000508 0.000939 F10 3.16978 16.7194 23.6763 0.000508 0.000508 0.297434 SMAD7A 3.17377 23.2823 7.90604 0.000508 0.046498 0.014287 TRIM25 3.18202 9.43226 22.0807 0.006571 0.000508 0.012707 ARHGAP21 3.25739 6.65078 6.65756 0.014062 0.014287 0.997074 KIAA0284 3.30032 6.37151 6.19441 0.032596 0.046498 0.945797 3.3069 15.4853 12.9723 0.000508 0.000508 0.658316 PMP22 3.35261 11.3209 21.0397 0.004604 0.000508 0.119606 MBNL2 3.48825 8.89566 8.91137 0.004314 0.004314 0.995875 JUP 217 Table A3 (Cont’d) ARRDC4 3.51399 14.5759 11.5788 0.000508 0.000508 0.453215 LCAT 3.52257 24.0232 15.2853 0.000508 0.000508 0.133525 KLF4 3.63126 11.7424 418.52 0.001707 0.000508 0.000508 SLC46A2 3.65269 13.9485 23.8908 0.023367 0.001337 0.372919 K123 3.74981 38.4522 45.4698 0.000508 0.000508 OLFML2A 3.76175 10.4987 22.559 0.007875 0.000508 0.028453 IL34 3.77065 33.7574 25.9901 0.000508 0.003083 0.646678 CRIP2 3.842 14.1945 18.8438 0.024939 0.008648 0.651942 GAS6 3.86588 9.10347 9.46873 0.017158 0.014731 0.929763 IGSF1 3.88626 8.15236 19.9483 0.042516 0.000508 0.008139 PLEKHG3 3.92112 9.48711 9.37216 0.032596 0.030101 0.981859 TNFRSF11A 3.93804 9.61676 20.5085 0.003707 0.000508 0.007875 EFNB2 3.99331 18.9242 26.2589 0.000508 0.000508 0.245044 CHST2 4.04376 19.8605 57.621 0.000508 0.000508 0.000508 CYP46A1 4.13707 8.11952 16.4152 0.049259 0.000508 0.029707 DENND4C 4.23717 7.63147 17.4612 0.036199 0.000508 0.003402 CX3CR1 4.27272 47.8291 100.933 0.000508 0.000508 0.003083 MAOA 4.36015 17.5283 136.06 0.001337 0.000508 0.000508 LHFP 4.46778 11.7735 38.4283 0.0281 0.000508 0.003707 ATP11A 4.51971 11.3786 34.7579 0.015384 0.000508 0.003707 LAMB1 4.54205 16.7902 22.5519 0.000508 0.000508 0.307152 ENSGALG00000026188 4.58184 11.7172 14.2484 0.001337 0.000508 0.565392 REV-ERBB 4.72928 10.7032 19.8868 0.028294 0.000508 0.063937 218 0.54621 Table A3 (Cont’d) TGM4 4.75004 68.7104 294.359 0.000508 0.000508 0.000508 MERTK 4.91421 11.5692 62.3949 0.002755 0.000508 0.000508 TESC 5.06102 20.2355 31.7507 0.005455 0.000939 0.400379 ST14 5.18044 19.941 15.431 0.000508 0.000508 5.4587 13.0119 40.6049 0.002413 0.000508 0.000508 AHR 5.51759 27.2075 14.5191 0.000508 0.000939 0.007615 VNN1 5.84053 77.7306 15.2045 0.000508 0.003083 0.000508 MAP4K4 5.88045 40.107 24.2251 0.000508 0.000508 0.164038 RUNX3 6.01675 13.6743 39.3157 0.002061 0.000508 0.000508 CBFA2T3 6.02418 20.1362 16.4519 0.000508 0.005455 0.624648 SOWAHC 6.07657 16.9537 170.749 0.007615 0.000508 0.000508 ENSGALG00000008755 6.08812 15.2984 22.1987 0.028453 0.003707 0.360152 ENSGALG00000010336 6.10817 76.9616 778.603 0.000508 0.000508 0.000508 ETS2 6.22195 34.9152 74.1951 0.000508 0.000508 0.002755 PLA2G4A 6.42729 14.5428 32.1337 0.002061 0.000508 0.001337 ATP2B4 6.6352 23.8871 77.9922 0.009893 0.000508 0.011803 AAED1 6.64458 20.0226 29.5981 0.040248 0.006571 0.446072 CPD 6.71025 16.3524 14.6723 0.000508 0.001707 0.732428 CLSTN1 6.7327 13.8602 26.6486 0.007615 0.000508 0.020792 ITGA6 6.77825 21.7023 37.8627 0.000508 0.000508 0.027709 HPGD 6.89161 23.3354 225.344 0.02626 0.000508 0.000508 SVIL 6.96261 12.8806 42.8868 0.0281 0.000508 0.000508 ENSGALG00000028304 6.98649 12.2243 37.6404 0.01606 0.000508 0.000508 SASH1 219 0.44242 Table A3 (Cont’d) EDNRA 7.16296 13.8902 13.9197 0.030101 0.029363 0.995352 GNG10 7.25267 66.5739 102.052 0.000508 0.000508 0.143124 BACH2 7.31154 14.2218 18.7513 0.031558 0.002413 MARK1 7.33468 16.2673 16.3064 0.003083 0.003083 0.994128 SESTD1 7.39224 16.9444 43.6512 0.004314 0.000508 0.000508 INPP5A 7.39908 13.5694 21.2495 0.036676 0.000939 0.128116 ADAM9 7.53558 14.0712 21.5508 0.035868 0.000508 0.177888 DNASE1L3 7.63404 17.6706 84.9297 0.044542 0.000508 0.000508 HES4 7.76319 29.602 61.8236 0.001337 0.000508 0.037211 ENSGALG00000008518 7.94889 59.4623 46.1109 0.000508 0.000508 0.507657 FOXI1 7.94947 18.1078 26.1027 0.007117 0.000508 0.259917 ABCC3 8.06548 19.8229 35.7362 0.046946 0.000508 0.227132 PIK3IP1 8.23974 34.201 101.858 0.000508 0.000508 0.000508 HIC1 8.28923 18.492 30.8397 0.008648 0.000508 0.128215 MCF2L 8.41862 16.1075 58.257 0.033141 0.000508 0.000508 SMPD1 8.49132 28.4941 74.3114 0.000508 0.000508 0.000508 CHDZ 8.77642 14.099 35.7552 0.047277 0.000508 0.000508 ANTXR2 9.04841 17.9383 42.7212 0.047112 0.000508 0.004024 MADH2 9.1565 21.433 18.3724 0.000939 0.007875 0.627012 ST3GAL2 9.1924 21.7255 53.9813 0.038701 0.000508 0.023951 AGPAT2 9.41875 23.4749 27.0785 0.031197 0.011573 0.771527 SULT1B1 9.4671 35.8784 37.6757 0.000508 0.000508 0.909989 9.54049 32.4843 37.8953 0.002755 0.000508 0.734885 BATF3 220 0.43691 Table A3 (Cont’d) KAT2B 9.59892 18.7419 50.6204 0.004889 0.000508 0.000508 MCCC2 9.90912 24.542 22.6469 0.001707 0.007357 0.834804 UGCG 9.96857 18.9801 40.8877 0.008392 0.000508 0.001337 SCARB2 10.3897 22.9079 39.6122 0.002755 0.000508 0.045154 TRIM3 10.4111 20.1098 31.4078 0.044542 0.000508 0.204185 SDC1 10.4561 34.821 33.3641 0.000508 0.000508 0.897981 FYB 10.4835 22.8407 129.24 0.000508 0.000508 0.000508 GNPTAB 10.8809 27.0677 52.5856 0.000508 0.000508 0.003402 ST3GAL6 11.0098 34.544 21.161 0.000508 0.035207 0.112432 TPRN 11.3196 24.8641 64.7043 0.003402 0.000508 0.000508 TNFSF11 11.9016 26.3119 34.0054 0.002755 0.000508 0.367069 S100A9 12.32 37.9278 50.3173 0.024352 0.005455 0.574868 SMPD1 12.4333 27.9909 84.0121 0.019233 0.000508 0.001707 CLCN5 12.5258 21.1835 53.5637 0.046199 0.000508 0.000508 ELF3 12.7239 58.9684 44.3292 0.000508 0.000508 0.440149 FABP7 13.3715 49.0606 440.517 0.000939 0.000508 0.000508 IRF-3 13.6281 33.7315 51.8144 0.007615 0.000508 0.254839 NFKBIE 13.8619 27.5447 69.07 0.017158 0.000508 0.000939 BDH2 13.8849 25.3178 42.8935 0.022785 0.000508 0.049562 PPDPF 14.0051 69.0516 143.646 0.000508 0.000508 0.025324 TMBIM1 14.1906 32.8311 56.4239 0.009387 0.000508 0.088765 PLOD2 14.2848 28.5609 28.1654 0.003402 0.005162 0.966534 SNTB1 14.7221 32.5636 51.9525 0.000508 0.000508 0.077064 221 Table A3 (Cont’d) PLAU 14.8344 34.9595 149.627 0.000939 0.000508 0.000508 RASGEF1A 15.1064 30.2156 56.8098 0.004889 0.000508 0.007357 TBC1D1 15.2898 29.97 28.1608 0.003707 0.010375 0.848791 B3GNT2 15.4667 47.576 52.3391 0.000508 0.000508 0.751878 TSPAN12 15.6636 32.2947 34.4499 0.007357 0.002413 0.858718 MPP1 16.2099 33.2076 53.3024 0.003083 0.000508 0.076905 SYDE1 16.6019 51.4967 535.765 0.029356 0.000859 0.000859 VDAC1 16.9436 46.7329 93.2086 0.003083 0.000508 0.027183 ZDHHC9 17.7021 32.6889 30.5284 0.009893 0.022182 0.842465 BRT-1 18.3421 63.9373 87.8242 0.000508 0.000508 0.430863 DUSP7 18.583 62.6784 79.7746 0.000508 0.000508 0.363473 ENSGALG00000026611 19.0873 109.709 93.5736 0.000859 0.000859 0.773962 TCP11L2 19.7596 32.3972 69.5213 0.03738 0.000508 0.001337 NFE2L2 19.9299 44.8117 56.0434 0.000939 0.000508 0.416203 S100A16 19.9739 72.0528 203.875 0.022182 0.000508 0.025324 SUB1 20.7937 35.206 56.0618 0.015156 0.000508 0.052287 FAM3C 20.9968 42.1351 71.7236 0.005162 0.000508 0.038204 PDLIM5 21.2027 38.6722 45.6607 0.004314 0.000508 0.545845 SGK3 21.5071 38.5786 59.721 0.004604 0.000508 0.066133 PLEKHB2 22.1496 61.6623 37.139 0.000508 0.014947 0.029006 TUBB6 22.2051 43.1896 63.5819 0.008904 0.000508 0.17283 CTNS 22.6631 41.255 68.6362 0.012919 0.000508 0.04743 LTB4R 23.4651 84.2222 160.847 0.000508 0.000508 0.015156 222 Table A3 (Cont’d) SAMHD1 23.6977 61.057 42.2576 0.000508 0.004889 0.129821 TGIF1 23.7211 46.0039 52.9727 0.021596 0.001707 0.687532 ADD3 24.7541 93.2358 110.55 0.000508 0.000508 0.541107 TAGAP 25.3725 42.189 73.8918 0.017989 0.000508 0.015384 CFD 25.3762 871.565 88.8297 0.000508 0.000508 0.000508 ASS1 26.0674 97.1358 226.195 0.000508 0.000508 0.000508 LTF 27.4783 1381.72 175.752 0.000508 0.000508 0.000508 CTNNB1 27.6302 100.267 62.8923 0.000508 0.000508 0.036839 PLSCR4 28.525 92.5301 120.117 0.000508 0.000508 0.393532 DRAM1 29.3442 53.5489 75.6942 0.009143 0.000508 0.184697 EPSTI1 31.2086 55.3298 70.2693 0.030653 0.001337 0.442132 ENSGALG00000011190 31.3789 164.558 1878.44 0.000939 0.000508 0.000508 HEXB 31.9978 68.2679 213.893 0.001707 0.000508 0.000508 LITAF 32.5128 69.7379 331.117 0.017772 0.000508 0.000508 C4BPA 32.5148 116.301 284.629 0.028816 0.000508 LYZ 33.8154 5986.69 452.803 0.000508 0.000508 0.000508 CD82 34.6493 76.1211 163.19 0.000508 0.000508 0.000508 gga-mir-147 36.0413 93.5739 66.9866 0.000508 0.007615 0.183878 GNE 36.1216 58.6151 73.1789 0.014062 0.000508 VCAM1 37.2466 58.9571 289.64 0.039913 0.000508 0.000508 PIK3CD 37.3971 75.4274 81.9943 0.009143 0.001707 GPR137B 38.7829 66.8007 177.188 0.043246 0.000508 0.000508 FAM49A 41.6386 90.4122 364.548 0.000508 0.000508 0.000508 223 0.06968 0.3883 0.80464 Table A3 (Cont’d) S100A11 42.144 178.771 109.105 0.000508 0.011803 0.161481 TAX1BP3 50.3501 88.4422 149.038 0.008392 0.000508 0.026069 APP 50.3658 243.817 161.855 0.000508 0.000508 0.075505 SMAP2 55.0645 90.871 313.284 0.037538 0.000508 0.000508 ISG12(2) 58.855 156.207 472.858 0.006301 0.000508 0.000508 HADH 61.5091 107.228 125.483 0.012919 0.001707 0.584976 SLC40A1 69.1872 291.578 1925.16 0.000508 0.000508 0.000508 gga-mir-3526 73.1726 145.732 119.473 0.000508 0.016284 SDC4 77.3579 167.166 224.543 0.006301 0.000508 0.361273 TIMD4 77.5062 562.83 393.011 0.000508 0.000508 0.162875 ITM2B 77.9683 187.133 273.665 0.000508 0.000508 0.142615 ATP1B1 78.6643 150.195 138.533 0.005455 0.016936 0.797835 NADK 81.5953 155.52 141.061 0.001337 0.003402 0.720354 CREG1 82.046 135.421 687.759 0.011088 0.000508 0.000508 ACVRL1 99.2499 244.363 2377.99 0.015801 0.000859 0.000859 ANXA11 99.3072 168.218 247.286 0.014519 0.000508 0.144181 ANKRD13D 115.107 362.664 518.865 0.002181 0.000859 0.480855 TSPAN3 127.301 215.206 257.599 0.011088 0.000939 0.538153 MYH9 164.506 244.164 488.105 0.043558 0.000508 0.007357 179.44 574.432 645.614 0.000508 0.000508 0.673321 184.523 345.639 581.711 0.000939 0.000508 192.66 710.049 650.191 0.000859 0.000859 0.873133 220.355 395.444 366.86 0.001707 0.009387 0.781003 ENSGALG00000026970 PNRC1 IL-1BETA SH3BGRL3 224 0.43925 0.01756 Table A3 (Cont’d) AKR1B10 287.807 476.292 689.201 0.046199 0.000508 0.234754 C3D 294.952 1090.11 1118.3 0.000859 0.000859 0.954525 IGJ 305.666 646.79 688.118 0.000508 0.001337 0.858204 LAPTM4A 327.313 533.039 836.049 0.008904 0.000508 0.044394 382.61 976.528 1588.47 0.001511 0.000859 0.203152 ENSGALG00000015398 530.181 1095.66 1125.56 0.008139 0.006301 0.927452 SP1 777.066 1503.76 2167.92 0.028769 0.000859 0.350366 PIK3R5 225 REFERENCES 226 REFERENCES FASTX Toolkit, http://hannonlab.cshl.edu/fastx_toolkit/index.html: Hannon Lab. Baba, T. W. & Humphries, E. H. (1985). Formation of a transformed follicle is necessary but not sufficient for development of an avian leukosis virus-induced lymphoma. Proc Natl Acad Sci U S A 82, 213-216. Bacon, L. D., Witter, R. L. & Fadly, A. M. (1989). Augmentation of retrovirus-induced lymphoid leukosis by Marek's disease herpesviruses in White Leghorn chickens. J Virol 63, 504-512. Bolger, A. M., Lohse, M. & Usadel, B. (2014). Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114-2120. Carroll, M. & Borden, K. L. (2013). The oncogene eIF4E: using biochemical insights to target cancer. J Interferon Cytokine Res 33, 227-238. Crittenden, L. B., Witter, R. L., Okazaki, W. & Neiman, P. E. (1979). Lymphoid neoplasms in chicken flocks free of infection with exogenous avian tumor viruses. J Natl Cancer Inst 63, 191-200. Crittenden, L. B., Hayward, W. S., Hanafusa, H. & Fadly, A. M. (1980). Induction of neoplasms by subgroup E recombinants of exogenous and endogenous avian retroviruses (Rous-associated virus type 60). J Virol 33, 915-919. Crittenden, L. B. & Astrin, S. M. (1981). Genes, viruses, and avian leukosis BioScience 31, 305-310. Crittenden, L. B., Fadly, A. M. & Smith, E. J. (1982). Effect of endogenous leukosis virus genes on response to infection with avian leukosis and reticuloendotheliosis viruses. Avian Dis 26, 279-294. Crittenden, L. B., Astrin, S. M. & Smith, E. J. (1983). Independent segregation of ev 10 and ev 11, genetic loci for spontaneous production of endogenous avian retroviruses. Virology 129, 514-516. Culjkovic-Kraljacic, B., Baguet, A., Volpon, L., Amri, A. & Borden, K. L. (2012). The oncogene eIF4E reprograms the nuclear pore complex to promote mRNA export and oncogenic transformation. Cell Rep 2, 207-215. Culjkovic, B., Tan, K., Orolicki, S., Amri, A., Meloche, S. & Borden, K. L. (2008). The eIF4E RNA regulon promotes the Akt signaling pathway. J Cell Biol 181, 51-63. 227 Fadly, A. M. & Witter, R. L. (1993). Effects of age at infection with serotype 2 Marek's disease virus on enhancement of avian leukosis virus-induced lymphomas. Avian Pathol 22, 565-576. Fadly, A. M. & Nair, V. (2008).Diseases of Poultry, pp. 514-568.: Iowa State University Press, Ames, Iowa. Franklin, S., Pho, T., Abreo, F. W., Nassar, R., De Benedetti, A., Stucker, F. J. & Nathan, C. O. (1999). Detection of the proto-oncogene eIF4E in larynx and hypopharynx cancers. Arch Otolaryngol Head Neck Surg 125, 177-182. Fynan, E., Block, T. M., DuHadaway, J., Olson, W. & Ewert, D. L. (1992). Persistence of Marek's disease virus in a subpopulation of B cells that is transformed by avian leukosis virus, but not in normal bursal B cells. J Virol 66, 5860-5866. Hood, M. I., Uzhachenko, R., Boyd, K., Skaar, E. P. & Ivanova, A. V. (2013). Loss of mitochondrial protein Fus1 augments host resistance to Acinetobacter baumannii infection. Infect Immun 81, 4461-4469. Ivanova, A. V., Ivanov, S. V., Pascal, V., Lumsden, J. M., Ward, J. M., Morris, N., Tessarolo, L., Anderson, S. K. & Lerman, M. I. (2007). Autoimmunity, spontaneous tumourigenesis, and IL-15 insufficiency in mice with a targeted disruption of the tumour suppressor gene Fus1. J Pathol 211, 591-601. Ivanova, A. V., Ivanov, S. V., Prudkin, L., Nonaka, D., Liu, Z., Tsao, A., Wistuba, I., Roth, J. & Pass, H. I. (2009). Mechanisms of FUS1/TUSC2 deficiency in mesothelioma and its tumorigenic transcriptional effects. Mol Cancer 8, 91. Ji, L. & Roth, J. A. (2008). Tumor suppressor FUS1 signaling pathway. J Thorac Oncol 3, 327-330. Kim, D., Pertea, G., Trapnell, C., Pimentel, H., Kelley, R. & Salzberg, S. L. (2013). TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol 14, R36. Kondo, M., Ji, L., Kamibayashi, C. & other authors (2001). Overexpression of candidate tumor suppressor gene FUS1 isolated from the 3p21.3 homozygous deletion region leads to G1 arrest and growth inhibition of lung cancer cells. Oncogene 20, 62586262. Kung, H. J. & Liu, J. L. (1997). Retroviral Oncogenesis. Philadelphia: Lippincott-Raven Publishers. Larsson, O., Li, S., Issaenko, O. A., Avdulov, S., Peterson, M., Smith, K., Bitterman, P. B. & Polunovsky, V. A. (2007). Eukaryotic translation initiation factor 4E induced progression of primary human mammary epithelial cells along the cancer 228 pathway is associated with targeted translational deregulation of oncogenic drivers and inhibitors. Cancer Res 67, 6814-6824. Lerman, M. I. & Minna, J. D. (2000). The 630-kb lung cancer homozygous deletion region on human chromosome 3p21.3: identification and evaluation of the resident candidate tumor suppressor genes. The International Lung Cancer Chromosome 3p21.3 Tumor Suppressor Gene Consortium. Cancer Res 60, 6116-6133. Mamane, Y., Petroulakis, E., Rong, L., Yoshida, K., Ler, L. W. & Sonenberg, N. (2004). eIF4E--from translation to transformation. Oncogene 23, 3172-3179. Marsh, J. D., Bacon, L. D. & Fadly, A. M. (1995). Effect of serotype 2 and 3 Marek's disease vaccines on the development of avian leukosis virus-induced pre-neoplastic bursal follicles. Avian Dis 39, 743-751. Meng, J., Majidi, M., Fang, B., Ji, L., Bekele, B. N., Minna, J. D. & Roth, J. A. (2013). The tumor suppressor gene TUSC2 (FUS1) sensitizes NSCLC to the AKT inhibitor MK2206 in LKB1-dependent manner. PLoS One 8, e77067. Motta, J. V., Crittenden, L. B., Purchase, H. G., Stone, H. A. & Witter, R. L. (1975). Low oncogenic potential of avian endogenous RNA tumor virus infection or expression. J Natl Cancer Inst 55, 685-689. Nathan, C. O., Liu, L., Li, B. D., Abreo, F. W., Nandy, I. & De Benedetti, A. (1997). Detection of the proto-oncogene eIF4E in surgical margins may predict recurrence in head and neck cancer. Oncogene 15, 579-584. Nathan, C. O., Franklin, S., Abreo, F. W., Nassar, R., De Benedetti, A. & Glass, J. (1999). Analysis of surgical margins with the molecular marker eIF4E: a prognostic factor in patients with head and neck cancer. J Clin Oncol 17, 2909-2914. Payne, L. N. & Rennie, M. (1975). B cell antigen markers on avian lymphoid leukosis tumour cells. Vet Rec 96, 454-455. Payne, L. N., Brown, S. R., Bumstead, N., Howes, K., Frazier, J. A. & Thouless, M. E. (1991). A novel subgroup of exogenous avian leukosis virus in chickens. J Gen Virol 72 ( Pt 4), 801-807. Prudkin, L., Behrens, C., Liu, D. D. & other authors (2008). Loss and reduction of FUS1 protein expression is a frequent phenomenon in the pathogenesis of lung cancer. Clin Cancer Res 14, 41-47. Pulaski, J. T., Tieber, V. L. & Coussens, P. M. (1992). Marek's disease virus-mediated enhancement of avian leukosis virus gene expression and virus production. Virology 186, 113-121. 229 Robinson, H. L., Pearson, M. N., DeSimone, D. W., Tsichlis, P. N. & Coffin, J. M. (1980). Subgroup-E avian-leukosis-virus-associated disease in chickens. Cold Spring Harb Symp Quant Biol 44 Pt 2, 1133-1141. Rovigatti, U. G. & Astrin, S. M. (1983). Avian endogenous viral genes. Curr Top Microbiol Immunol 103, 1-21. Ruggero, D., Montanaro, L., Ma, L., Xu, W., Londei, P., Cordon-Cardo, C. & Pandolfi, P. P. (2004). The translation factor eIF-4E promotes tumor formation and cooperates with c-Myc in lymphomagenesis. Nat Med 10, 484-486. Senchenko, V., Liu, J., Braga, E. & other authors (2003). Deletion mapping using quantitative real-time PCR identifies two distinct 3p21.3 regions affected in most cervical carcinomas. Oncogene 22, 2984-2992. Smith, E. J. & Fadly, A. M. (1988). Influence of congenital transmission of endogenous virus-21 on the immune response to avian leukosis virus infection and the incidence of tumors in chickens. Poult Sci 67, 1674-1679. Soni, A., Akcakanat, A., Singh, G., Luyimbazi, D., Zheng, Y., Kim, D., GonzalezAngulo, A. & Meric-Bernstam, F. (2008). eIF4E knockdown decreases breast cancer cell growth without activating Akt signaling. Mol Cancer Ther 7, 1782-1788. Trapnell, C., Roberts, A., Goff, L. & other authors (2012). Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc 7, 562-578. Uzhachenko, R., Issaeva, N., Boyd, K., Ivanov, S. V., Carbone, D. P. & Ivanova, A. V. (2012). Tumour suppressor Fus1 provides a molecular link between inflammatory response and mitochondrial homeostasis. J Pathol 227, 456-469. Vogt, P. K. (1997).Genetics of RNA tumor viruses. In Comprehensive Virology pp. 341455. Edited by H. Fraenkel-Conrat & R. R. Wagner: New York & London: Plenum Press. Weiss, R., Teich, N., Varmus, H. & Coffin, J. (1982). RNA Tumor Viruses: New York: Cold Spring Harbor Laboratory. Witter, R. L. (1997). Avian tumor viruses: persistent and evolving pathogens. Acta Vet Hung 45, 251-266. Zabarovsky, E. R., Lerman, M. I. & Minna, J. D. (2002). Tumor suppressor genes on chromosome 3p involved in the pathogenesis of lung and other cancers. Oncogene 21, 6915-6935. Zhang, H. M., Bacon, L. D., Cheng, H. H. & Hunt, H. D. (2005). Development and validation of a PCR-RFLP assay to evaluate TVB haplotypes coding receptors for 230 subgroup B and subgroup E avian leukosis viruses in White Leghorns. Avian Pathol 34, 324-331. Zimmer, S. G., DeBenedetti, A. & Graff, J. R. (2000). Translational control of malignancy: the mRNA cap-binding protein, eIF-4E, as a central regulator of tumor formation, growth, invasion and metastasis. Anticancer Res 20, 1343-1351. 231