AN ABSTRACT OF THE DISSERTATION OF Zhixing Wang for the degree of Doctor of Philosophy in Pharmacy presented on June 5, 2012. Title: Transcriptional Regulation Of Mouse Epidermal Permeability Barrier Development And Homeostasis By Ctip2 Abstract approved________________________________________ Arup Kumar Indra Skin is the largest organ in the body that protects the organism from environmental, chemical and physical traumas of each passing day. The protective skin epidermal permeability barrier (EPB) is formed within the exterior layers of the epidermis, which are regularly sloughed off and repopulated by movement of inner cells. The epidermal permeability barrier is established during in utero development and maintained through lifetime. Impaired epidermal barrier formation is one of the major features of several dermatoses such as psoriasis and atopic dermatitis. Chicken ovalbumin upstream promoter transcription factor (COUP-TF)interacting protein 2 (Ctip2), also known as Bcl11b, is a C2H2 zinc finger protein expressed in many organs and tissues. It has been shown to regulate the development of thymocyte, tooth and corticospinal motor neurons. Ctip2 is highly expressed in mouse epidermis during skin organogenesis and in adulthood. It is crucial for epidermal homeostasis and protective barrier formation in developing mouse embryos. Germline (Ctip2- null mice) and selective ablation of Ctip2 in mouse epidermis (Ctip2ep-/- mice) leads to increased transepidermal water loss (TEWL), impaired epidermal proliferation and terminal differentiation as well as altered lipid distribution during embryogenesis. Sphingolipids account for ~50% of total skin lipids by weight and are crucial components of epidermal barrier. We have recently identified Ctip2 as a key regulator of skin lipid metabolism. Germline deletion of Ctip2 in mouse embryos leads to altered lipid composition in the developing mouse epidermis by modulating the expression levels of key enzymes involved in lipid metabolism (bio-synthesis and catabolism). We also demonstrated that Ctip2 is recruited to the promoter regions of several genes involved in the ceramide and sphingomyelin biosynthesis pathways and could directly regulate their expression. Thus, we have identified Ctip2 as a key regulator of several lipid metabolizing genes and hence epidermal sphingolipid biosynthesis during skin development. To study the role of Ctip2 in adult skin homeostasis, we have utilized Ctip2ep-/- mouse model in which Ctip2 is selectively deleted in epidermal keratinocytes. We showed that keratinocytic ablation of Ctip2 leads to atopic dermatitis (AD)-like skin inflammation, characterized by alopecia, pruritus and scaling, as well as high infiltration of T lymphocytes and immune cells. We have also observed increased expression of Th2type cytokines and chemokines in the mutant skin, as well as systemic immune responses that share similarity with human AD patients. Furthermore, we discovered that thymic stromal lymphopoietin (TSLP) expression is significantly upregulated in the mutant epidermis as early as postnatal day 1 and Ctip2 was recruited to the promoter region of the TSLP gene in mouse epidermal keratinocytes. The results suggest that upregulation of TSLP expression in the Ctip2ep-/- epidermis could be due to a derepression of gene transcription in absence of Ctip2. Thus, our data demonstrated a cell-autonomous role of Ctip2 in barrier maintenance and epidermal homeostasis in adult skin, as well as a non-cell autonomous role of keratinocytic Ctip2 in suppressing skin inflammatory responses by regulating the expression of Th2-type cytokines in adult mouse skin. Present results establish an initiating role of epidermal TSLP in AD pathogenesis via a novel repressive regulatory mechanism mediated by Ctip2 in mouse epidermal keratinocytes. Altogether, our study indicates that Ctip2 could be involved in a diverse range of biological events in skin including barrier formation, maintenance and epidermal homeostasis. Ctip2 appears to be a master regulator in skin barrier functions by directly regulating the transcription of a subset of genes involved in lipid metabolism and inflammatory responses. ©Copyright by Zhixing Wang June 5, 2012 All Rights Reserved Transcriptional Regulation Of Mouse Epidermal Permeability Barrier Development And Homeostasis By Ctip2 by Zhixing Wang A DISSERTATION submitted to Oregon State University in partial fulfillment of the requirement for the degree of Doctor of Philosophy Presented June 5, 2012 Commencement June, 2013 Doctor of Philosophy dissertation of Zhixing Wang presented on June 5, 2012. APPROVED: _______________________________________ Major Professor, representing Pharmacy _______________________________________ Dean of the College of Pharmacy _______________________________________ Dean of the Graduate School I understand that my dissertation will become part of the permanent collection of Oregon State University libraries. My signature below authorizes release of my dissertation to any reader upon request. Zhixing Wang, Author DEDICATION I would like to dedicate my thesis to my parents. CONTRIBUTION OF AUTHORS Dr. Arup Kumar Indra helped design research experiments and Zhixing Wang primarily performed the research. Zhixing Wang and Dr. Arup Kumar Indra analyzed most of the data and wrote the manuscripts. Chapter 2: Zhixing Wang performed most of the experiments and analyzed the data. Jay Kirkwood and Alan Taylor performed Mass spectrometry experiments. Drs. Jan F. Stevens, Mark Leid and Gitali Ganguli-Indra provided valuable suggestions and criticism for the manuscript. Chapter 3: Zhixing Wang performed most of the experiments. Dr. Lingjuan Zhang assisted with sample collection, and contributed to Figure 3.7, 3.8, 3.10 and 3.13. Dr. Gunjan Guha did ELISA in Figure 3.9. Dr. Chrissa Kioussi provided the transgenic mice. Drs. Mark Leid and Gitali Ganguli-Indra provided valuable suggestions during the course of the conducted research. TABLE OF CONTENTS Page Chapter 1 General Introduction .............................................................. 2 1.1 Skin homeostasis and human health ............................................ 3 1.1.1 The structure and morphogenesis of mammalian skin ........... 3 1.1.2 Skin permeability barrier formation ......................................... 4 1.1.3 Lipids in epidermal permeability barrier .................................. 5 1.1.4 Impaired barrier and skin inflammatory disorders................... 6 1.2 COUP-TF interacting proteins ....................................................... 9 1.2.1 Nuclear receptors (NRs) ......................................................... 9 1.2.2 Chicken ovalbumin upstream promoter transcription factors (COUP-TFs)................................................................................... 10 1.2.3 COUP-TF interacting proteins .............................................. 11 1.2.4 COUP-TF interacting protein 1 (Ctip1) ................................. 12 1.2.5 COUP-TF interacting protein 2 (Ctip2) ................................. 13 1.3 The role of Ctip2 in skin development and homeostasis ............ 15 1.3.1 Ctip2 expression in mouse developing skin.......................... 15 1.3.2 Ctip2 expression in adult mouse skin ................................... 16 1.3.3 Ctip2 in epidermal barrier formation ..................................... 16 1.3.4 Ctip2 in epidermal surface lipid distribution .......................... 18 1.3.5 Ctip2 and cornified envelop (CE) formation .......................... 18 1.4 CTIP2 and skin diseases ............................................................ 19 1.4.1 Expression of CTIP2 in human head and neck squamous cell carcinoma (HNSCC) ...................................................................... 19 1.4.2 Role of Ctip2 in cutaneous wound healing ........................... 21 1.5 Research objectives .................................................................... 23 1.6 References .................................................................................. 31 TABLE OF CONTENTS (Continued) Page Chapter 2 Transcription factor Ctip2 controls epidermal lipid metabolism and regulates expression of genes involved in sphingolipid biosynthesis during skin development ...................................................................... 40 2.1 Abstract ....................................................................................... 41 2.2 Introduction ................................................................................. 42 2.3 Materials and methods ................................................................ 45 2.4 Results ........................................................................................ 51 2.4.1 Altered profile of sphingolipids in the developing epidermis of Ctip2-null mice ............................................................................... 52 2.4.2 Ctip2 directly regulates expression of a subset of genes involved in lipid metabolism in the developing skin ....................... 54 2.5 Discussion ................................................................................... 58 2.6 References .................................................................................. 78 Chapter 3 Selective Ablation of Ctip2/Bcl11b in Epidermal Keratinocytes Triggers Atopic Dermatitis-like Skin Inflammatory Responses in Adult Mice ...................................................................... 84 3.1 Abstract ....................................................................................... 84 3.2 Introduction ................................................................................. 87 3.3 Materials and Methods ................................................................ 90 3.4 Results ........................................................................................ 97 3.4.1 Selective ablation of Ctip2 in epidermal keratinocytes leads to altered epidermal proliferation, differentiation and spontaneous dermatitis in adult skin ................................................................... 97 3.4.2 Preferential induction of Th2-type cytokines and chemokines in adult Ctip2ep-/- skin ................................................................... 100 TABLE OF CONTENTS (Continued) Page 3.4.3 TSLP is a direct target of Ctip2 in epidermal keratinocytes 102 3.4.4 Systemic inflammatory responses in older Ctip2ep-/- mice .. 103 3.5 Discussion ................................................................................. 105 3.6 References ................................................................................ 132 Chapter 4 General Conclusion ........................................................... 138 Bibliography .................................................................................... 146 LIST OF FIGURES Figure Page Figure 1.1 Schematic representation of epidermal permeability barrier. (Adapted from Segre et al., 2006). ................................................ 24 Figure 1.2 Amino acid sequence, diagram of amino acid sequence alignment of CTIP1 and CTIP2. (Taken from Avram et al., 2000) . 25 Figure 1.3 Expression of Ctip2 in the mouse fetal skin. (Adapted from Golonzhka et al., 2007).................................................................. 27 Figure 1.4 Ctip2 mutant mice exhibit permeability barrier defects and epidermal hypoplasia. (Adapted from Golonzhka et al., 2007) ...... 29 Figure 1.5 Ctip2 is required for the formation and integrity of cornified envelope in mouse epidermis. ....................................................... 30 Figure 2.1 Liquid chromatography tandem mass-spectrometry (LC/MS/MS) analyses of mouse epidermal saturated ceramides during skin development. ............................................................... 63 Figure 2.2 Liquid chromatography tandem mass-spectrometry (LC/MS/MS) analyses of mouse epidermal unsaturated ceramides during skin development. ............................................................... 64 Figure 2.3 LC/MS/MS analyses of mouse epidermal saturated sphingomyelins during skin development. ..................................... 65 Figure 2.4 LC/MS/MS analyses of mouse epidermal unsaturated sphingomyelins during skin development. ..................................... 66 Figure 2.5 LC/MS/MS analyses of mouse epidermal sphingosines and sphinganines during skin development.......................................... 67 Figure 2.6 UPLC/MS/MS analyses of mouse epidermal cholesterol and fatty acids during skin development............................................... 68 LIST OF FIGURES (Continued) Figure Page Figure 2.7 Ctip2 regulates the expression of genes encoding lipid metabolizing enzymes during skin development. .......................... 69 Figure 2.8 Relative expression of genes encoding lipid-metabolizing enzymes during skin development. ............................................... 70 Figure 2.9 Immunoblot analyses of lipid metabolizing enzymes in the developing murine epidermis. ........................................................ 71 Figure 2.10 Ctip2 interacts directly with the promoter region of lipid metabolizing genes. ....................................................................... 72 Figure 2.11 ChIP analyses on murine keratinocytes for lipid metabolizing genes. ....................................................................... 74 Figure 2.12 Schematic representation of Ctip2 mediated differential regulation of genes encoding different enzymes of the ceramide synthesis pathways........................................................................ 75 Figure 3.1 Ctip2ep-/- mice develop a chronic skin lesions and epidermal hyperplasia. ................................................................................. 114 Figure 3.2 Ctip2ep-/- mice develop enhanced epidermal proliferation. 116 Figure 3.3 Ablation of Ctip2 leads to increased epidermal terminal differentiation. .............................................................................. 117 Figure 3.4 Enhanced eosinophil and mast cell infiltrates in dorsal skin of Ctip2ep-/- adult mice. ..................................................................... 118 Figure 3.5 Characterization of inflammatory cell infiltrates in dorsal skin of WT and in Ctip2ep-/- adult mice. ................................................ 119 LIST OF FIGURES (Continued) Figure Page Figure 3.6 Increased inflammatory cell infiltrate in Ctip2ep-/- adult mice. ..................................................................................................... 121 Figure 3.7 Th2-dependent cytokine and chemokine expression levels in WT and Ctip2ep-/- skin. ................................................................. 123 Figure 3.8 Relative expression levels of cytokines and chemokines in WT and Ctip2ep-/- skin. ................................................................. 125 Figure 3.9 Systemic immunological abnormalities in Ctip2ep-/- adult mice. ............................................................................................ 126 Figure 3.10 Ctip2ep-/- adult mice exhibited systemic immune responses. ..................................................................................................... 127 Figure 3.11 Immunological abnormalities of spleen and lymph node in Ctip2ep-/- adult mice. ..................................................................... 128 Figure 3.12 Relative expression levels of RXRa and genes involved in Notch signaling pathway. ............................................................. 129 Figure 3.13 Relative expression levels of FLG in 1w and 2w mice epidermis and dermis. ................................................................. 130 LIST OF TABLES Table Page Table 2.1 Primer sequences used in q-PCR assays. ........................... 76 Table 2.2 Primer sequences used in ChIP assays............................... 77 Table 3.1 List of primers used for RT-qPCR and ChIP assays .......... 131 Transcriptional Regulation Of Mouse Epidermal Permeability Barrier Development And Homeostasis By Ctip2 2 Chapter 1 General Introduction 3 1.1 Skin homeostasis and human health 1.1.1 The structure and morphogenesis of mammalian skin Skin is the largest organ in the body that protects the organism from environmental, chemical and physical traumas of each passing day (Lippens et al., 2009). Skin is composed of epidermis and dermis, which are separated by a basement membrane of extracellular matrix (Fuchs and Raghavan, 2002). The epidermis is a stratified epithelium that forms the uppermost compartment of the skin and is composed of four layers: basal, spinous, granular and stratum corneum (Fuchs and Raghavan, 2002). Keratinocytes are the most abundant cell type in the epidermis, that move from proliferative cells in the basal layer through the granular layer to form cornified envelope, finally to stratum corneum as flattened, dead cells (Candi et al., 2005) (Figure 1.1). The mouse epidermis develops from a single‐layered multipotent embryonic ectoderm, which is highly proliferative (Mack et al., 2005). Skin proliferation markers keratin 5 (K5) and keratin 14 (K14) expressions were observed as early as embryonic day 9.5 (E9.5), facilitating the stratification events that lead to the formation of the periderm (E9‐E12), which overlies the ectoderm (Byrne et al., 2003; Mack et al., 2005). After a series of proliferation and terminal differentiation events, the epidermis is fully formed by E18.5, consisting 4 of three live morphologically distinct living layers (basal, spinous, and granular) and a dead (cornified) layer (Byrne et al., 2003; Mack et al., 2005) (Figure 1.1). 1.1.2 Skin permeability barrier formation Barrier function is mainly conferred by the outermost layer of the epidermis, the stratum corneum (Hardman et al., 1998). The process of terminal differentiation starts when basal cells withdraw from the cell cycle and detach from the basement membrane. In the spinous layer, the cells are reinforced by a cytoskeletal framework of keratin filaments. Lamellar bodies in the granular layer are the reservoir of lipids. Keratins bundled with macrofibrils though filaggrin, and cornified envelopes (CEs) are assembled by precursor proteins underneath plasma membranes such as transglutaminase (TGM). As the cell membrane disintegrates, transglutaminase crosslink the CE proteins irreversibly, creating tough insoluble sac surrounding the keratin fibers. Finally, lipids are extruded onto the CE scaffold and the epidermal permeability barrier is formed (Segre, 2006). Once built, this barrier has a “brick and mortar” structure, with keratin macrofibrils and CEs as bricks and the lipids as the mortar to seal together the CEs (Elias, 1983; Nemes and Steinert, 1999) (Figure 1.1). 5 1.1.3 Lipids in epidermal permeability barrier The exterior layer of skin, the stratum corneum (SC), plays a critical role in skin permeability barrier formation and maintenance. Mammalian SC contains a large number of lipids, such as ceramides, sphingomyelins, cholesterols and fatty acids (Elias and Menon, 1991; Ishikawa et al., 2010). Ceramides (CERs) are the most abundant lipid types in the SC (50% by weight) and can be divided into at least 11 species (Oda et al., 2008; Ishikawa et al., 2010). It has been shown that reduced content of CERs leads to epidermal water loss as well as skin epidermal dysfunctions, and causes diseases such as atopic dermatitis (AD) (Imokawa, 2009; Ishikawa et al., 2010). Spingomyelins are another large group of epidermal sphingolipids, which have been reported as precursors of ceramides (Schmuth et al., 2000; Uchida et al., 2000; Holleran et al., 2006). Cholesterols and fatty acids are also major skin lipids, while cholesterol 3-sulfate (CSO4) is a ubiquitous metabolite of cholesterol, which is involved in cornified envelope formation by interacting with transglutaminase1 (Nemes et al., 2000). A large variety of skin fatty acids are synthesized in epidermis, and disruption of the skin permeability barrier leads to a rapid and marked increase in fatty acid synthesis by upregulating the activity and mRNA levels of several key enzymes involved in fatty acid biosynthesis (Feingold, 2007, 2009). 6 Furthermore, following acute barrier disruption, the recovery of permeability barrier function is delayed, correlating with reduced fatty acid synthesis (Feingold, 2007). Moreover, the elongation of fatty acids is also critical as mice deficient in elongation of very-long-chain fatty acid-like 4 (Elovl4) have a severely disrupted permeability barrier and die perinatally (Cameron et al., 2007; Li et al., 2007; Vasireddy et al., 2007). 1.1.4 Impaired barrier and skin inflammatory disorders The epidermal permeability barrier is established during in utero development and maintained throughout the lifetime. Impaired epidermal barrier is one of the major features of several dermatoses such as psoriasis and atopic dermatitis, which are two of the most common inflammatory skin disorders (Ghadially et al., 1996; Segre, 2006). Psoriasis is a chronic inflammatory skin disease that shows distinct erythrosquamous silvery scaly skin either in the areas of inflection or the entire body (Segre, 2006). Psoriasis has a 2%-3% of prevalence in western populations, affecting mostly adults. Psoriatic skin is characterized by inflammation, hyperproliferation, and abnormal 7 differentiation. Psoriasis was initially identified as a Th1-dependent disease due to the observation of T helper type 1 (Th1) cell secreted cytokines such as interleukin-1 (IL-1), tumor necrosis factor alpha (TNFα) and interferon gamma (INFG) (Gudjonsson et al., 2010). However, studies performed in psoriatic lesions have also identified cytokines such as IL-17 and IL-22 that are produced by Th17 cells (Roberson and Bowcock, 2010). The causes of psoriasis have not yet been fully understood; however, genes involved in skin barrier formation such as loricrin (LOR), involucrin (IVL), the small proline rich proteins (SPRRs) and late cornified envelope proteins (LCEs) were altered in the skin of psoriasis patients, indicating a correlation between psoriasis and skin barrier defects (Roberson and Bowcock, 2010).10%30% of psoriasis patients develop psoriatic arthritis, which causes destruction of joints if not treated aggressively (Nograles et al., 2009). Psoriasis also increases the risk of heart diseases and stroke (Gelfand et al., 2006, 2009). Atopic dermatitis (AD) is a chronic, genetically predisposed skin inflammatory disease that starts at early childhood with a prevalence of 10%-20% in children and 1%-3% in adults (Elias and Menon, 1991; Ishikawa et al., 2010). Clinical features of AD include skin xerosis, 8 pruritus and eczematoid skin lesion (Leung and Bieber, 2003). AD is characterized by both skin barrier deficiencies and immunological responses (Scharschmidt and Segre, 2008). In AD, a defective skin barrier is thought to permit the penetration of allergens and induces the interactions of the allergens with immune cells, promoting the subsequent release of pro-inflammatory cytokines and chemokines and elevation of IgE level (Cork et al., 2009). The molecular pathways involved in AD pathogenesis remain unclear. Distinct from psoriasis, there have been many reports on the connections between atopic dermatitis and Th2 inflammatory pathways. Pro-Th2 cytokines such as IL-4, IL-13, IL-5, and IL10 are elevated in AD patients (B. Brandt and Sivaprasad, 2011). One possible candidate that initiates inflammatory responses in AD is thymic stromal lymphopoietin (TSLP), which is a member of hematopoietic cytokine family and highly expressed in skinderived epithelial cells and also in fibroblasts and dendritic cells (Comeau and Ziegler, 2010; Ziegler, 2010; De Monte et al., 2011; Kashyap et al., 2011; Yamada et al., 2012). TSLP signals through a heterodimeric receptor formed by TSLP receptor (TSLPR) as well as IL7 receptor alpha (IL7Ra) (Comeau and Ziegler, 2010). TSLP is a key keratinocyte-derived cytokine, whose expression was greatly increased in the epidermis of a group of acute and chronic atopic dermatitis 9 patients (Soumelis et al., 2002). In mouse models with skin barrier deficiencies, mutation of genes (e.g. RXRα, VDR or Notch1) that are involved in barrier functions also leads to TSLP induction, subsequently causing AD-like phenotypes (Li, 2005, 2006; Demehri et al., 2008; Ziegler, 2010). 1.2 COUP-TF interacting proteins 1.2.1 Nuclear receptors (NRs) Nuclear receptors (NRs) are a class of proteins that interact with steroid and thyroid hormones, as well as certain other proteins, to regulate transcription in response to ligands. By regulating the expression of specific genes, NRs control diverse aspects of the organism such as embryonic development and adult homeostasis (Mangelsdorf et al., 1995; Robinson-Rechavi et al., 2003). NRs have the ability to bind to DNA and regulate the expression of adjacent genes when activated by ligands; hence, they are classified as members of the transcription factor family of proteins (Evans, 1988; Olefsky, 2001). NRs are divided into two broad classed according to their mechanism of action. Type I NRs are located in the cytosol in the absence of the ligand. Hormone binding facilitates the dissociation of 10 heat shock protein (HSP) from NR, allowing NR dimerization and translocation to the nucleus to bind to genomic DNA. The NR/DNA complex recruits other proteins to regulate transcription and eventually the cell functions. Members of Type I NRs include androgen receptor (AR), estrogen receptor (ER), glucocorticoid receptor (GR), progesterone receptor, as well as orphan receptors (Linja, 2004). In contrast to Type I, Type II NRs are located in the nucleus regardless of the presence of ligands. Without ligands, Type II NRs bind to the corepressor proteins; however, ligand binding leads to dissociation of corepressor and recruitment of the co-activator proteins to regulate gene expression. Retinoic acid receptors (RAR α, β and γ), retinoid X receptors (RXR α, β and γ) and thyroid hormone receptor (TR) are members of the Type II NRs (Mangelsdorf et al., 1995; Klinge et al., 1997; 1999). 1.2.2 Chicken ovalbumin upstream promoter transcription factors (COUP-TFs) Chicken ovalbumin upstream promoter transcription factors (COUPTFs) were originally identified to bind to the upstream promoter region of the ovalbumin gene to activate its transcription (Pastorcic et al., 1986; Sagami et al., 1986; Wang et al., 1987; Pereira et al., 1995). 11 They are members of the Type I NR superfamily and classified as orphan receptors as no ligand has been identified for COUP-TFs to date (Pereira et al., 1995; Kruse et al., 2008). COUP-TFs are generally considered as transcription repressors with different mechanisms of action. For example, COUP-TF has been shown to bind promiscuously to a variety of different repeats including the AGGTCA motif and hormone response elements of VDR, TR and RAR, leading to silencing of basal transcription activity (Tsai, 1997). It’s also well known that COUP-TF forms heterodimers with RXRs, which is a universal partner for other nuclear receptors such as VDR, RAR, PPAR and other orphan receptors (Tsai, 1997). This mechanism may involve the recruitment of nuclear receptor repressor (NCoR) and silencing mediator for retinoid and thyroid hormone receptor (SMRT), which bind to RAR and TR in the absence of ligands (Tsai, 1997). Other possible mechanisms involve active repression and transrepression (Tsai, 1997). 1.2.3 COUP-TF interacting proteins COUP-TF interacting proteins (CTIPs) were observed to interact directly with COUP-TF family members (COUP-TFI/NR2F1, COUPTFII/ARP1/NR2F2 and COUP-TFIII/EAR2/NR2F6) in yeast and in vitro 12 (Avram et al., 2000). CTIP1 (BCL11A, EVI9) and CTIP2 (BCL11B, RIT1b) are highly related members of the CTIP family, which share high sequence identity over the entire protein, and contain several conserved C2H2 zinc finger motifs (Avram et al., 2000) (Figure 1.2). They have been shown to repress the transcription of genes harbouring COUP-TF regions by binding to the canonical GC box (Avram et al., 2002). As mediators of transcriptional repression, their activity has been found to be insensitive to reversal by trichostatin A (TSA), an inhibitor of histone deacetylases (HDACs) ( Avram et al., 2000; Topark-Ngarm et al., 2006). 1.2.4 COUP-TF interacting protein 1 (Ctip1) Developmental studies in mice have shown that expression of Ctip1 starts as early 10.5 days post-coitum embryos (P10.5) in a diffused pattern in forelimb bud, branchial arches and CNS (Avram et al., 2000; Leid et al., 2004). Its expression became more restricted at later stages at E12.5 and E14.5 with the highest expression in the developing forebrain (Leid et al., 2004). CNS expression of Ctip1 remains strong with low diffuse expression throughout the E18.5 fetus (Leid et al., 2004). 13 In humans and mice, CTIP1 is also expressed in several hematopoietic tissues such as bone marrow, splenic B and T cells, monocytes and germinal B cells (Liu et al., 2003). Several reports indicated the important role of CTIP1 as a myeloid or B cell proto-oncogene in mice and humans as it is a common site of retroviral integration in myeloid leukemia (Nakamura et al., 2000; Satterwhite, 2001; Uda et al., 2008). CTIP1 is also required for normal lymphoid development as well as human fatal hemoglobin expression (Liu et al., 2003; Sankaran et al., 2008). 1.2.5 COUP-TF interacting protein 2 (Ctip2) Both mouse Ctip2 and human CTIP2 gene contains 4 exons with the majority of its open reading frame (ORF) encoded by exon 4 (Figure 1.2). The expression of Ctip2 during mouse development shares a similar pattern with Ctip1. It is expressed diffusely at E10.5 and becomes more restricted at E12.5 in the same region as Ctip1, except in the cerebellar domain as well as mandibular and nasal mesenchyme (Leid et al., 2004). A relatively high expression level of Ctip2 was observed to be expressed in thymus at E14.5 when most of the thymic cells are CD4-CD8-. The thymus becomes the most dominant organ that expresses Ctip2 at E18.5. Ctip2 expression has been detected in 14 the developing mice skin and in adult mice and human skin (Golonzhka et al., 2007; Ganguli-Indra et al., 2009a) Ctip2 expression was found to be upregulated in the skin of human AD patients and allergic contact dermatitis (ACD) patients (Ganguli-Indra et al., 2009a) Ctip2 has been reported to interact with nucleosome remodelling and deacetylation (NuRD) complex with histone deacetylase activity and repress the transcription of target genes such as p57KIP2 (ToparkNgarm et al., 2006). However, new evidence suggested the role of Ctip2 as a transcriptional activator. One-third of all Ctip2-regulated genes in thymocytes are activated by Ctip2 (Kastner et al., 2010). Ablation of Ctip2 in mouse embryonic skin leads to downregulation of several genes involved in skin lipid homeostasis (Wang et al, 2012, submitted). Hence, Ctip2 could also activate transcription under certain biological conditions. As Ctip2 null mice die perinatally, several studies have investigated the possible causes of the lethality. Ctip2 null mice show defects in thymocyte development and rearranged T cell receptors (TCR) signalling, suggesting Ctip2 is required in differentiation and survival of T lymphocytes (Wakabayashi et al., 2003). Ctip2 also plays a critical role in the development of corticospinal motor neuron (CSMN) axonal projections to the spinal cord in vivo (Arlotta et al., 2005). Thymocytes 15 isolated from Ctip2 null newborn mice exhibit increased vulnerability to DNA replication stress and damage, which leads to increased apoptosis (Kamimura et al., 2007; Karanam et al., 2010). Recent reports suggested crucial roles of Ctip2 in tooth and skin development (Golonzhka et al., 2007, 2008, 2009). 1.3 The role of Ctip2 in skin development and homeostasis 1.3.1 Ctip2 expression in mouse developing skin Ctip2 expression is detected in the ectoderm as early as E10.5 in the outermost layer of skin. Some of the positive cells also express keratin 14 (K14), which is the marker of the basal layer (Golonzhka et al., 2007). Embryonic basal layer is formed at E12.5, in which every cell is found to express Ctip2 (Golonzhka et al., 2007). Strong expression of Ctip2 is observed at E14.5, mainly in the basal layer and suprabasal layer of the epidermis. Further investigation of Ctip2 expression was performed on E16.5 and E18.5 mouse skin. Ctip2 expression remains high in the basal layer at both stages with small groups of positive cells in the suprabasal layer. Also, Ctip2 expression was found in hair follicles and hair bulbs at later developmental stages with a few positive 16 cells in the dermis (Golonzhka et al., 2007). Ctip2 expression is overlapping with cell proliferation marker Ki67 positive signals in the epidermis and hair follicles at majority of developmental stages, indicating that Ctip2 positive cells are highly proliferative during skin development (Golonzhka et al., 2007) (Figure 1.3). 1.3.2 Ctip2 expression in adult mouse skin Similar to embryonic skin, in 8-10 week-old mouse skin, Ctip2 is expressed in most of the cells in the basal layer, and some cells in the suprabasal layer and hair follicles. However, Ctip2 can be barely detected in the dermal compartment. Compared with E18.5 embryos, the expression level is much lower in the adult skin (Golonzhka et al., 2007). Ctip2 is also expressed in the bulge region of adult mouse hair follicles, which harbour stem cells and contribute to the generation of hair follicle and inter-follicular epidermis (Golonzhka et al., 2007). 1.3.3 Ctip2 in epidermal barrier formation As Ctip2 null mice die after birth, it’s likely that these mice have epidermal defects, together with other developmental abnormality. An X-gal diffusion assay was performed to determine the barrier function at 17 E17.5 and E18.5. Ctip2 null mice, but not wildtype, show strong X-gal staining at E17.5, especially on the ventral surface and head, indicating a delayed dorsal to ventral barrier establishment in the null mice (Golonzhka et al., 2008) (Figure 1.4.A). Similar assays performed at E18.5 didn’t reveal any significant difference between wildtype and null mice (Figure 1.4.A). Trans-epidermal water loss (TEWL) studies indicate that Ctip2 null embryos have significant higher water loss compared with wildtype at E17.5 and E18.5 (Figure 1.4.B). Ctip2 null mice also exhibited reduced epidermal thickness at E17.5 with defects in proliferation and differentiation (Golonzhka et al., 2008) (Figure 1.4.D-E). Real-time quantitive PCR (RT-qPCR) studies performed on wildtype and Ctip2 null mice revealed downregulation of genes involved in epidermal terminal differentiation and barrier formation in the null mice, such as transglutaminase-1 (Tgase1), Gata3, and caspase-14 (Golonzhka et al., 2008). Hence, Ctip2 regulates epidermal barrier formation during embryonic development by modulating the expression of genes involved in barrier formation and terminal differentiation (Golonzhka et al., 2008). 18 1.3.4 Ctip2 in epidermal surface lipid distribution By Nile Red staining, dense and evenly distributed neutral lipids were stained in the wildtype fetus along the cornified layer, while uneven and thinner neutral lipid layer was observed in the Ctip2 null mice at E17.5 (Golonzhka et al., 2008) (Figure 1.4.C). The ultrastructure of the epidermis also revealed defects in lipid disc formation, loosely packed corneocytes, as well as disorganized intercellular lamellar body membranes in the cornified layer of Ctip2 null mice compared with wildtype (Golonzhka et al., 2008). These results indicated impaired lipid distribution following ablation of Ctip2 during development (Golonzhka et al., 2008). The defects in neutral lipid distribution and irregular lamellar body formation in the mutant mice could be due to changed gene expression profile. Several genes involved in lipid homeostasis were studied with RT-qPCR. Dgat2, Elovl4, eLox3 and Alox12b were downregulated in the null embryos at E17.5, suggesting that Ctip2 could regulate the expression of lipid homeostasis genes, thus contributing to lipid barrier formation during development (Golonzhka et al., 2008). 1.3.5 Ctip2 and cornified envelop (CE) formation Ctip2 also has an important role in epidermal barrier formation by regulating the formation and integrity of CEs (Figure 1.5, unpublished 19 data). CE suspension was sonicated for different lengths of time and the morphology of the CEs were monitored using a phase contrast microscope. The mutant CEs were less in number, morphologically abnormal and irregular in shape compared to the wildtypes. Also, the mutant CEs were more fragile during sonication, and were completely destroyed after 2 minutes of sonication. Thus, Ctip2 may regulate the epidermal barrier by involving in the formation of functional and integral CEs. 1.4 CTIP2 and skin diseases 1.4.1 Expression of CTIP2 in human head and neck squamous cell carcinoma (HNSCC) Head and neck cancer is the 6th most common cancer in the world. In United States, head and neck cancer accounts for less than 5% of all cancers and less than 3% of all cancer deaths. The most common type of head and neck cancer is head and neck squamous cell carcinoma (HNSCC) (Hunter et al., 2005). Disappointingly, the survival rate of this disease cannot be improved even with aggressive and multidisciplinary treatment and medicine currently available. The localization of CTIP2 in 20 human HNSCC was studied by immunohistochemical staining (GanguliIndra et al., 2009b). In tumor adjacent normal epithelium tissue, CTIP2 is exclusively observed in the nucleus of basal layer keratinocytes. In dysplasia samples, the expression of CTIP2 is much stronger than normal tissue, with extended expression in the spinous layer. In welldifferentiated tumors, CTIP2 expresses in basal layer of the keratinized horn pearl. Moderately and poorly differentiated tumors showed homologous and strong CTIP2 staining (Ganguli-Indra et al., 2009b). Overall, increased expression of CTIP2 correlated well with the progression of carcinoma (Ganguli-Indra et al., 2009b). CTIP2 expression has been reported to be linked to poor differentiation status of the tumor (Ganguli-Indra et al., 2009b). The expression level of CTIP2 in human cell lines confirmed above results. Low level of CTIP2 is found in primary human epidermal keratinocytes (Hek-a) and in spontaneously immortalized non- tumorigenic human skin keratinocytes (Hacat), while high expression of CTIP2 has been detected in head and neck carcinoma cell lines (SCC4, SCC25, etc) and other carcinomas (Ganguli-Indra et al., 2009b). Altogether, results indicate that CTIP2 expression is linked to HNSCC progression. 21 1.4.2 Role of Ctip2 in cutaneous wound healing Cutaneous wound healing is a highly organized process involving crosstalk between different cell types, epidermal barrier homeostasis recovery, as well as re-epithelialization carried out by cell migration and proliferation (Gurtner et al., 2008). Various pathways regulate this process with key factors such as integrins, stem cell markers, growth factors, cytokines as well as other regulatory proteins (Werner et al., 2007; Barrientos et al., 2008; Liang et al., 2012). Tape stripping creates a model for mechanical skin injury in mouse skin. It induces transient epidermal hyperplasia with altered epidermal proliferation and differentiation (Oyoshi et al., 2010). Another model to study more several mechanical injuries is to create an excisional wound in mouse skin and monitor the healing process by measurement of the gap between the migratory tongues from the wound site (Wong et al., 2011). In wildtype mice, the expression level of Ctip2 increases after 24-48 hours post tape stripping, and 7-11 days post wounding, mainly expressed in epidermal basal layer as well as post-mitotic suprabasal layers during later phases of wound healing (Liang et al., 2012). Since Ctip2 null mice die after birth, a new mouse line was generated in which Ctip2 is selectively deleted in mouse epidermal keratinocytes (Ctip2ep-/-) using Cre-LoxP strategies and K14/Cre deletor mice (Indra et 22 al., 1999; Golonzhka et al., 2008). Ctip2ep-/- mice exhibit increased epidermal proliferation and terminal differentiation in unwounded skin. After wounds were created, the mutant mice show significantly delayed wound healing with lower re-epithelialization efficiency compared with wildtype. Also, Ctip2ep-/- mice have thick and blunted migratory tongues in the wound margin while the wildtypes have thinner ones. The mutant mice also have impaired keratinocyte activation and altered cell proliferation and differentiation, which could contribute to the delayed cutaneous wound healing (Liang et al., 2012). Altogether, Ctip2 plays a critical role in mouse skin wound healing and re-epithelialization by regulating keratinocyte activation, proliferation and differentiation in a cell-autonomous manner(Liang et al., 2012). During mice skin wound healing, the expression of alpha SMA which is responsible for development of extracellular matrix (ECM) was reduced in the mutant on day 5 after wounding. Several stem cell markers, such as K15, CD133 as well as CD34 were significantly downregulated in the Ctip2ep-/- mice 5 days post wounding. These results indicate aberrant expression levels of regulatory proteins in the mouse cutaneous wound healing process in the absence of Ctip2 (Liang et al., 2012). 23 1.5 Research objectives In the study described under Chapter 2, we have evaluated how Ctip2 regulates lipid composition in mouse epidermis in connection to the role of Ctip2 in epidermal permeability barrier formation. The molecular mechanisms of Ctip2 mediated regulation of sphingolipids biosynthesis were also addressed. In Chapter 3, our study focused on the regulation of adult skin barrier maintenance and homeostasis by Ctip2. The role of Ctip2 in atopic dermatitis-like mouse skin inflammatory disease was studied. Key factors involved in this disease progression were identified. 24 Figure 1.1 Schematic representation of epidermal permeability barrier. (Adapted from Segre et al., 2006). Epidermal keratinocytes undergo a series of proliferation and differentiation from basal cells to spinous cells to enucleated granular cells, resulting finally in differentiated squames in the stratum corneum, which is composed of keratin macrofibrils and cross-linked cornified envelopes (CEs). Tight junction also plays an important role in epidermal permeability barrier. 25 Figure 1.2 Amino acid sequence, diagram of amino acid sequence alignment of CTIP1 and CTIP2. (Taken from Avram et al., 2000) (A) and (B) Amino acid sequence of CTIP1 and CTIP2, respectively. The conserved cysteine and histidine in zinc finger motifs for both proteins are underlined. (C) Schematic diagram of CTIP1 and CTIP2 amino acid alignment. Black boxes indicate homologous regions between CTIP1 and CTIP2, and percentage of identity is listed. 26 27 Figure 1.3 Expression of Ctip2 in the mouse fetal skin. (Adapted from Golonzhka et al., 2007) (A) Highly expressed Ctip2 (green) was observed in the ectoderm at E10.5 (upper panel) and E12.5 (lower panel) and is co-localized with the expression of K14 (red). (B) Ctip2 expression was mainly in the basal cells and upper layers of the epidermis of E14.5 (upper panel), E16.5 (middle panel) and E18.5 embryos (lower panel). Ctip2 (red) was co-stained with K14 (basal layer) and K10 (suprabasal layer) (in red). At E16.5 and E18.5 stages of development CTIP2 is highly expressed in the basal layer of epidermis as well as in the dermis and hair follicles. All sections were counterstained with DAPI (in blue). Images were taken at 40X magnification. EC: ectoderm, M: mesoderm, E: epidermis, D: dermis, B: basal cell layer, SB: suprabasal cell layers, HB: hair bulb, HF: hair follicle. 28 !" #" %" " " " " " &" $" 29 Figure 1.4 Ctip2 mutant mice exhibit permeability barrier defects and epidermal hypoplasia. (Adapted from Golonzhka et al., 2007) (A) X-gal diffusion assay performed on wildtype and Ctip2 null mice at E17.5 and E18.5. (B) Transepidermal water loss (TEWL) measurement of E17.5 and E18.5 embryonic dorsal and ventral skin. *P<0.05, # not statistically significant. (C) Nile red staining of wildtype and Ctip2 null mice at E18.5. Yellow ribbon indicates neutral lipid layer. Yellow arrows indicate loss of neutral lipid in the Ctip2 null epidermis. Scale bar: 50 µm. E, epidermis; D, dermis. (D) Immunohistochemical staining of wildtype and null mice dorsal skin at E18.5 with proliferation markers Ki67 (red), K14 (red) and early differentiation marker K10 (red). (E) Immunohistochemical staining of wildtype and null mice dorsal skin at E18.5 with terminal differntiation markers Filaggrin (red), Involucrin (red) and Loricrin (red). All sections were counterstained with DAPI (blue). Scale bar: 100 µm. 30 Figure 1.5 Ctip2 is required for the formation and integrity of cornified envelope in mouse epidermis. Pictures represent the morphology of cornified envelope extracted from wildtype and Ctip2ep-/- after different time of sonication. 31 1.6 References Arlotta, P., Molyneaux, B.J., Chen, J., Inoue, J., Kominami, R., and Macklis, J.D. (2005). Neuronal Subtype-Specific Genes that Control Corticospinal Motor Neuron Development In Vivo. Neuron 45, 207–221. Avram, D., Fields, A., Pretty On Top, K., Nevrivy, D.J., Ishmael, J.E., and Leid, M. (2000). Isolation of a novel family of C(2)H(2) zinc finger proteins implicated in transcriptional repression mediated by chicken ovalbumin upstream promoter transcription factor (COUP-TF) orphan nuclear receptors. J Biol Chem 275, 10315–10322. Avram, D., Fields, A., Senawong, T., Topark-Ngarm, A., and Leid, M. (2002). COUP-TF (chicken ovalbumin upstream promoter transcription factor)-interacting protein 1 (CTIP1) is a sequence-specific DNA binding protein. Biochem J 368, 555–563. B. Brandt, E., and Sivaprasad, U. (2011). Th2 Cytokines and Atopic Dermatitis. Journal of Clinical & Cellular Immunology 02,. Barrientos, S., Stojadinovic, O., Golinko, M.S., Brem, H., and TomicCanic, M. (2008). PERSPECTIVE ARTICLE: Growth factors and cytokines in wound healing. Wound Repair and Regeneration 16, 585– 601. Byrne, C., Hardman, M., and Nield, K. (2003). Covering the limb-formation of the integument. J Anat 202, 113–123. Cameron, D.J., Tong, Z., Yang, Z., Kaminoh, J., Kamiyah, S., Chen, H., Zeng, J., Chen, Y., Luo, L., and Zhang, K. (2007). Essential role of Elovl4 in very long chain fatty acid synthesis, skin permeability barrier function, and neonatal survival. Int. J. Biol. Sci. 3, 111–119. Candi, E., Schmidt, R., and Melino, G. (2005). The cornified envelope: a model of cell death in the skin. Nat. Rev. Mol. Cell Biol. 6, 328–340. Comeau, M.R., and Ziegler, S.F. (2010). The influence of TSLP on the allergic response. Mucosal Immunol 3, 138–147. Cork, M.J., Danby, S.G., Vasilopoulos, Y., Hadgraft, J., Lane, M.E., Moustafa, M., Guy, R.H., MacGowan, A.L., Tazi-Ahnini, R., and Ward, 32 S.J. (2009). Epidermal Barrier Dysfunction in Atopic Dermatitis. Journal of Investigative Dermatology 129, 1892–1908. Demehri, S., Liu, Z., Lee, J., Lin, M.-H., Crosby, S.D., Roberts, C.J., Grigsby, P.W., Miner, J.H., Farr, A.G., and Kopan, R. (2008). NotchDeficient Skin Induces a Lethal Systemic B-Lymphoproliferative Disorder by Secreting TSLP, a Sentinel for Epidermal Integrity. PLoS Biology 6, e123. Elias, P.M. (1983). Epidermal lipids, barrier function, desquamation. J. Invest. Dermatol. 80 Suppl, 44s–49s. and Elias, P.M., and Menon, G.K. (1991). Structural and lipid biochemical correlates of the epidermal permeability barrier. Adv. Lipid Res 24, 1– 26. Evans, R.M. (1988). The steroid and thyroid hormone receptor superfamily. Science 240, 889–895. Feingold, K.R. (2007). Thematic review series: skin lipids. The role of epidermal lipids in cutaneous permeability barrier homeostasis. J Lipid Res 48, 2531–2546. Feingold, K.R. (2009). The outer frontier: the importance of lipid metabolism in the skin. J. Lipid Res. 50 Suppl, S417–422. Fuchs, E., and Raghavan, S. (2002). Getting under the skin of epidermal morphogenesis. Nat Rev Genet 3, 199–209. Ganguli-Indra, G., Liang, X., Hyter, S., Leid, M., Hanifin, J., and Indra, A.K. (2009a). Expression of COUP-TF-interacting protein 2 (CTIP2) in human atopic dermatitis and allergic contact dermatitis skin. Experimental Dermatology 18, 994–996. Ganguli-Indra, G., Wasylyk, C., Liang, X., Millon, R., Leid, M., Wasylyk, B., Abecassis, J., Indra, A.K., and Indra, A. (2009b). CTIP2 expression in human head and neck squamous cell carcinoma is linked to poorly differentiated tumor status. PLoS ONE 4, e5367. Gelfand, J.M., Dommasch, E.D., Shin, D.B., Azfar, R.S., Kurd, S.K., Wang, X., and Troxel, A.B. (2009). The Risk of Stroke in Patients with Psoriasis. Journal of Investigative Dermatology 129, 2411–2418. 33 Gelfand, J.M., Neimann, A.L., Shin, D.B., Wang, X., Margolis, D.J., and Troxel, A.B. (2006). Risk of Myocardial Infarction in Patients With Psoriasis. JAMA: The Journal of the American Medical Association 296, 1735–1741. Ghadially, R., Reed, J.T., and Elias, P.M. (1996). Stratum Corneum Structure and Function Correlates with Phenotype in Psoriasis. Journal of Investigative Dermatology 107, 558–564. Golonzhka, O., Leid, M., Indra, G., and Indra, A.K. (2007). Expression of COUP-TF-interacting protein 2 (CTIP2) in mouse skin during development and in adulthood. Gene Expr Patterns 7, 754–760. Golonzhka, O., Liang, X., Messaddeq, N., Bornert, J.-M., Campbell, A.L., Metzger, D., Chambon, P., Ganguli-Indra, G., Leid, M., and Indra, A.K. (2008). Dual Role of COUP-TF-Interacting Protein 2 in Epidermal Homeostasis and Permeability Barrier Formation. J Investig Dermatol 129, 1459–1470. Golonzhka, O., Metzger, D., Bornert, J.-M., Bay, B.K., Gross, M.K., Kioussi, C., and Leid, M. (2009). Ctip2/Bcl11b controls ameloblast formation during mammalian odontogenesis. Proc. Natl. Acad. Sci. U.S.A. 106, 4278–4283. Gudjonsson, J.E., Ding, J., Johnston, A., Tejasvi, T., Guzman, A.M., Nair, R.P., Voorhees, J.J., Abecasis, G.R., and Elder, J.T. (2010). Assessment of the Psoriatic Transcriptome in a Large Sample: Additional Regulated Genes and Comparisons with In Vitro Models. J Investig Dermatol 130, 1829–1840. Gurtner, G.C., Werner, S., Barrandon, Y., and Longaker, M.T. (2008). Wound repair and regeneration. Nature 453, 314–321. Hardman, M.J., Sisi, P., Banbury, D.N., and Byrne, C. (1998). Patterned acquisition of skin barrier function during development. Development 125, 1541–1552. Holleran, W.M., Takagi, Y., and Uchida, Y. (2006). Epidermal sphingolipids: metabolism, function, and roles in skin disorders. FEBS Lett. 580, 5456–5466. 34 Hunter, K.D., Parkinson, E.K., and Harrison, P.R. (2005). Opinion: Profiling early head and neck cancer. Nature Reviews Cancer 5, 127– 135. Imokawa, G. (2009). A possible mechanism underlying the ceramide deficiency in atopic dermatitis: Expression of a deacylase enzyme that cleaves the N-acyl linkage of sphingomyelin and glucosylceramide. Journal of Dermatological Science 55, 1–9. Indra, A.K., Warot, X., Brocard, J., Bornert, J.M., Xiao, J.H., Chambon, P., and Metzger, D. (1999). Temporally-controlled site-specific mutagenesis in the basal layer of the epidermis: comparison of the recombinase activity of the tamoxifen-inducible Cre-ER(T) and CreER(T2) recombinases. Nucleic Acids Res. 27, 4324–4327. Ishikawa, J., Narita, H., Kondo, N., Hotta, M., Takagi, Y., Masukawa, Y., Kitahara, T., Takema, Y., Koyano, S., Yamazaki, S., et al. (2010). Changes in the Ceramide Profile of Atopic Dermatitis Patients. Journal of Investigative Dermatology 130, 2511–2514. Kamimura, K., Mishima, Y., Obata, M., Endo, T., Aoyagi, Y., and Kominami, R. (2007). Lack of Bcl11b tumor suppressor results in vulnerability to DNA replication stress and damages. Oncogene 26, 5840–5850. Kastner, P., Chan, S., Vogel, W.K., Zhang, L.-J., Topark-Ngarm, A., Golonzhka, O., Jost, B., Le Gras, S., Gross, M.K., and Leid, M. (2010). Bcl11b represses a mature T-cell gene expression program in immature CD4(+)CD8(+) thymocytes. Eur. J. Immunol. 40, 2143–2154. Karanam, N.K., Grabarczyk, P., Hammer, E., Scharf, C., Venz, S., Gesell-Salazar, M., Barthlen, W., Przybylski, G.K., Schmidt, C.A., and Völker, U. (2010). Proteome analysis reveals new mechanisms of Bcl11b-loss driven apoptosis. J. Proteome Res. 9, 3799–3811. Kashyap, M., Rochman, Y., Spolski, R., Samsel, L., and Leonard, W.J. (2011). Thymic stromal lymphopoietin is produced by dendritic cells. J. Immunol. 187, 1207–1211. Klinge, C.M., Bodenner, D.L., Desai, D., Niles, R.M., and Traish, A.M. (1997). Binding of type II nuclear receptors and estrogen receptor to full and half-site estrogen response elements in vitro. Nucleic Acids Res. 25, 1903–1912. 35 Kruse, S.W., Suino-Powell, K., Zhou, X.E., Kretschman, J.E., Reynolds, R., Vonrhein, C., Xu, Y., Wang, L., Tsai, S.Y., Tsai, M.-J., et al. (2008). Identification of COUP-TFII Orphan Nuclear Receptor as a Retinoic Acid–Activated Receptor. PLoS Biology 6, e227. Leid, M., Ishmael, J.E., Avram, D., Shepherd, D., Fraulob, V., and Dolle, P. (2004). CTIP1 and CTIP2 are differentially expressed during mouse embryogenesis. Gene Expr Patterns 4, 733–739. Leung, D.Y., and Bieber, T. (2003). Atopic dermatitis. The Lancet 361, 151–160. Li, M. (2005). Retinoid X receptor ablation in adult mouse keratinocytes generates an atopic dermatitis triggered by thymic stromal lymphopoietin. Proceedings of the National Academy of Sciences 102, 14795–14800. Li, M. (2006). Topical vitamin D3 and low-calcemic analogs induce thymic stromal lymphopoietin in mouse keratinocytes and trigger an atopic dermatitis. Proceedings of the National Academy of Sciences 103, 11736–11741. Li, W., Sandhoff, R., Kono, M., Zerfas, P., Hoffmann, V., Ding, B.C.-H., Proia, R.L., and Deng, C.-X. (2007). Depletion of ceramides with very long chain fatty acids causes defective skin permeability barrier function, and neonatal lethality in ELOVL4 deficient mice. Int. J. Biol. Sci. 3, 120–128. Liang, X., Bhattacharya, S., Bajaj, G., Guha, G., Wang, Z., Jang, H.-S., Leid, M., Indra, A.K., and Ganguli-Indra, G. (2012). Delayed Cutaneous Wound Healing and Aberrant Expression of Hair Follicle Stem Cell Markers in Mice Selectively Lacking Ctip2 in Epidermis. PLoS ONE 7, e29999. Linja, M.J. (2004). Expression of Androgen Receptor Coregulators in Prostate Cancer. Clinical Cancer Research 10, 1032–1040. Lippens, S., Hoste, E., Vandenabeele, P., Agostinis, P., and Declercq, W. (2009). Cell death in the skin. Apoptosis 14, 549–569. Liu, P., Keller, J.R., Ortiz, M., Tessarollo, L., Rachel, R.A., Nakamura, T., Jenkins, N.A., and Copeland, N.G. (2003). Bcl11a is essential for normal lymphoid development. Nature Immunology 4, 525–532. 36 Mack, J.A., Anand, S., and Maytin, E.V. (2005). Proliferation and cornification during development of the mammalian epidermis. Birth Defects Res C Embryo Today 75, 314–329. Mangelsdorf, D.J., Thummel, C., Beato, M., Herrlich, P., Schütz, G., Umesono, K., Blumberg, B., Kastner, P., Mark, M., Chambon, P., et al. (1995). The nuclear receptor superfamily: the second decade. Cell 83, 835–839. De Monte, L., Reni, M., Tassi, E., Clavenna, D., Papa, I., Recalde, H., Braga, M., Di Carlo, V., Doglioni, C., and Protti, M.P. (2011). Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J. Exp. Med. 208, 469–478. Nakamura, T., Yamazaki, Y., Saiki, Y., Moriyama, M., Largaespada, D.A., Jenkins, N.A., and Copeland, N.G. (2000). Evi9 encodes a novel zinc finger protein that physically interacts with BCL6, a known human B-cell proto-oncogene product. Mol. Cell. Biol. 20, 3178–3186. Nemes, Z., Demény, M., Marekov, L.N., Fésüs, L., and Steinert, P.M. (2000). Cholesterol 3-sulfate interferes with cornified envelope assembly by diverting transglutaminase 1 activity from the formation of cross-links and esters to the hydrolysis of glutamine. J. Biol. Chem. 275, 2636–2646. Nemes, Z., and Steinert, P.M. (1999). Bricks and mortar of the epidermal barrier. Exp. Mol. Med. 31, 5–19. Nograles, K.E., Brasington, R.D., and Bowcock, A.M. (2009). New insights into the pathogenesis and genetics of psoriatic arthritis. Nature Clinical Practice Rheumatology 5, 83–91. Oda, Y., Uchida, Y., Moradian, S., Crumrine, D., Elias, P.M., and Bikle, D.D. (2008). Vitamin D Receptor and Coactivators SRC2 and 3 Regulate Epidermis-Specific Sphingolipid Production and Permeability Barrier Formation. J Investig Dermatol 129, 1367–1378. Olefsky, J.M. (2001). Nuclear receptor minireview series. J. Biol. Chem. 276, 36863–36864. Oyoshi, M.K., Larson, R.P., Ziegler, S.F., and Geha, R.S. (2010). Mechanical injury polarizes skin dendritic cells to elicit a T(H)2 37 response by inducing cutaneous thymic stromal lymphopoietin expression. J. Allergy Clin. Immunol. 126, 976–984, 984.e1–5. Pastorcic, M., Wang, H., Elbrecht, A., Tsai, S.Y., Tsai, M.J., and O’Malley, B.W. (1986). Control of transcription initiation in vitro requires binding of a transcription factor to the distal promoter of the ovalbumin gene. Mol. Cell. Biol. 6, 2784–2791. Pereira, F.A., Qiu, Y., Tsai, M.J., and Tsai, S.Y. (1995). Chicken ovalbumin upstream promoter transcription factor (COUP-TF): expression during mouse embryogenesis. J. Steroid Biochem. Mol. Biol. 53, 503–508. Roberson, E.D.O., and Bowcock, A.M. (2010). Psoriasis genetics: breaking the barrier. Trends in Genetics 26, 415–423. Robinson-Rechavi, M., Escriva Garcia, H., and Laudet, V. (2003). The nuclear receptor superfamily. J. Cell. Sci. 116, 585–586. Sagami, I., Tsai, S.Y., Wang, H., Tsai, M.J., and O’Malley, B.W. (1986). Identification of two factors required for transcription of the ovalbumin gene. Mol. Cell. Biol. 6, 4259–4267. Sankaran, V.G., Menne, T.F., Xu, J., Akie, T.E., Lettre, G., Van Handel, B., Mikkola, H.K.A., Hirschhorn, J.N., Cantor, A.B., and Orkin, S.H. (2008). Human Fetal Hemoglobin Expression Is Regulated by the Developmental Stage-Specific Repressor BCL11A. Science 322, 1839– 1842. Satterwhite, E. (2001). The BCL11 gene family: involvement of BCL11A in lymphoid malignancies. Blood 98, 3413–3420. Scharschmidt, T.C., and Segre, J.A. (2008). Modeling atopic dermatitis with increasingly complex mouse models. J. Invest. Dermatol. 128, 1061–1064. Schmuth, M., Man, M.Q., Weber, F., Gao, W., Feingold, K.R., Fritsch, P., Elias, P.M., and Holleran, W.M. (2000). Permeability barrier disorder in Niemann-Pick disease: sphingomyelin-ceramide processing required for normal barrier homeostasis. J. Invest. Dermatol. 115, 459–466. Segre, J.A. (2006). Epidermal barrier formation and recovery in skin disorders. J Clin Invest 116, 1150–1158. 38 Soumelis, V., Reche, P.A., Kanzler, H., Yuan, W., Edward, G., Homey, B., Gilliet, M., Ho, S., Antonenko, S., Lauerma, A., et al. (2002). Human epithelial cells trigger dendritic cell–mediated allergic inflammation by producing TSLP. Nature Immunology. Topark-Ngarm, A., Golonzhka, O., Peterson, V.J., Barrett, B., Martinez, B., Crofoot, K., Filtz, T.M., and Leid, M. (2006). CTIP2 associates with the NuRD complex on the promoter of p57KIP2, a newly identified CTIP2 target gene. J Biol Chem 281, 32272–32283. Tsai, S.Y. (1997). Chick Ovalbumin Upstream Promoter-Transcription Factors (COUP-TFs): Coming of Age. Endocrine Reviews 18, 229–240. Uchida, Y., Hara, M., Nishio, H., Sidransky, E., Inoue, S., Otsuka, F., Suzuki, A., Elias, P.M., Holleran, W.M., and Hamanaka, S. (2000). Epidermal sphingomyelins are precursors for selected stratum corneum ceramides. J. Lipid Res. 41, 2071–2082. Uda, M., Galanello, R., Sanna, S., Lettre, G., Sankaran, V.G., Chen, W., Usala, G., Busonero, F., Maschio, A., Albai, G., et al. (2008). Genome-wide association study shows BCL11A associated with persistent fetal hemoglobin and amelioration of the phenotype of thalassemia. Proceedings of the National Academy of Sciences 105, 1620–1625. Vasireddy, V., Uchida, Y., Salem, N., Jr, Kim, S.Y., Mandal, M.N.A., Reddy, G.B., Bodepudi, R., Alderson, N.L., Brown, J.C., Hama, H., et al. (2007). Loss of functional ELOVL4 depletes very long-chain fatty acids (> or =C28) and the unique omega-O-acylceramides in skin leading to neonatal death. Hum. Mol. Genet. 16, 471–482. Wakabayashi, Y., Watanabe, H., Inoue, J., Takeda, N., Sakata, J., Mishima, Y., Hitomi, J., Yamamoto, T., Utsuyama, M., Niwa, O., et al. (2003). Bcl11b is required for differentiation and survival of alphabeta T lymphocytes. Nat Immunol 4, 533–539. Wang, L.H., Tsai, S.Y., Sagami, I., Tsai, M.J., and O’Malley, B.W. (1987). Purification and characterization of chicken ovalbumin upstream promoter transcription factor from HeLa cells. J. Biol. Chem. 262, 16080–16086. 39 Werner, S., Krieg, T., and Smola, H. (2007). Keratinocyte-fibroblast interactions in wound healing. J Invest Dermatol 127, 998–1008. Wong, V.W., Sorkin, M., Glotzbach, J.P., Longaker, M.T., and Gurtner, G.C. (2011). Surgical Approaches to Create Murine Models of Human Wound Healing. Journal of Biomedicine and Biotechnology 2011, 1–8. Yamada, T., Saito, H., Kimura, Y., Kubo, S., Sakashita, M., Susuki, D., Ito, Y., Ogi, K., Imoto, Y., and Fujieda, S. (2012). CpG-DNA suppresses poly(I:C)-induced TSLP production in human laryngeal arytenoid fibroblasts. Cytokine 57, 245–250. Ziegler, S.F. (2010). The role of thymic stromal lymphopoietin (TSLP) in allergic disorders. Current Opinion in Immunology 22, 795–799. (1999). A unified nomenclature system for the nuclear receptor superfamily. Cell 97, 161–163. 40 Chapter 2 Transcription factor Ctip2 controls epidermal lipid metabolism and regulates expression of genes involved in sphingolipid biosynthesis during skin development Zhixing Wang, Jay S. Kirkwood, Alan W. Taylor, Jan F. Stevens, Mark Leid, Gitali Ganguli-Indra and Arup K. Indra Journal of Investigative Dermatology In revision 41 2.1 Abstract The stratum corneum is composed of a continuous sheath of proteinenriched corneocytes embedded in an intercellular matrix enriched in nonpolar lipids organized as lamellar layers and give rise to the epidermal permeability barrier (EPB). EPB defects play an important role in the pathophysiology of skin diseases such as eczema. The transcriptional control of skin lipid metabolism is poorly understood. We have discovered that mouse lacking a transcription factor COUP-TF interacting protein 2 (Ctip2) exhibit EPB defects including altered keratinocyte terminal differentiation, delayed skin barrier development and interrupted neutral lipid distribution in the epidermis. We adapted herein a targeted lipidomics approach using mass spectrometry, and have determined that Ctip2-/- mice (germline deletion of Ctip2 gene) display altered composition of major epidermal lipids such as ceramides and sphingomyelins compared to the wildtype at different stages of skin development. Interestingly, expressions of several genes involved in skin sphingolipid biosynthesis and metabolism were altered in mutant skin. Ctip2 was found to be recruited to the promoter region of a subset of those genes, suggesting their possible direct regulation by Ctip2. Our Ctip2 mouse model exhibiting EPB defects can be utilized to screen for new drugs for treatment and recovery of barrier dysfunctions. 42 2.2 Introduction The exterior layer of skin, stratum corneum (SC), plays a critical role in skin permeability barrier formation and maintenance (Fuchs and Raghavan, 2002; Elias, 2004). It forms a protective barrier against external stress and environmental insults. Skin barrier defects are caused not only by altered levels of protein(s) involved in keratinocytes terminal differentiation, cross-linking of cornified envelops (CEs), but also by impaired formation and maintenance of skin lipid barrier (Elias and Menon, 1991; Nemes and Steinert, 1999; Feingold, 2007). Mammalian SC contains a large number of lipids, such as ceramides, sphingomyelins, cholesterols and fatty acids (Elias and Menon, 1991; Ishikawa et al., 2010). Ceramides (CERs) are the most abundant lipid types in SC (50% by weight) and can be divided into at least 11 species (Oda et al., 2008; Ishikawa et al., 2010). It has been shown that reduced content of CERs leads to epidermal water loss as well as skin epidermal dysfunctions, and causes diseases such as atopic dermatitis (AD) or eczema (Imokawa, 2009; Ishikawa et al., 2010). Spingomyelins are another large group of epidermal sphingolipids, which have been reported as precursors of ceramides (Schmuth et al., 2000; Uchida et al., 2000; Holleran et al., 2006). Cholesterols and fatty acids are also major skin lipids, and cholesterol 3-sulfate (CSO4) is a ubiquitous 43 metabolite of cholesterol, which is involved in cornified envelope formation by interacting with transglutaminase1 (Nemes et al., 2000). Ceramide biosynthesis is regulated at multiple levels. Four pathways have been reported so far, de novo pathway, sphingomyelinase pathway, salvage pathway and exogenous ceramide recycling pathway (Kitatani et al., 2008). In de novo pathway, serine and fatty acyl-CoA are the initiators of a series of reactions. Ceramide synthases 1-6 (Lass1-Lass6) play an important role since each of them has specificities on synthesizing certain ceramides (Mizutani et al., 2009). Other enzymes such as palmitoyltransferases (Sptlc1-3), dihydroceramide desaturase (Degs1) and ketodihydro-sph reductase (Kdsr) are also key enzymes in de novo pathway. The salvage pathway has been reported recently as a complex mechanism, commencing as the catabolism of complex sphingolipids into glucosylceramid by glucosylceramide synthase (Ugcg), and followed by the formation of ceramide by acid beta-glucosidase (Gba2) (Ogretmen, 2002; Becker, 2004). These ceramides can be broken down into sphingosines by acid ceramidase (Asah1 and Asah2), which are reused to produce ceramides. In sphingomyelinase pathway, ceramide is formed by hydrolysis of sphingomyelins, and the reverse reaction is controlled by sphingomyelin synthase (Marchesini and Hannun, 2004). 44 Exogenous short chain ceramide can also be utilized to generate sphingosine, thus leads to the synthesis of endogenous long chain ceramides (Ogretmen, 2002; Sultan et al., 2006). COUP-TF-interacting protein 2 (Ctip2), also known as Bcl11b, is a C2H2 zinc finger protein expressed in many organs and tissues including developing mouse epidermis (Avram et al., 2000, 2002; Golonzhka et al., 2008). It has been reported that Ctip2 null mice exhibit altered epidermal proliferation and late terminal differentiation, as well as barrier defects during embryogenesis (Golonzhka et al., 2008). The barrier defects were correlated with altered surface lipid distribution in the absence of Ctip2 (Golonzhka et al., 2008). Liquid chromatography-tandem mass spectrometry (LC/MS/MS) has been used to characterize ceramide species in mouse and human stratum corneum (Masukawa et al., 2008; Park et al., 2009; Ishikawa et al., 2010; Janssens et al., 2011; van Smeden et al., 2011). Here we adopted the LC/MS/MS methodology to elucidate the CER [NS], sphingmyelin and sphingosine profile in wildtype and Ctip2 null (Ctip2-/-) embryonic skin epidermis. The expression levels of genes encoding various enzymes involved in sphingolipid metabolism were analyzed by RT-qPCR and immuno-blotting in wildtype and Ctip1-null skin. Furthermore, Ctip2 recruitment to the promoter regions of several 45 genes that were altered in Ctip2-/- mice was evaluated by chromatin immunoprecipitation (ChIP) analyses. Results indicated that Ctip2 control sphingolipid metabolism in the developing skin by directly or indirectly regulating expression of a subset of genes involved in the sphingolipid bio-synthesis pathways. 2.3 Materials and methods Mice Ctip2 null (Ctip2-/-) mice were generated by floxing exon 4 of Ctip2 locus, which encodes for three-fourth of open reading frame (ORF) (Golonzhka et al., 2008). OSU IACUC approval was obtained for animal experiments. Lipid extraction Fetus from E16.5 to E18.5 and neonatal mice were collected and genotyped (Golonzhka et al., 2008). A minimum of three wildtype and Ctip2-/- fetuses were taken from the same litter and compared between littermates. A minimum of three litters were analyzed from each developmental stages. Skin was isolated, weighed and incubated overnight in CnT-07 medium (CELLnTEC, Bern, Switzerland) 46 containing 1 mg ml−1 dispase (Gibco, Grand Island, NY) at 4 °C to dissociate epidermis. Raw skin lipid was extracted using Bligh and Dyer method (Folch et al., 1957; Bligh and Dyer, 1959). Briefly, skin epidermis was incubated in extraction buffer (chloroform: methanol: water (1:2:0.8), 10-15mg tissue/1ml extraction buffer) overnight at room temperature. Fresh extraction buffer was added to epidermis samples the second day and old extraction buffer was saved. Samples were sonicated for 5 cycles, 30 seconds each, and the suspension was shaked for 30min in room temperature. After centrifuged at 2000rpm for 10min, the pellets were discarded and the extraction buffer was combined. Chloroform and water were added to extraction buffer in a ratio of extraction buffer: chloroform: water (7.6: 2: 2). After mixing and centrifuging, upper phase was discarded and the lower phase (chloroform) was washed twice with chloroform: methanol: water (1: 1: 0.9). Lower phase was dried in nitrogen, weighed and saved in -20°C. LC-MS/MS Raw skin lipid was further purified as in previous reports (Merrilljr et al., 2005; Monette et al., 2011). 10µl sphingolipid internal standard mixture (Avanti, Alabaster, Alabama) and 1mg raw lipid were added to 1.5ml of 47 chloroform: methanol (1:2) and sonicated in the bath sonicator for 30 seconds. After 2 hours of incubation at 48 °C, the samples were cooled on ice. Samples were then incubated for 1 hour at 37°C with 75μl 1M KOH in methanol to remove the free fatty acids and triacylglycerol, and cooled on ice. 12μl of glacial acetic acid was added to neutralize the pH. Phase separation was achieved by adding 2ml chloroform and 4ml water and samples were gently vortexed. After centrifuge for 15 min at 2000rcf, the chloroform layer was aspirated and dried under nitrogen. The samples were reconstituted in 200μl chloroform: methanol (3:1) and diluted 1:4 with acetonitrile: methanol: acetic acid (97:2:1) with 5mM ammonium acetate. LC-tandem mass spectrometry was performed and relative quantification was based on skin weight and sphingolipid standards (Monette et al., 2011). In brief, lipids were separated by HPLC using a Supelco Discovery column (2mm x 50mm; Sigma-Aldrich) with 300μl/min flow rate. Mobile phase A: methanol: water (60:40) and mobile phase B: methanol: chloroform (60:40) both contained 0.2% (v/v) formic acid and 10 mM ammonium acetate. The column was preequilibrated at 100% phase A and 5μl sample was injected; 100% mobile phase A was maintained for 1min, followed by a linear increase to 40% mobile phase B in a 7 min period; followed by a linear increase 48 to 70% mobile phase B in next 6 min, which was maintained for the remainder (6min) of the 20 min run. Sample detection was performed using a triple-quadrupole mass spectrometer operated in positive mode (Applied Biosystems/ MDS Sciex, API300) with multiple reaction monitoring. Quantitation was based on comparison to a synthetic sphingolipid standard mixture (Monette et al., 2010). UPLC-MS/MS Ultra high pressure liquid chromatography was performed on a Shimadzu Nexera system (Shimadzu, Columbia, MD) coupled to a hybrid quadrupole-time of flight mass spectrometer (TripleTOFTM 5600, AB SCIEX) with raw skin lipids. Chromatographic separations were carried out on a Brownlee Analytical C18 column (50 × 2.1 mm, 1.9 mm; PerkinElmer, Boston, MA). The flow rate was 0.35 mL/min and mobile phases consisted of water (A) and acetonitrile (B), both with 0.1% formic acid. Elution gradient was as follows: 0 min, 25% B; 2 min, 55% B; 7 min, 100 % B; 9 min, 100% B. The column was equilibrated 3 minutes prior to each injection. Column temperature was held at 40 °C and the injection volume was 3 µL. 49 Mass spectrometry was performed on an AB SCIEX TripleTOFTM 5600 equipped with an electrospray ionization source. The instrument was operated in information dependent MS/MS acquisition mode with the collision energy set at 30 V and a collision energy spread of 10 V. TOF MS acquisition time was 0.15 seconds and MS/MS acquisition time was 0.10 seconds. Scan range was 70-1000 m/z for TOF MS and 40-1000 m/z for MS/MS. Ion source gas 1 and 2 and curtain gas (all nitrogen) were set at 50 and 40 and 25, respectively. Source temperature was set at 500 °C and IonSpray voltage at 5.5/-5.5 kV. A set of standards was run periodically throughout the batch to assess system variance and stability. The instrument was automatically calibrated every five runs. Stearic acid, palmitic acid, cholesterol, and cholesterol-3-sulfate were identified using authentic standards with accurate mass measurements, isotope distribution, MS/MS fragmentation, and retention time. RT-qPCR RNA was extracted and cDNA was synthesized from fetus and neonatal skin (Indra et al., 2005b; Wang et al., 2011). Real-time PCR was performed on an ABI 7500 Real-Time PCR system using SYBR green methodology (Indra et al., 2005a, 2005b). Relative gene expression 50 analysis of the RT-qPCR data was performed using Gapdh as an internal control, and the specific primers were indicated in Table 2.1 (Wang et al., 2011). All assays were performed in triplicates. Western blotting Protein was extracted from mouse skin biopsies and western blotting was performed due to previous reports using specific antibodies against Lass2, Gba2 and eLox3. (Wang et al., 2011; Santa Cruz Biotechnology, Santa Cruz, CA). Briefly, skin samples were homogenized, and proteins were extracted with a lysis buffer (150mM NaCl, 50mM Tris pH 7.5, 5mM EDTA, and 1% Nonidet P-40, 0.1% SDS, 0.5% sodium deoxycholate) containing protease inhibitors (2µg/ml Aprotinin, 2µg/ml Leupeptin, 100µg/ml Pefabloc and 1µg/ml Pepstatin). Equal amounts of protein extract (25 µg) from each lysate were resolved using SDS polyacrylamide-gel electrophoresis and transferred onto a nitrocellulose membrane. Blots were blocked overnight with 5% nonfat dry milk and incubated with specific antibodies. All western blot experiments were done in triplicates, using a minimum of three biological replicates from each group of mice. Chromatin immunoprecipitation (ChIP) 51 ChIP assay was performed according to Hyter et al., 2010 with minor modifications. Briefly, mouse keratinocytes were isolated from neonatal wildtype and Ctip2-/- mice skin epidermis and fixed in 1% formaldehyde. Each ChIP started with 3x106 cells, which are sonicated and immunoprecipitated with 2µg anti-Ctip2 antibody (Abcam, Cambridge, MA) overnight at 4°C. Rat IgG was used as control. 25ml protein G beads (Invitrogen, Carlsbad, CA) were added to each ChIP and incubated at 4°C for four hours. DNA was uncrosslinked at 65°C overnight and purified by QIAquick PCR Purification Kit (Qiagen, Germantown, MD). DNA was amplified using ABI-7500 Real-time PCR machine with specific primers indicated in Supplementary table 2. Statistics Statistical significance of differences between groups was analyzed using GraphPad Prism software using student’s unpaired t-test. All statistical analyses were performed in a double-blinded manner. 2.4 Results 52 2.4.1 Altered profile of sphingolipids in the developing epidermis of Ctip2-null mice We hypothesized that impaired epidermal permeability barrier in the Ctip2-null (Ctip2-/- ) mice could be at least in part due to altered skin lipid composition. In order to determine the lipid composition of embryonic skin, wildtype and Ctip2-null epidermis were isolated at different developmental stages, E16.5, E17.5 E18.5 and neonatal (P0). Lipidomic studies were then performed using LC/MS/MS technique and absolute quantities of the different skin sphingolipids were analyzed (for details see materials and methods). Both ceramides and sphingomyelins were abundant during mouse skin development, especially during the earlier stages of skin development (E16.5, E17.5) (Figure 2.1-2.4). Ceramide C14:0 level remained very low from E16.5-P0. Long chain ceramide C16:0 was the most abundant saturated ceramide at E16.5 and was 2-fold higher in the mutant epidermis compared with wildtype (Figure 2.1 A). Significant induction of ceramide C18:0 was observed in the E16.5 mutant skin (Figure 2.1 A). The amount of long chain ceramides (C16:0, C18:0, C20:0 and C22:0) was in a very low abundance at E17.5-E18.5, with no discernable difference observed between wildtype and mutant skin; however, their quantities are larger at P0, each having a ~2-fold reduction in the Ctip2-/- skin (Figure 2.1 B- 53 D). At E17.5 and E18.5, very long chain ceramide C26:0 became the dominant subtype in mouse skin, modestly reduced at E17.5 and 2-fold decreased at E18.5 in Ctip2-/- skin (Figure 2.1 B-C). Modest decrease of ceramide C24:0 was also observed in Ctip2-/- epidermis at P0 (Figure 2.1 D). Unsaturated ceramides were less abundant compared with saturated ceramides in the wild type epidermis. Long chain unsaturated ceramide C16:1 was significantly reduced at E16.5 in the mutant epidermis, unaffected at E17.5 and E18.5, with a 2-fold decrease in P0 mutant epidermis (Figure 2.2 A-D). Among saturated sphingomyelins, long chain sphingomyelins C16:0, C20:0 and C22:0 were the most abundant subtypes in embryonic epidermis at most of the stages, which were highly induced in the mutant skin, especially at E17.5 and E18.5 (Figure 2.3 B and C). Very long chain sphingomyelins (C24:1, C26:1, C28:1) were also increased in the mutant skin at E16.5-E18.5 (Figure 2.3 A-C). unsaturated sphingomyelins were less Interestingly, predominant in all developmental stages; however, they showed the similar trend as saturated ones, with higher amount in embryonic mutant epidermis. As early as E16.5, long chain unsaturated sphingomyelin C24:1 was significantly elevated in Ctip2-/- skin (Figure 2.4 A). Higher quantities of 54 long chain sphingomyelins C20:1, C22:1, and C24:1 were noted in the mutant epidermis at E17.5 and E18.5 (Figure 2.4 B-D). The profiles of sphingosine and sphinganine were also determined by LC/MS/MS. Compared with ceramides and sphingomyelins, the quantity of free sphingoid bases were smaller in embryonic skin, with no significant change observed between wildtype and mutant epidermis at all developmental stages (Figure 2.5 A-D). We also performed UPLC/MS/MS analyses to determine the relative amount of cholesterols and fatty acids. Levels of cholesterol and cholesterol 3-sulfate were significantly reduced in the epidermis of Ctip2-/- mice at E16.5, but were unaltered at all later timepoints (Figure 2.6 A-B). Although the amount of palmitic acid and stearic acid was similar between wildtype and null skin at E16.5-E18.5, their levels doubled in the P0 Ctip2-/- epidermis compared to the wildtype epidermis (Figure 2.6 C-D). Altogether, our results indicate that epidermal lipid composition is altered in absence of transcriptional regulator Ctip2. 2.4.2 Ctip2 directly regulates expression of a subset of genes involved in lipid metabolism in the developing skin Previous studies have identified four major pathways involved in ceramide synthesis: de novo pathway, salvage pathway, sphingomyelinase pathway and exogenous ceramide-recycling pathway 55 (Kitatani et al., 2008; Mizutani et al., 2009). We hypothesized that altered sphingolipid profiles in mutant mice could be due to altered expression of genes encoding proteins involved in the sphingolipid metabolism. To test that, we evaluated the expression of genes involved in the different ceramide biosynthesis pathways by RT-qPCR analyses in the wild type control and mutant skin (for details see materials and methods). Expressions of ceramide synthase 3 (Lass3), palmitoyltransferases (Sptlc1 and Sptlc3) were downregulated at E16.5 and E17.5, while ceramide synthases Lass1 and Lass2 were significantly reduced at E18.5 in Ctip2-/- skin (Figure 2.7 A-C). Lass6 was consistently downregulated from E16.5 to E18.5 in the mutant skin (Figure 2.7 A-C). Thus, genes encoding enzymes in de novo pathway were altered in ablation of Ctip2. Similarly, transcript levels of enzymes involved in sphingomyelinase pathway such as sphingomyelinases (Smpd1 and Smpd2) were significantly downregulated at E18.5, whereas sphingomyelin synthase (Sgms2) level was upregulated at E16.5 in null embryos compared to the wildtypes (Figure 2.7 D-F). Expression of glucosylceramide synthase (Ugcg), which turns complex sphingolipids into glucosylcerarimde, was also reduced at E18.5 while that of acid beta-glucosidase (Gba2), which is involved in salvage pathway, was downregulated in skin of null mice from E16.5 to E18.5 56 (Figure 2.7 D-F). Ceramidases are involved in the breaking down of short-chain ceramide, facilitating its conversion to sphingosine. In particular, reduced expression of acid ceramidase 2 (Asah 2) and alkaline ceramidase 1 (Acer1) was observed in all developmental stages in the mutant epidermis (Figure 2.7 G-I). Levels of all other members of acid ceramidases (Asah 1 and Asah 3) and alkaline ceramidases (Acer 2 and Acer 3) were unaltered in the mutants at all stages of development (Figure 2.7 G-I). Similarly, sphingosine kinases (Sphk1, Sphk2) are responsible for the disposal of sphingosine to sphingosine-1-phosphate, whose expression level was reduced at E16.5 in the mutant epidermis (Figure 2.7 G). No significant changes in the relative mRNA levels of several other enzymes involved in ceramide synthesis pathways was observed between wildtype and Ctip2-/embryonic skin (Figure 2.7 G-I, 2.8 A-C). Furthermore, protein levels of Lass2, Gba2 and eLox3 were significantly reduced in the Ctip2 null skin compared with the wildtype skin at E18.5 (Figure 2.9 A-B) (Golonzhka et al., 2008). Taken together, our results indicate that expression of a subset of genes encoding enzymes involved in the sphingolipid biosynthesis pathways was altered during skin organogenesis in absence of Ctip2. 57 To investigate whether some of the lipid-metabolizing genes, which are dysregulated in the mutant skin, are direct targets of transcriptional regulator Ctip2, we conducted in vivo chromatin immunoprecipitation (ChIP) assay on neonatal (P0) mouse epidermal keratinocyte extracts. The promoter regions of several genes encoding lipid-metabolizing enzymes such as Lass1-3, 6, Sptlc1-3, Smpd1-2, Ugcg, Gba2, and eLox3 (Golonzhka et al., 2008), whose expressions were altered in the Ctip2-/- skin, were selected and primers were designed to determine possible Ctip2 binding within -6 kb upstream of transcription starting sites (TSS). Our results indicated that Ctip2 was recruited within -1 kb promoter region of Lass2, Gba2 and eLox3, as well as between 5-6 kb promoter region of eLox3 relative to TSS (Figure 2.10 A-C). However, we did not observe the direct recruitment of Ctip2 to the promoter regions of several other lipid-metabolizing genes (Figure 2.11 A-K). Our results indicate that Ctip2 is directly recruited to the promoter region of a subset of lipid metabolizing genes and possibly regulates their expression in the developing epidermis. Altogether, results suggest that Ctip2 directly or indirectly regulates expression of a subset of lipid metabolizing genes and control epidermal sphingolipid biosynthesis during skin organogenesis. 58 2.5 Discussion The establishment of an effective physical barrier protecting the living organism from its surrounding environment is the key function of mammalian skin (Hoffjan and Stemmler, 2007). The transport of most substances across the stratum corneum (SC) takes place through the lipid bilayer, suggesting its essential role for the epidermal barrier function (Jungersted et al., 2008). The major SC lipids, ceramides, cholesterol and fatty acids have distinct roles in maintaining normal barrier functions (Elias and Menon, 1991; Ishikawa et al., 2010). Ceramides as well as cholesterol were shown to have an influence on (and are influenced by) the integrity of the SC, while free fatty acids play a major role in the lipid bilayer formation and pH maintenance (Baroni et al., 2012). We have identified Ctip2 as a key regulator of skin lipid metabolism, which plays an important role in establishment of epidermal permeability barrier (EPB) during development. Ablation of Ctip2 leads to altered lipid composition in the developing mouse epidermis, such as reduced ceramide at E17.5-P0, as well as increased sphingomyelins at E16.5-E18.5. Ctip2 regulate sphingolipid homeostasis in mouse embryonic skin by modulating the expression of several key enzymes involved in lipid metabolism. Here we demonstrate that transcription factor Ctip2 is recruited to the promoter 59 regions of a subset of genes (elox3, Lass2 and Gba2) involved in the sphingolipid biosynthesis pathways and could directly regulate their expression (Figure 2.12). Skin barrier defects are caused not only by altered levels of protein(s) involved in keratinocytes terminal differentiation, cross-linking of cornified envelops (CEs), formation of intercellular junctions, but also by impaired formation and maintenance of skin lipid barrier (Furuse et al., 2002; Kirschner and Brandner, 2012). Our previous studies have shown that Ctip2 regulate keratinocyte proliferation and late terminal differentiation in a cell-autonomous manner; the ultrastructural analyses of the epidermis have revealed defects in lipid disc formation, loosely packed corneocytes, as well as disorganized intercellular lamellar body membranes in Ctip2 null mice (Golonzhka et al., 2008 and unpublished data). In this study, we have focused on the role of Ctip2 in controlling epidermal lipid barrier by modulating lipid metabolism. Herein, we have applied a mass spectrometry method to systemically detect and profile ceramides, sphingomyelins, sphingosines and sphinganines simultaneously in murine epidermis. As a crucial member of stratum corneum lipids, there are few reports on profiling of skin sphingomyelins. Our present data confirm deregulated levels of both long chain and very long chain saturated and unsaturated ceramides in 60 the mutant epidermis at different stages of skin development. Along the same line, overall levels of long chain and very long chain saturated and unsaturated sphingomyelins were significantly enhanced in the mutants at all stages analyzed. In the present study, we have specifically looked at ceramide [NS] but were unable to perform massspectrometry study on other subclasses such as NP, NH, AS, AP, AH, EOS, EOP and EOH, due to the lack of available deuterated ceramide internal standards for each subclasses. Additional analyses for the different subclasses will be performed in future using specific internal standard for each subclass. Our results indicate that altered expression(s) of ceramide synthases 16 (Lass 1-6), sphingomyelinases (Smpd1 and Smpd2), glucosylceramide synthase (Ugcg) and that of acid beta-glucosidase (Gba2) at different stages of skin development contribute to an overall decrease in the epidermal ceramide levels in the mutant embryonic skin. Lass 1-6 have been reported to be involved in UVB-induced apoptosis and their expression was increased after UVB irradiation in normal human keratinocytes, but reduced in aged skin (Uchida et al., 2003; Jensen et al., 2005). A recent study has reported that Lass3 mutant mice die after birth due to increased skin barrier defects, characterized by increased TEWL, loss of ultra-long chain ceramides as 61 well as non-functional cornified envelopes (Jennemann et al., 2011). Upregulation of sphingomyelin synthase (Sgms2) in Ctip2-/- mice may partially account for increased amount of sphingomyelins observed at later stages in the mutant skin (Figure 2.3 B-C(Wang et al., 2011)). Furthermore, loss of Gba2, whose expression is altered in the mutant epidermis, has been reported to lead to barrier defects in Gaucher’s disease patients (Holleran et al., 1994). Similarly, eLox3 as well as its substrate 12R-lipoxygenase (12R-LOX) are involved in the formation of oxidized ceramides in mouse epidermis; 12R-LOX deficient mice die shortly after birth due to severe barrier abnormalities with absence of protein-bound EOS ceramide (Zheng et al., 2011). Interestingly, we have reported earlier that eLox3 expression is downregulated in the Ctip2-/- embryonic skin (Golonzhka et al., 2008). Here we have furthered our studies and have shown that Ctip2 could directly regulate the expression of eLox3, Gba2 and Lass2 in mouse embryonic skin by recruitment to their promoter regions. Altogether, we have identified Ctip2 as a key regulator of several lipid metabolizing genes and hence epidermal sphingolipid biosynthesis during skin development. Sphingolipids are key components in skin barrier homeostasis, as well as in other cellular processes such as cell cycle arrest, apoptosis and stress responses (Hannun, 1996; Ogretmen and Hannun, 2004). 62 Ceramide is a central metabolite, whose biosynthesis is regulated at multiple levels with spatial separation of enzymes involved in ceramide formation (Futerman and Riezman, 2005; Kitatani et al., 2008). It remains unclear that if alteration of the enzymes in one of the pathways could have an effect on other pathways, and finally modulate the biological functions. It would be interesting to systemically compare the lipid composition in healthy subjects vs patients with skin disorders such as atopic dermatitis and psoriasis. It is likely possible that besides Ctip2, other regulatory pathways are also involved in controlling ceramide biosynthesis. B-cell lymphoma/leukemia 11A (Bcl11a/Ctip1) is a homolog of Ctip2, which was found to be upregulated in Ctip2 null mice at E18.5 (Golonzhka et al., 2008). The role of Ctip1 in epidermal barrier development and maintenance is still unclear. Further investigation about the expression pattern and distribution of Ctip1, as well as its role in lipid barrier homeostasis in mouse epidermis will be performed utilizing Ctip1-/- mice. 63 E16.5 150 100 N-acyl chain length 28 100 28 26 24 22 28 26 24 N-acyl chain length 20 50 0 22 20 18 16 14 * WT Ctip2-/- 18 50 150 16 * 100 D 14 WT Ctip2-/- Ceramide Content (pmol/mg skin) Ceramide Content (pmol/mg skin) 26 P0 150 0 24 N-acyl chain length E18.5 C 22 0 20 50 18 26 24 22 * 20 18 16 0 * WT Ctip2-/- 200 16 100 250 14 150 50 B Ceramide Content (pmol/mg skin) WT Ctip2-/- 14 Ceramide Content (pmol/mg skin) 200 28 A E17.5 N-acyl chain length Figure 2.1 Liquid chromatography tandem mass-spectrometry (LC/MS/MS) analyses of mouse epidermal saturated ceramides during skin development. Epidermal sphingolipids were extracted from skin of (A) E16.5; (B) E17.5; (C) E18.5 embryos and (D) P0 wildtype and Ctip2-/- mice. Amount of saturated ceramides was determined according to their Nacyl chain length using LC/MS/MS analyses. Statistical analyses were performed by student’s unpaired t-test using Graphpad Prism software. *p<0.05. 64 E16.5 15 WT Ctip2-/- 10 N-acyl chain length N-acyl chain length 28:1 26:1 WT Ctip2-/- 30 20 28:1 26:1 24:1 22:1 20:1 0 18:1 10 16:1 28:1 26:1 24:1 22:1 20:1 18:1 16:1 5 40 14:1 10 D Ceramide Content (pmol/mg skin) WT Ctip2-/- 14:1 Ceramide Content (pmol/mg skin) 24:1 P0 15 0 22:1 N-acyl chain length E18.5 C 20:1 0 18:1 5 16:1 26:1 24:1 22:1 20:1 18:1 0 16:1 5 14:1 Ceramide Content (pmol/mg skin) 10 B 14:1 WT Ctip2-/- * Ceramide Content (pmol/mg skin) 15 28:1 A E17.5 N-acyl chain length Figure 2.2 Liquid chromatography tandem mass-spectrometry (LC/MS/MS) analyses of mouse epidermal unsaturated ceramides during skin development. Epidermal sphingolipids were extracted from skin of (A) E16.5; (B) E17.5; (C) E18.5 embryos and (D) P0 wildtype and Ctip2-/- mice. Amount of unsaturated ceramides was determined according to their Nacyl chain length using LC/MS/MS analyses. Statistical analyses were performed by student’s unpaired t-test using Graphpad Prism software. *p<0.05. 65 E16.5 100 50 N-acyl chain length 28 26 24 22 0 20 28 26 24 22 20 18 16 * 50 18 20 100 16 * WT Ctip2-/- 14 ** 40 150 Sphingomyelin Content (pmol/mg skin) WT Ctip2-/- ** 14 Sphingomyelin Content (pmol/mg skin) 28 P0 D 60 0 26 N-acyl chain length E18.5 C 24 N-acyl chain length * 22 0 28 26 24 22 20 18 0 16 * 150 20 500 WT Ctip2-/- 200 18 1000 250 16 * B 14 WT Ctip2-/- Sphingomyelin Content (pmol/mg skin) 1500 14 Sphingomyelin Content (pmol/mg skin) A E17.5 N-acyl chain length Figure 2.3 LC/MS/MS analyses of mouse epidermal saturated sphingomyelins during skin development. Epidermal sphingolipids were extracted from skin of (A) E16.5; (B) E17.5; (C) E18.5 embryos and (D) P0 wildtype and Ctip2-/- mice. Amount of saturated sphingomyelins was determined according to their N-acyl chain length using LC/MS/MS analyses. Statistical analyses were performed by student’s unpaired t-test using Graphpad Prism software. *p<0.05; **p<0.005. 66 E16.5 60 40 N-acyl chain length N-acyl chain length 15 WT Ctip2-/- 10 28:1 26:1 24:1 22:1 20:1 18:1 0 16:1 5 14:1 28:1 26:1 24:1 22:1 20:1 18:1 16:1 2 Sphingomyelin Content (pmol/mg skin) * 14:1 Sphingomyelin Content (pmol/mg skin) D WT Ctip2-/- *** 0 28:1 P0 6 4 26:1 N-acyl chain length E18.5 C 24:1 ** 20 0 28:1 26:1 24:1 22:1 20:1 16:1 0 18:1 ** * 80 22:1 100 WT Ctip2-/- 20:1 200 100 18:1 300 B 16:1 WT Ctip2-/- 14:1 * Sphingomyelin Content (pmol/mg skin) 400 14:1 Sphingomyelin Content (pmol/mg skin) A E17.5 N-acyl chain length Figure 2.4 LC/MS/MS analyses of mouse epidermal unsaturated sphingomyelins during skin development. Epidermal sphingolipids were extracted from skin of (A) E16.5; (B) E17.5; (C) E18.5 embryos and (D) P0 wildtype and Ctip2-/- mice. Amount of unsaturated sphingomyelins was determined according to their N-acyl chain length using LC/MS/MS analyses. Statistical analyses were performed by student’s unpaired t-test using Graphpad Prism software. *p<0.05; **p<0.005; ***p<0.001. 67 E16.5 A E17.5 WT Ctip2-/- B 25 Lipid Content (pmol/mg skin) Lipid Content (pmol/mg skin) 6 4 2 WT Ctip2-/- 20 15 10 5 0 C 16 18 20 16 18 20 Sphingosine Sphinganine 16 18 20 16 18 20 Sphingosine Sphinganine N-acyl chain length N-acyl chain length E18.5 P0 15 WT Ctip2-/- D 10 Lipid Content (pmol/mg skin) Lipid Content (pmol/mg skin) 0 10 5 WT Ctip2-/- 8 6 4 2 0 16 18 20 16 18 20 Sphingosine Sphinganine N-acyl chain length 0 16 18 20 16 18 20 Sphingosine Sphinganine N-acyl chain length Figure 2.5 LC/MS/MS analyses of mouse epidermal sphingosines and sphinganines during skin development. Epidermal sphingosines and sphinganines were measured in the skin of (A) E16.5, (B) E17.5, (C) E18.5 embryos and (D) P0 wildtype and Ctip2-/- mice. The amount of skin sphingosines and sphinganines were determined according to their N-acyl chain length by LC/MS/MS analyses, quantified utilizing sphingosine and sphinganine standards, and expressed as pmol/ mg of skin (for details see methods). Statistical analyses were performed by student’s unpaired t-test using Graphpad Prism software. The results depicted in this figure are representative of at least two additional studies that have been conducted using different litters of mice of identical genotypes. 68 200 P0 E18.5 100 P0 E18.5 stearic acid 300 WT Ctip2-/- 200 * 100 0 P0 300 E17.5 0 E18.5 WT Ctip2-/- 400 peak area /skin dry weight (mg) 500 0 5000 D palmitic acid * 10000 E17.5 P0 E18.5 E17.5 0 E16.5 50 * 15000 E16.5 100 WT Ctip2-/- 20000 E17.5 150 cholesterol 3-sulfate 25000 E16.5 WT Ctip2-/- peak area /skin dry weight (mg) 200 C peak area /skin dry weight (mg) B cholesterol E16.5 peak area /skin dry weight (mg) A Figure 2.6 UPLC/MS/MS analyses of mouse epidermal cholesterol and fatty acids during skin development. Epidermal lipids were extracted from skin of E16.5, E17.5, E18.5 embryos and P0 wildtype and Ctip2-/- mice. Amount of (A) cholesterol, (B) cholesterol 3-sulfate, (C) palmitic acid and (D) stearic acid were determined using UPLC/MS/MS analyses. Statistical analyses were performed by student’s unpaired t-test using Graphpad Prism software. *p<0.05; **p<0.005. 69 * * 0.5 * ** Acer3 Acer1 Acer2 Sphk2 0.0 1.0 * ** ** 0.0 * * Lass1 Lass2 ** Sptlc3 Lass6 Sptlc1 Lass3 WT 0.0 WT 1.5 Ctip2-/- 1.0 * * Smpd2 * 0.5 Smpd1 2.0 1.5 Gba2 WT Ctip2-/- 1.0 0.5 Ugcg WT 0.0 Sgms2 Relative Expression Level Relative Expression Level Sptlc3 Lass6 Sptlc1 Lass3 Lass2 WT Lass1 Ctip2-/- 0.5 0.5 * * * 0.0 Acer3 1.0 I WT 1.5 Ctip2-/- 1.0 WT Asah1 Asah2 Sphk1 Sphk2 Acer1 Acer2 Ctip2-/- 0.0 Relative Expression Level H WT * Acer2 Acer3 Gba2 Ugcg Sgms2 Smpd2 Smpd1 WT 1.5 Asah2 Sphk1 Relative Expression Level 0.0 0.5 1.5 Gba2 * * Acer1 * 0.5 1.0 Ugcg 1.0 Ctip2-/- Sgms2 1.5 * WT F WT 1.5 Smpd2 Ctip2-/- * 0.0 Sphk1 Sphk2 WT 2.0 * 0.5 E Relative Expression Level Sptlc3 Lass6 Sptlc1 Lass3 Lass2 WT Lass1 0.0 WT Asah1 Relative Expression Level * ** 1.0 WT * 0.5 Ctip2-/- Smpd1 * 1.5 E18.5 C WT WT 1.0 D G Relative Expression Level Ctip2-/- Relative Expression Level Relative Expression Level 1.5 E17.5 B WT Asah1 Asah2 E16.5 A Figure 2.7 Ctip2 regulates the expression of genes encoding lipid metabolizing enzymes during skin development. Expression of (A, B, C) ceramide synthases (Lass1-Lass3, Lass6), serine palmitoyltransferases (Sptlc1, Sptlc3); (D, E, F) sphingomyeinases (Smpd1, Smpd2), sphingomyelin synthase 2 (Sgms2), glucosylceramide synthase (Ugcg) and acid beta-glucosidase (Gba2); (G, H, I) N-acylsphingosine amidohydrolases (acid ceramidases: Asah1, Asah2), sphingosine kinases (Sphk1, Sphk2) and alkaline ceramidases (Acer1, Acer2) was analyzed by RT-qPCR using specific primers indicated in table S1. All values represent relative transcript level after normalization with Gapdh transcripts. Statistical analyses were performed by student’s unpaired t-test using Graphpad Prism software. *p<0.05; **p<0.005. The results depicted in this figure are representative of at least three additional studies that have been conducted using numerous litters of mice of identical genotypes. 70 E16.5 1.0 Sgms1 Kdsr Figure 2.8 Relative expression of genes encoding Wang et al., 2012 metabolizing enzymes Figure duringS3, skin development. Degs1 Lass5 0.0 Sptlc2 0.5 WT Degs1 Sgms1 Kdsr Lass5 Sptlc2 0.0 WT 0.5 WT Ctip2-/- 1.5 Lass4 1.0 Relative Expression Level C WT Ctip2-/- 1.5 Lass4 Degs1 Sgms1 Kdsr Lass5 Sptlc2 Lass4 0.5 Relative Expression Level 1.0 0.0 E18.5 B WT Ctip2-/- 1.5 WT Relative Expression Level A E17.5 lipid- Expression of ceramide synthases (Lass4-5), serine palmitoyltransferase 2 (Sptlc2), dihydroceramide desaturase (Degs1), Ketodihydro-sph reductase (Kdsr) and sphingomyelin synthase 1 (Sgms1) was studied at (A) E16.5, (B) E17.5 and (C) E18.5 stages of development by RT-qPCR using specific primers indicated in table S1. Values represent relative transcript level after normalization with Gapdh transcripts. Statistical analyses were performed by student’s unpaired ttest using Graphpad Prism software. 71 Figure 2.9 Immunoblot analyses of lipid metabolizing enzymes in the developing murine epidermis. (A) Western blotting analyses was performed on protein extracted from E18.5 wildtype (WT) and Ctip2-/- mice epidermis to study the expression level of eLox3, Gba2 and Lass2 using specific antibodies. β-actin was used as an internal control. (B) Protein levels of eLox3, Gba2 and Lass2 were quantified in wildtype and mutant epidermis by densitometry analyses of western blots. All data was normalized by the corresponding β-actin levels. Statistical analyses were performed by student’s unpaired t-test using Graphpad Prism software. *p<0.05. The results depicted in this figure are representative of at least three additional studies that have been conducted using different litters of mice of identical genotypes. 72 8 6 4 30 ** 10 0 0 di st al pr ox im al 3' -U TR 5 di st al pr ox im al 3' -U TR 2 15 eLox3 IgG Ctip2 IgG Ctip2 ** 20 10 ** 0 di st al pr ox im al 3' -U TR * IgG Ctip2 C Gba2 Relative input % 10 Relative input % B Lass2 Relative input % A Figure 2.10 Ctip2 interacts directly with the promoter region of lipid metabolizing genes. Chromatin immunoprecipitation (ChIP) assay was performed on freshly isolated neonatal mouse skin keratinocytes using anti-Ctip2 antibody and results were analyzed by qPCR using primers indicated in table S2. Rat IgG was used as a control. Ctip2 was recruited to the promoter regions of (A) Lass2, (B) Gba2 and (C) eLox3. Statistical analyses were performed by student’s unpaired t-test using Graphpad Prism software. *p<0.05; **p<0.005. 0.20 0.15 0.10 0.05 0.00 0.0 Smpd3 H IgG Ctip2 0.3 0.2 0.1 0.0 IgG Ctip2 4 Dgat2 K 3 2 1 0 0.0 E Smpd1 F IgG Ctip2 1.5 1.0 0.5 0.0 Sgms2 I IgG Ctip2 3 2 1 0 Aloxe12b IgG Ctip2 pr ox im al 0.5 pr ox im al di st al 1.0 Relative input % IgG Ctip2 Relative input % pr ox im al C pr ox im al 0.5 2.0 di st al 1.0 1.5 di st al IgG Ctip2 Lass3 Relative input % Ugcg pr ox im al di st al 0.0 pr ox im al 0.4 di st al 0.5 Relative input % 1.0 B pr ox im al 0.5 di st al 1.5 Relative input % IgG Ctip2 Relative input % pr ox im al Lass1 di st al J pr ox im al di st al Relative input % 1.5 Relative input % G pr ox im al di st al Relative input % D pr ox im al di st al Relative input % A di st al Relative input % 73 Lass6 3 IgG Ctip2 2 1 0 Smpd2 2.0 IgG Ctip2 1.5 1.0 0.5 0.0 Elovl4 2.5 IgG Ctip2 2.0 1.5 1.0 0.5 0.0 74 Figure 2.11 ChIP analyses on murine keratinocytes for lipid metabolizing genes. Chromatin immunoprecipitation (ChIP) assay was performed on freshly isolated primary keratinocytes from neonatal mouse skin using antiCtip2 antibody and results were analyzed by qPCR using specific primers indicated in table S2. Rat IgG was used as a control. Ctip2 was not recruited to distal and proximal promoter regions of (A) Lass1, (B) Lass2, (C) Lass6, (D) Ugcg, (E) Smpd1, (F) Smpd2, (G) Smpd3, (H) Sgms2, (I) Elovl4, (J) Dgat2 and (K) Aloxe12b. 75 de novo pathway Salvage pathway palmitoyl-CoA L-serine complex sphingolipids Sptlc1 (E16.5-E17.5) Sptlc2 Sptlc3 (E16.5) glucosyl-cer ketodihydro-sph !" Kdsr dihydro-sph Lass4 Lass5 Lass6 (E16.5-E18.5) Gba2 (E16.5, E17.5) Ctip2 Lass1 (E18.5) Lass2 (E18.5) Lass3 (E16.5) dihydro-cer Exogenous ceramide recycling Ugcg (E18.5) Ceramide !" Degs1 Asah1 Asah2 (E16.5-E18.5) Ceramide Lass/CerSs Short chain ceramide Asah1 Asah2 (E16.5-E18.5) Acer1 (E16.5-E18.5) Acer2 Acer3 Sphingosine Asahs Acers Smpd1 (E16.5-E18.5) Smpd2 (E18.5) Sgms1 Sgms2 (E16.5) Sphk1 (E16.5) Sphk2 (E16.5) sphingomyelin Sphingomyelinase pathway Sphingosine1-phosphate Figure S5, Wang et al., 2012 Figure 2.12 Schematic representation of Ctip2 mediated differential regulation of genes encoding different enzymes of the ceramide synthesis pathways. The scheme indicates metabolic pathways for ceramide biosynthesis: de novo pathway, salvage pathway, sphingomyelinase pathway and exogenous ceramide recycling. Genes involved in these pathways are indicated. Stages at which altered gene expression were observed are indicated within brackets. Red: downregulation of gene expression; green: upregulation of gene expression. Ctip2 positively regulates Gba2 and Lass2 expressions by direct recruitment to their promoter regions. Expression of ceramide synthases 1-6 (Lass 1-6), serine palmitoyltransferases 1-3 (Sptlc 1-3), dihydroceramide desaturase (Degs1), Ketodihydro-sph reductase (Kdsr), glucosylceramide synthase (Ugcg), acid beta-glucosidase (Gba2), sphingomyelin synthases 1-2 (Sgms1-2), sphingomyeinases 1-2 (Smpd1, Smpd2),N-acylsphingosine amidohydrolases (acid ceramidases: Asah1, Asah2), alkaline ceramidases 1-2 (Acer1, Acer2), sphingosine kinases 1-2 (Sphk1, Sphk2) are indicated for the different pathways. 76 Table 2.1 Primer sequences used in q-PCR assays. 77 Table 2.2 Primer sequences used in ChIP assays. 78 2.6 References Avram, D., Fields, A., Pretty On Top, K., Nevrivy, D.J., Ishmael, J.E., and Leid, M. (2000). Isolation of a novel family of C(2)H(2) zinc finger proteins implicated in transcriptional repression mediated by chicken ovalbumin upstream promoter transcription factor (COUP-TF) orphan nuclear receptors. J Biol Chem 275, 10315–10322. Avram, D., Fields, A., Senawong, T., Topark-Ngarm, A., and Leid, M. (2002). COUP-TF (chicken ovalbumin upstream promoter transcription factor)-interacting protein 1 (CTIP1) is a sequence-specific DNA binding protein. Biochem J 368, 555–563. Baroni, A., Buommino, E., De Gregorio, V., Ruocco, E., Ruocco, V., and Wolf, R. (2012). Structure and function of the epidermis related to barrier properties. Clinics in Dermatology 30, 257–262. Becker, K.P. (2004). Selective Inhibition of Juxtanuclear Translocation of Protein Kinase C II by a Negative Feedback Mechanism Involving Ceramide Formed from the Salvage Pathway. Journal of Biological Chemistry 280, 2606–2612. Bligh, E.G., and Dyer, W.J. (1959). A rapid method of total lipid extraction and purification. Can J Biochem Physiol 37, 911–917. Elias, P.M. (2004). The epidermal permeability barrier: from the early days at Harvard to emerging concepts. J. Invest. Dermatol 122, xxxvi– xxxix. Elias, P.M., and Menon, G.K. (1991). Structural and lipid biochemical correlates of the epidermal permeability barrier. Adv. Lipid Res 24, 1– 26. Feingold, K.R. (2007). Thematic review series: skin lipids. The role of epidermal lipids in cutaneous permeability barrier homeostasis. J Lipid Res 48, 2531–2546. Folch, J., Lees, M., and Sloane Stanley, G.H. (1957). A simple method for the isolation and purification of total lipides from animal tissues. J. Biol. Chem. 226, 497–509. 79 Fuchs, E., and Raghavan, S. (2002). Getting under the skin of epidermal morphogenesis. Nat Rev Genet 3, 199–209. Furuse, M., Hata, M., Furuse, K., Yoshida, Y., Haratake, A., Sugitani, Y., Noda, T., Kubo, A., and Tsukita, S. (2002). Claudin-based tight junctions are crucial for the mammalian epidermal barrier: a lesson from claudin-1-deficient mice. J. Cell Biol. 156, 1099–1111. Futerman, A.H., and Riezman, H. (2005). The ins and outs of sphingolipid synthesis. Trends Cell Biol. 15, 312–318. Golonzhka, O., Liang, X., Messaddeq, N., Bornert, J.-M., Campbell, A.L., Metzger, D., Chambon, P., Ganguli-Indra, G., Leid, M., and Indra, A.K. (2008). Dual Role of COUP-TF-Interacting Protein 2 in Epidermal Homeostasis and Permeability Barrier Formation. J Investig Dermatol 129, 1459–1470. Hannun, Y.A. (1996). Functions of Ceramide in Coordinating Cellular Responses to Stress. Science 274, 1855–1859. Hoffjan, S., and Stemmler, S. (2007). On the role of the epidermal differentiation complex in ichthyosis vulgaris, atopic dermatitis and psoriasis. British Journal of Dermatology 157, 441–449. Holleran, W.M., Ginns, E.I., Menon, G.K., Grundmann, J.U., Fartasch, M., McKinney, C.E., Elias, P.M., and Sidransky, E. (1994). Consequences of beta-glucocerebrosidase deficiency in epidermis. Ultrastructure and permeability barrier alterations in Gaucher disease. J. Clin. Invest. 93, 1756–1764. Holleran, W.M., Takagi, Y., and Uchida, Y. (2006). Epidermal sphingolipids: metabolism, function, and roles in skin disorders. FEBS Lett. 580, 5456–5466. Hyter, S., Bajaj, G., Liang, X., Barbacid, M., Ganguli-Indra, G., and Indra, A.K. (2010). Loss of nuclear receptor RXRα in epidermal keratinocytes promotes the formation of Cdk4-activated invasive melanomas. Pigment Cell Melanoma Res 23, 635–648. Imokawa, G. (2009). A possible mechanism underlying the ceramide deficiency in atopic dermatitis: Expression of a deacylase enzyme that cleaves the N-acyl linkage of sphingomyelin and glucosylceramide. Journal of Dermatological Science 55, 1–9. 80 Indra, A.K., Dupe, V., Bornert, J.M., Messaddeq, N., Yaniv, M., Mark, M., Chambon, P., and Metzger, D. (2005a). Temporally controlled targeted somatic mutagenesis in embryonic surface ectoderm and fetal epidermal keratinocytes unveils two distinct developmental functions of BRG1 in limb morphogenesis and skin barrier formation. Development 132, 4533–4544. Indra, A.K., Mohan, W.S., Frontini, M., Scheer, E., Messaddeq, N., Metzger, D., and Tora, L. (2005b). TAF10 is required for the establishment of skin barrier function in foetal, but not in adult mouse epidermis. Dev Biol 285, 28–37. Ishikawa, J., Narita, H., Kondo, N., Hotta, M., Takagi, Y., Masukawa, Y., Kitahara, T., Takema, Y., Koyano, S., Yamazaki, S., et al. (2010). Changes in the Ceramide Profile of Atopic Dermatitis Patients. Journal of Investigative Dermatology 130, 2511–2514. Janssens, M., van Smeden, J., Gooris, G.S., Bras, W., Portale, G., Caspers, P.J., Vreeken, R.J., Kezic, S., Lavrijsen, A.P.M., and Bouwstra, J.A. (2011). Lamellar Lipid Organization and Ceramide Composition in the Stratum Corneum of Patients with Atopic Eczema. Journal of Investigative Dermatology 131, 2136–2138. Jennemann, R., Rabionet, M., Gorgas, K., Epstein, S., Dalpke, A., Rothermel, U., Bayerle, A., van der Hoeven, F., Imgrund, S., Kirsch, J., et al. (2011). Loss of ceramide synthase 3 causes lethal skin barrier disruption. Hum. Mol. Genet. Jensen, J.-M., Förl, M., Winoto-Morbach, S., Seite, S., Schunck, M., Proksch, E., and Schütze, S. (2005). Acid and neutral sphingomyelinase, ceramide synthase, and acid ceramidase activities in cutaneous aging. Exp. Dermatol 14, 609–618. Jungersted, J.M., Hellgren, L.I., Jemec, G.B.E., and Agner, T. (2008). Lipids and skin barrier function – a clinical perspective. Contact Dermatitis 58, 255–262. Kirschner, N., and Brandner, J.M. (2012). Barriers and more: functions of tight junction proteins in the skin. Ann. N. Y. Acad. Sci. 1257, 158– 166. 81 Kitatani, K., Idkowiak-Baldys, J., and Hannun, Y.A. (2008). The sphingolipid salvage pathway in ceramide metabolism and signaling. Cell. Signal 20, 1010–1018. Marchesini, N., and Hannun, Y.A. (2004). Acid and neutral sphingomyelinases: roles and mechanisms of regulation. Biochem. Cell Biol. 82, 27–44. Masukawa, Y., Narita, H., Shimizu, E., Kondo, N., Sugai, Y., Oba, T., Homma, R., Ishikawa, J., Takagi, Y., Kitahara, T., et al. (2008). Characterization of overall ceramide species in human stratum corneum. The Journal of Lipid Research 49, 1466–1476. Merrilljr, A., Sullards, M., Allegood, J., Kelly, S., and Wang, E. (2005). Sphingolipidomics: High-throughput, structure-specific, and quantitative analysis of sphingolipids by liquid chromatography tandem mass spectrometry. Methods 36, 207–224. Mizutani, Y., Mitsutake, S., Tsuji, K., Kihara, A., and Igarashi, Y. (2009). Ceramide biosynthesis in keratinocyte and its role in skin function. Biochimie 91, 784–790. Monette, J.S., Gómez, L.A., Moreau, R.F., Bemer, B.A., Taylor, A.W., and Hagen, T.M. (2010). Characteristics of the rat cardiac sphingolipid pool in two mitochondrial subpopulations. Biochemical and Biophysical Research Communications 398, 272–277. Monette, J.S., Gómez, L.A., Moreau, R.F., Dunn, K.C., Butler, J.A., Finlay, L.A., Michels, A.J., Shay, K.P., Smith, E.J., and Hagen, T.M. (2011). (R)-α-Lipoic acid treatment restores ceramide balance in aging rat cardiac mitochondria. Pharmacological Research 63, 23–29. Nemes, Z., Demény, M., Marekov, L.N., Fésüs, L., and Steinert, P.M. (2000). Cholesterol 3-sulfate interferes with cornified envelope assembly by diverting transglutaminase 1 activity from the formation of cross-links and esters to the hydrolysis of glutamine. J. Biol. Chem. 275, 2636–2646. Nemes, Z., and Steinert, P.M. (1999). Bricks and mortar of the epidermal barrier. Exp. Mol. Med. 31, 5–19. 82 Oda, Y., Uchida, Y., Moradian, S., Crumrine, D., Elias, P.M., and Bikle, D.D. (2008). Vitamin D Receptor and Coactivators SRC2 and 3 Regulate Epidermis-Specific Sphingolipid Production and Permeability Barrier Formation. J Investig Dermatol 129, 1367–1378. Ogretmen, B. (2002). Biochemical Mechanisms of the Generation of Endogenous Long Chain Ceramide in Response to Exogenous Short Chain Ceramide in the A549 Human Lung Adenocarcinoma Cell Line. ROLE FOR ENDOGENOUS CERAMIDE IN MEDIATING THE ACTION OF EXOGENOUS CERAMIDE. Journal of Biological Chemistry 277, 12960–12969. Ogretmen, B., and Hannun, Y.A. (2004). Biologically active sphingolipids in cancer pathogenesis and treatment. Nat. Rev. Cancer 4, 604–616. Park, H., Haynes, C.A., Nairn, A.V., Kulik, M., Dalton, S., Moremen, K., and Merrill, A.H. (2009). Transcript profiling and lipidomic analysis of ceramide subspecies in mouse embryonic stem cells and embryoid bodies. The Journal of Lipid Research 51, 480–489. Schmuth, M., Man, M.Q., Weber, F., Gao, W., Feingold, K.R., Fritsch, P., Elias, P.M., and Holleran, W.M. (2000). Permeability barrier disorder in Niemann-Pick disease: sphingomyelin-ceramide processing required for normal barrier homeostasis. J. Invest. Dermatol. 115, 459–466. van Smeden, J., Hoppel, L., van der Heijden, R., Hankemeier, T., Vreeken, R.J., and Bouwstra, J.A. (2011). LC/MS analysis of stratum corneum lipids: ceramide profiling and discovery. J. Lipid Res. 52, 1211–1221. Sultan, I., Senkal, C.E., Ponnusamy, S., Bielawski, J., Szulc, Z., Bielawska, A., Hannun, Y.A., and Ogretmen, B. (2006). Regulation of the sphingosine-recycling pathway for ceramide generation by oxidative stress, and its role in controlling c-Myc/Max function. Biochemical Journal 393, 513. Uchida, Y., Hara, M., Nishio, H., Sidransky, E., Inoue, S., Otsuka, F., Suzuki, A., Elias, P.M., Holleran, W.M., and Hamanaka, S. (2000). Epidermal sphingomyelins are precursors for selected stratum corneum ceramides. J. Lipid Res. 41, 2071–2082. 83 Uchida, Y., Nardo, A.D., Collins, V., Elias, P.M., and Holleran, W.M. (2003). De novo ceramide synthesis participates in the ultraviolet B irradiation-induced apoptosis in undifferentiated cultured human keratinocytes. J. Invest. Dermatol. 120, 662–669. Wang, Z., Coleman, D.J., Bajaj, G., Liang, X., Ganguli-Indra, G., and Indra, A.K. (2011). RXRα ablation in epidermal keratinocytes enhances UVR-induced DNA damage, apoptosis, and proliferation of keratinocytes and melanocytes. J. Invest. Dermatol. 131, 177–187. Zheng, Y., Yin, H., Boeglin, W.E., Elias, P.M., Crumrine, D., Beier, D.R., and Brash, A.R. (2011). Lipoxygenases Mediate the Effect of Essential Fatty Acid in Skin Barrier Formation: A PROPOSED ROLE IN RELEASING OMEGA-HYDROXYCERAMIDE FOR CONSTRUCTION OF THE CORNEOCYTE LIPID ENVELOPE. Journal of Biological Chemistry 286, 24046–24056. 84 Chapter 3 Selective Ablation of Ctip2/Bcl11b in Epidermal Keratinocytes Triggers Atopic Dermatitis-like Skin Inflammatory Responses in Adult Mice Zhixing Wang, Ling-juan Zhang, Gunjan Guha, Kateryna Kyrylkova, Chrissa Kioussi, Mark Leid, Gitali Ganguli-Indra, Arup K. Indra Submitted to PLoSOne 3.1 Abstract 85 Ctip2 is crucial for epidermal homeostasis and protective barrier formation in developing mouse embryos. Selective ablation of Ctip2 in mouse epidermis leads to increased transepidermal water loss (TEWL) from skin, impaired epidermal proliferation and terminal differentiation as well as altered lipid composition during development. However, little is known about the role of Ctip2 in adult mice skin homeostasis. To study the role of Ctip2 in adult skin homeostasis, we utilized a Ctip2ep-/mouse model in which Ctip2 is selectively deleted in epidermal keratinocytes. Measurement of TEWL, followed by histological, immuno-histochemical and RT-qPCR analyses on skin biopsies from control and mutant mice revealed an important role of Ctip2 in barrier maintenance and in regulating adult skin homeostasis. We have shown that keratinocytic ablation of Ctip2 leads to atopic dermatitis (AD)-like skin inflammation, characterized by alopecia, pruritus and dermatitis, as well as high infiltration of T lymphocytes and immune cells. We have also observed increased expression of Th2-type cytokines and chemokines in the mutant skin, as well as systemic immune responses that share similarity with human AD patients. Furthermore, we discovered that thymic stromal lymphopoietin (TSLP) expression is significantly upregulated in the mutant epidermis as early as postnatal day 1 and Ctip2 was recruited to the promoter region of the TSLP gene 86 in epidermal keratinocytes. The results suggest that upregulation of TSLP expression in the Ctip2ep-/- epidermis could be due to a derepression of gene transcription in absence of Ctip2. Thus, our data demonstrated a cell-autonomous role of Ctip2 in barrier maintenance and epidermal homeostasis in adult skin, as well as a non-cell autonomous role of keratinocytic Ctip2 in suppressing skin inflammatory responses by regulating the expression of Th2-type cytokines in adult mouse skin. Present results establish an initiating role of epidermal TSLP in AD pathogenesis via a novel repressive regulatory mechanism mediated by Ctip2. 87 3.2 Introduction Mammalian skin forms the first defense barrier in the body for protecting against physical injuries, ultraviolet radiation, bacterial infections as well as excessive loss of water (Fuchs and Raghavan, 2002). The most abundant cell type in epidermis is keratinocytes, which forms four layers: basal, spinous, granular and stratum corneum (Fuchs and Raghavan, 2002; Proksch et al., 2008). Atopic dermatitis (AD) is a chronic, inflammatory disease of the skin that starts at early childhood. AD patients are genetically predisposes to the disease, which has a prevalence of 10%-20% in children and 1%-3% in adults (Leung and Bieber, 2003; Tokura, 2010). Clinical features of AD include skin xerosis, pruritus and eczematoid skin lesions (Leung and Bieber, 2003). AD is characterized by both skin barrier deficiencies and immunological responses (Scharschmidt and Segre, 2008). In atopic dermatitis, a defective skin barrier is thought to permit the penetration of allergens and induces the interactions of the allergens with immune cells, promoting the subsequent release of pro-inflammatory cytokines and chemokines and elevation of IgE level (Cork et al., 2009). The molecular pathways involved in AD pathogenesis remain unclear. There have been many reports on the connections between atopic 88 dermatitis and Th2 inflammatory pathways (Leung and Bieber, 2003; Biedermann et al., 2004; B. Brandt and Sivaprasad, 2011). Pro-Th2 cytokines, such as IL-4, IL-13, IL-5, and IL10, are elevated in AD patients (B. Brandt and Sivaprasad, 2011). One possible candidate that initiates inflammatory responses in AD is thymic stromal lymphopoietin (TSLP), which is a member of hematopoietic cytokine family and highly expressed in skin-derived epithelial cells, fibroblasts, and dendritic cells (Comeau and Ziegler, 2010; Ziegler, 2010; Kashyap et al., 2011). TSLP signals through a heterodimeric receptor formed by TSLP receptor (TSLPR) as well as IL-7 receptor alpha (IL7Ra) (Comeau and Ziegler, 2010). TSLP is a key keratinocyte-derived cytokine, whose expression was greatly increased in the epidermis of a group of acute and chronic atopic dermatitis patients (Soumelis et al., 2002). Mice that are deficient of nuclear receptors (NRs) retinoid X receptor a (RXRa), vitamin D receptor (VDR) or Notch1 in epidermal keratinocytes, exhibit epidermal permeability barrier (EPB) defects, express elevated levels of TSLP in skin, and subsequently develop AD-like phenotypes (Li, 2005, 2006; Demehri et al., 2008; Ziegler, 2010). Chicken ovalbumin upstream promoter transcription factor (COUP-TF) interacting protein 2 (Ctip2), also known as Bcl11b, is a C2H2 zinc finger 89 protein that is crucial in the development of central nervous and immune system (Avram et al., 2000; Arlotta et al., 2005; Topark-Ngarm et al., 2006; Kastner et al., 2010). Ctip2 is also expressed in human and mouse adult epidermis (Golonzhka et al., 2007; Ganguli-Indra et al., 2009). Studies with mice harboring germline deletion and epidermal specific ablation of Ctip2 revealed its critical role(s) in epidermal proliferation and terminal differentiation, as well as barrier formation during mouse embryonic development in both cell autonomous and non-cell autonomous ways (Golonzhka et al., 2008). Ctip2 was shown to be a “master regulator” in skin homeostasis by interacting with the promoter regions of many genes involved in skin development such as eLox3, Gba2, Lass2 and Notch1 (Wang et al., 2012, submitted). Since Ctip2 null mice die at birth, the function(s) of keratinocytic Ctip2 in adult mice EPB maintenance as well as in epidermal homeostasis is unknown. Here we report a novel role of Ctip2 in regulating epidermal homeostasis and skin inflammation by selective Cre-recombinase mediated ablation of Ctip2 gene in epidermal keratinocytes of adult mice skin (Indra et al., 1999; Dassule et al., 2000). We show that ablation of Ctip2 in adult mice epidermal keratinocytes results in impaired EPB maintenance, increased epidermal hyperplasia and a 90 severe form of AD-like skin inflammation that becomes disseminated with progression of the disease, all of which are very similar to AD in humans. Our present data indicates that keratinocytic Ctip2 plays a crucial role in triggering skin inflammatory responses by regulating the expression of genes encoding Th2-type cytokines in adult mouse skin. The results presented herein establish an initiating role of epidermal TSLP in AD pathogenesis and demonstrate a key anti-inflammatory role of Ctip2, which strongly represses TSLP expression in wild-type skin. 3.3 Materials and Methods Mice The function of Ctip2 in adult mice skin homeostasis was characterized using Ctip2ep-/- mice that selectively lacked Ctip2 in epidermal keratinocytes after Cre-recombinase mediated deletion of Ctip2 gene in keratinocytes using K14-Cre deleter mice (Indra et al., 1999; Dassule et al., 2000; Golonzhka et al., 2008). Ctip2 floxed mice (Ctip2 L2/L2 , WT) were used as controls (Indra et al., 1999; Golonzhka et al., 2008). 8 to 10 mice from multiple litters were used in each group at each time point. Mice were maintained in a temperature/humidity-controlled facility 91 with a 12-hour light/dark cycle. OSU IACUC approval was obtained for animal experiments. Transepidermal water loss (TEWL) measurement Transepidermal water loss was measured using the calibrated Tewameter TM300 with Multi Probe Adapter (CK electronic GmbH, Köln, Germany) in accordance with manufacture operating instructions. Data was expressed in g/m2h, and represents the mean ± S.E.M. for 8 independent animals (8 measurements each animal) of each genotype. Statistical analysis was performed using Graphpad Prism software with student’s unpaired t-test. Histological analysis Skin biopsies were fixed in 4% paraformaldehyde overnight and embedded in paraffin blocks. 5mm paraffin sections were sectioned using a Leica RM2255 microtome (Bannockburn, IL). Hematoxylin and eosin staining was performed according to general protocols (Wang et al., 2011). Combined eosinophil/mast cell stain (C.E.M) was performed using a commercial kit according to the manufacturer’s protocol (American MasterTech, Lodi, CA). Briefly, slides were deparaffinized with xylene 92 and hydrated through alcohols. After washed in running tap water, slides were placed in astra blue staining solution for 30 minutes and rinsed in tap water. Slides were put in vital new red staining solution for another 30 minutes and rinsed with tap water. After that, slides were placed in modified Mayer’s hematoxylin for 15-30 seconds and rinsed again with tap water for 2 minutes. Finally, dehydration of slides was performed with 3 changes of absolute alcohol and xylene before mounting. Mast cells showed bright blue color while eosinophils were red. Nuclei were stained blue. Toluidine blue staining was performed as described (Li, 2005). Toluidine blue working solution was prepared by mixing 5ml 1% toluidine blue EtOH solution (Sigma-Aldrich, St. Louis, MO) with 45ml 1% sodium chloride (pH2.3). Slides were deparaffinized and hydrated in distilled water. Sections were stained with toluidine blue working solution (pH2.0-2.5) for 2-3 minutes before washed in 3 changes of distilled water. Slides were dehydrated through alcohols and xylenes and mounted with resinous mounting medium. Immunohistochemistry IHC staining of paraffin and frozen sections was described previously (Wang et al., 2011; Liang et al., 2012). In brief, paraffin sections were 93 deparaffinized and antigen retrieval was performed with pH6.0 citrate buffer and 95-100° C for 20 minutes and cooled down. For frozen sections, slides were fixed with cold acetone for 20 minutes and allowed for air dry. Slides were washed with 0.1% PBST and blocked with 10% normal goat serum (Vector Laboratories, Builingame, CA) for 30 minutes before incubated with primary antibodies overnight at 4°C. Fluorescently labeled secondary antibodies were applied to slides for an hour at room temperature. Nuclei were visualized with DAPI. After the final washes, slides were dehydrated and mounted. Images were captured at X20 magnification using a Carl Zeiss Axio Imager Z1 fluorescent microscope and AxioCam camera. Data were analyzed and quantified using Adobe Photoshop and ImageJ software. Multiple IHC fields were randomly chosen and 10-15 such fields per slide were counted independently in a double-blinded manner. All experiments in each category were repeated in triplicates. The concentrations of antibodies used were as follows: Ki67 (Abcam, Cambridge, MA, 1:500), K14 (Abcam, 1:1000), K10 (Abcam, 1:1000), Filaggrin (Abcam, 1:1000), Loricrin (Abcam, 1:500), F4/80 (Biolegend, San Diego, CA, 1:250), CD11b (Abcam, 1:100), CD45 (Abcam, 1:500), Ly6g (Abcam, 1:250), CD11c (BD Pharmingen, San Jose, CA, 1:100), RXRα (Santa Cruz Biotechnology, Santa Cruz, CA, 1:1000), Notch1 94 (Cell Signaling technology, Danvers, MA, 1:500), Cy3 goat anti rabbit (Jackson ImmunoResearch Laboratories Inc, West Grove, PA, 1:500), Cy3 goat anti rat (Jackson ImmunoResearch, 1:500), Cy2 goat anti rat (Jackson ImmunoResearch, 1:500), Cy3 goat anti mouse (Jackson ImmunoResearch, 1:1000). RT-qPCR RNA extraction and cDNA preparation were performed as described (Indra et al., 2005b). Real-time PCR was performed on an ABI 7500 Real-Time PCR system using SYBR Green methodology using specific sets of primers as indicated in Supplementary Table1 (Indra et al., 2005a, 2005b). Relative gene expression analysis of the RT-qPCR data was performed using HPRT as an internal control. All assays were performed in triplicates. ELISA ELISA kits for detecting mouse IL-4, IL-13, TNFα and TSLP were obtained from eBioscience, Inc. (San Diego, CA, USA). Mouse IgE ELISA kit was procured from Bethyl Laboratories, Inc. (Montgomery, TX, USA), while CCL17 immunoassay kit was purchased from R&D Systems, Inc. (Minneapolis, MN, USA). 95 1-month and 4-month-old mice (6 WT and 6 Ctip2ep-/- from each age group) were selected for studying secondary systemic response to Ctip2ep-/- genotype. Blood plasma was isolated from the mice following the method of Argmann and Auwerx (Argmann and Auwerx, 2006). ELISA was performed for estimation of the levels of IL-4, IL-13, IgE, CCL17, TNFα and TSLP using kits following the respective manufacturer’s instructions. All samples were assayed in triplicates. Chromatin immunoprecipitation (ChIP) ChIP assay was performed according to Hyter et al., 2010 with minor modifications (Hyter et al., 2010). Briefly, mouse epidermal keratinocytes from 1-day postnatal wildtype mice skin were isolated and fixed in 1% formaldehyde. Cell lysate from 3x106 keratinocytes (each ChIP) were obtained after incubation with hypotonic buffer (10mM Tris/HCl, pH7.5, 10mM NaCl, 3mM MgCl2 and 0.5% NP40). Nuclei were suspended in ChIP sonication buffer (50mM Hepes, pH7.9, 150mM NaCl, 1mM EDTA, 1% Triton X-100, 0.1% SDS, 0.1% sodium deoxycholate) with 0.2% SDS. Nuclei were sonicated and immunoprecipitated with 2mg anti-Ctip2 antibody (Abcam, Cambridge, MA) overnight at 4°C. Rat IgG was used as a control. 25ml preequilibrated protein G beads (Invitrogen, Carlsbad, CA) were added to 96 each ChIP and incubated for four hours at 4°C. After washing twice with ChIP sonication buffer, buffer A (50mM Hepes, 500mM NaCl, 1mM EDTA, 1% Triton X-100, 0.1% SDS and 0.1% sodium deoxycholate), buffer B (20mM Tris/HCl, pH8.0, 1mM EDTA, 250mM LiCl, 0.5% NP40 and 0.5% sodium deoxycholate) and once with TE buffer, DNA was uncrosslinked at 65°C overnight and purified by QIAquick PCR Purification Kit (Qiagen, Germantown, MD). All the buffers used in the study were added with PMSF and protease inhibitors before use. DNA was amplified using ABI-7500 Real-time PCR machine with specific primers indicated in Supplementary Table 1. Primers were designed from proximal (-200bp) and distal (-1kb) promoter regions of TSLP to study the possible interaction between Ctip2 and TSLP, and primers selected from 3’-UTR region was used as a control. Statistics Statistical significance of differences between groups was analyzed using GraphPad Prism software using student’s unpaired t-test. All statistical analyses were performed in a double-blinded manner. 97 3.4 Results 3.4.1 Selective ablation of Ctip2 in epidermal keratinocytes leads to altered epidermal proliferation, differentiation and spontaneous dermatitis in adult skin The function of Ctip2 in adult mice skin homeostasis was characterized using Ctip2ep-/- mice that selectively lacked Ctip2 in epidermal keratinocytes after Cre-recombinase mediated deletion of Ctip2 gene in keratinocytes using K14-Cre deleter mice (Indra et al., 1999; Dassule et al., 2000). At approximately 8-10 weeks after birth, 67% Ctip2ep-/- mice (n=18) developed dry and scaly skin and 17% mutant mice also developed spontaneous lesions on the dorsal skin at the same age, which were absent in wildtype (WT) mice (Fig 3.1 A). The severity of all of these abnormalities worsened with age (Figure 3.1 A). By 4 months of age, almost all the mutants (89%, n=18) developed spontaneous dermatitis that occurred predominantly on the dorsal skin, in the neck region, and on the face (Figure 3.1 A). Furthermore, progressive alopecia was evident in most of the 4 month-old Ctip2ep-/- mice (~85%) (Figure 3.1 A). We hypothesized that the skin lesions observed in the mutant mice could be due to impaired function of the skin barrier. To test this hypothesis, trans-epidermal water loss (TEWL) was measured in wildtype and Ctip2ep-/- mice. Interestingly, the mutant mice showed 98 increased TEWL as early as 2 weeks of age, and the values were ~10fold higher than wildtype mice at 4 months of age (Figure 3.1 B). Histological analysis of hematoxylin- and eosin- (H&E) stained skin sections revealed significant epidermal hyperplasia in Ctip2ep-/- mice as early as 2 weeks of age (Figures 3.1 C and D). In order to determine if increased epidermal hyperplasia could be due to altered keratinocyte proliferation, immunohistochemical (IHC) analyses for proliferation marker Ki67 and keratin 14 (K14) were performed on control and mutant skin (Figures 3.2 A and D). The percentage of Ki67-positive cells was significantly higher in Ctip2ep-/epidermis compared to the wild-type littermates at all time-points (Figure 3.2 A and D). Similarly, the expression level of K14 in the mutant epidermis was prominently increased in Ctip2ep-/- mice at 2 and 4 months of age (Figure 3.2 B). Expression levels of the early differentiation marker K10 were not significantly different between wildtype and Ctip2ep-/- mice (Figure 3.2 C). Expression of terminal differentiation markers, filaggrin and loricrin, were also upregulated in Ctip2ep-/- mice at later time points (Figure 3.3 A-B). These results indicate that selective, somatic ablation of Ctip2 in epidermal keratinocytes leads to increased epidermal proliferation and altered terminal differentiation during adulthood. Altogether, results suggest 99 that adult Ctip2ep-/- mice exhibited impaired barrier functions and developed spontaneous dermatitis, which worsened as the mice aged. 3.4.2. Increased infiltration of inflammatory cells in adult Ctip2ep-/mice skin Dermal infiltrates were notable in Ctip2ep-/- mice starting around 2 months of age, as revealed by histological analyses (Figure 3.1 C). Combined eosinophil and mast cell (C.E.M.) staining revealed increased eosinophils in the dermis of Ctip2ep-/- skin around 1 month of age, while mast cells were most numerous in the mutant dermis at 4 months of age (Figures 3.4 A and B). IHC staining for T lymphocytes revealed a two-fold increase in CD3+ T cells in the skin of 4 month-old Ctip2ep-/- mice compared to the wild-type skin (Figures 3.5 A-B). A similar increase in the number of CD4+ T cells was observed in 4 month-old mutant skin compared to the wildtype skin (Figure 3.5 C and D). The number of macrophages in the dermis was significantly higher in Ctip2ep-/- mice skin at 4 months of age (Figure 3.6 A-B), as were the number of infiltrating neutrophils and monocytes (Figure 3.6 C-F). In addition, CD45+ cell infiltration was observed in one month-old Ctip2ep-/mice skin, but this resolved by 4 months of age (Figure 3.6 G-H). 100 Taken together, these results demonstrate an increased inflammatory response in Ctip2ep-/- skin that progressively increased with time. 3.4.2 Preferential induction of Th2-type cytokines and chemokines in adult Ctip2ep-/- skin Upregulated expression of cytokines and chemokines play a crucial role in inflammatory skin diseases (Grewe et al., 1998; B. Brandt and Sivaprasad, 2011). Differential expression profiles of cytokines and/or chemokines orchestrate and determine the type and outcome of inflammatory responses in mouse and human skin. We next determined if expression of pro-inflammatory cytokines/chemokines were dysregulated in mutant skin. RT-qPCR analyses revealed a preferential induction of Th2-type cytokines, including TSLP, CCL17, IL13, IL4, IL6, and IL10 in mutant skin (Figures 3.7 A-E and 3.10 A). In contrast, neither expression of Th1-type (IL1α, IL2, IL12a/b; Figure 3.10 B-E) nor that of Th17-type (IL17a, IL18 and IL23; Figure 3.10 F-H) cytokines was unaltered in mutant skin. Time course analyses also revealed several distinct induction patterns of cytokines and chemokines in the mutant skin. TSLP, a master initiator of Th2 inflammatory responses, was induced ~30-fold at postnatal day 1 (P1), and we observed a ~200-fold induction at P7 101 compared to control skin (Liu, 2006). Expression of TSLP remained significantly high at all later time points (~1000-fold induction between 1 month to 4 months after birth; Figure 3.7 A). Interestingly, the high level of TSLP expression was observed predominantly in the mutant epidermis, whereas TSLP transcripts were barely detectable in the dermal layer of the mutant skin (Figure 3.7 F). These data suggest that TSLP may be an initial triggering cytokine that is induced in the absence of Ctip2 in the epidermis. Chemokine CCL17 is one of known downstream genes whose expression can be stimulated by TSLP (Ziegler and Artis, 2010). CCL17 and IL13 transcripts were significantly induced in the mutant skin at around 1 week and 2 weeks after birth, respectively. Induction of these two genes was transient, peaking at 1 month, followed by a decrease of expression during later time points (2 - 4 months; Figure 3.7 B and C). In contrast to these early and transient inductions, we observed a strong, late induction of cytokines IL4 and IL6 and chemokines CCL3 and CXCL2 in the mutant skin (Figure 3.7 D-E, 3.8 L-M). We also observed a late onset of induction of Th1-type cytokines, including TNFα and IFNγ, at 4 months of age (Figure 3.8 N-O). These results suggest that deletion of Ctip2 in the epidermis leads to an increased 102 inflammatory response, which is characterized by a Th2-type cytokine/chemokine profile. 3.4.3 TSLP is a direct target of Ctip2 in epidermal keratinocytes Recent studies have revealed that ablation of RXRα and RXRβ in adult murine keratinocytes leads to an AD-like phenotype triggered by induction of TSLP (Li, 2005).. Similarly, TSLP is also elevated in Notch signaling-deficient keratinocytes (deficient in either Notch1/2 or recombining binding protein suppressor of hairless (RBP-j; Refs (Li, 2005; Demehri et al., 2008; Dumortier et al., 2010)). Thus, it is possible that Ctip2 regulates TSLP expression via the nuclear receptor or Notch signaling pathways in epidermis. To test that, we determined the expression of key players in both the Notch and RXR signaling pathways, including Notch1, Notch2, RBP-j, RXRα and RXRβ. Expression of the corresponding transcripts was not affected by loss of Ctip2 in the epidermis at both one and four months after birth (Figure 3.12 A-D, data not shown). Similarly, epidermal protein levels of Notch1 and RXRα were comparable between wildtype and mutants at all timepoints, suggesting an alternative mechanism of Ctip2-mediated regulation of TSLP expression in the adult murine skin (Figure 3.12 EF). 103 In order to determine if Ctip2 directly regulates TSLP expression in murine keratinocytes, we conducted chromatin immunoprecipitation (ChIP) assays on neonatal mouse epidermal extracts. Primers were designed from the proximal (-200bp) and distal (-1kb) promoter region relative to the transcriptional start site of TSLP to determine the possible interaction of Ctip2 with TSLP. We observed that Ctip2 interacted with the distal but not the proximal region of TSLP promoter, suggesting Ctip2 may directly repress TSLP transcription in mouse keratinocytes (Figure 3.7 G). Hence, ablation of Ctip2 in epidermal keratinocytes could lead to upregulation of TSLP expression due a derepression mechanism. 3.4.4 Systemic inflammatory responses in older Ctip2ep-/- mice We hypothesized that chronic skin inflammation developed in Ctip2ep-/mice may trigger systemic immune responses in the mutant skin. To test that we determined blood plasma concentrations of IgE as well as various Th2-type cytokines/chemokines (IL-4, IL-13, CCL17 and TSLP) and Th1-type cytokine TNF-α from 1-month and 4-months-old mice by ELISA (Figure 3.9 A-F). Plasma level for all of the tested cytokines, were observed to be higher in both 1-month and 4-month old mutant mice (Figure 3.9 A-E), including IL4 and the related IL-13 (Figure 3.9 A, 104 B), CCL17 (Figure 3.9 C), TSLP (Figure 3.9 D), and TNFα (Figure 3.9 E). While circulating IgE levels were elevated in both wild-type and mutant mice at four months of age, IgE levels in mutant mice were 2.5 times greater than those in wildtype mice (Figure 3.9 F). Ctip2ep-/- mice exhibited inguinal lymphadenopathy smaller convoluted thymus at four months of age (Figure 3.10 A, data not shown). The mutant mice displayed progressive splenomegaly starting from one month of age, with a 2-fold increase in spleen weight at the age of four months (Figure 3.10 A-B). Histological studies of the spleens revealed an increased cell infiltration in the 4-month-old mutant spleen in comparison with wildtype (Figure 3.11 A, D, G, J). 4 month-old Ctip2ep-/mice exhibited increased eosinophil infiltration in the spleen and lymph node compared with wildtype (Figure 3.11, B, E, H, K). Very few mast cells were observed in both wildtype and mutant mice spleens and the number of mast cells was larger in the lymph node, indistinguishable between 4 month-old wildtype and mutant mice (Figure 3.11, C, F, I, L). These results clearly indicate that Ctip2ep-/- mice display a systemic immune response that may be driven by primarily TSLP expression from keratinocytes. 105 3.5 Discussion Epidermal keratinocytes have long been suspected to play a crucial role in initiating and directing the immune response in inflammatory skin diseases, such as AD. In the present report we have shown that skin of adult Ctip2ep-/- mice, with an epidermal-specific deletion of the gene encoding Ctip2, display histological and cellular features that are typical of human AD. First, Ctip2ep-/- mice develop skin lesions, alopecia and pruritus, which are major clinical features in patients afflicted with AD (Leung and Bieber, 2003; Dubrac et al., 2010; Boguniewicz and Leung, 2011). Second, Ctip2ep-/- mice exhibit epidermal hyperplasia that is characterized by increased epidermal proliferation and differentiation, similar to what has been observed in AD patients (Proksch et al., 2008). Third, the skin of Ctip2ep-/- mice is characterized by a dramatic upregulation of Th2-type cytokines and chemokines, including TSLP, CCL17, IL13 and IL4. TSLP, all of which were induced early in Ctip2ep-/epidermis, and are crucially involved in the initiation of pathogenesis in human AD (Tokura, 2010; B. Brandt and Sivaprasad, 2011; Novak and Leung, 2011). Finally, Ctip2ep-/- adult mice exhibit Th2-like systemic immune syndrome that is similar to that found in most AD patients (Spergel, 2003). Based on the phenotypic spectrum of Ctip2ep-/- adult 106 mice, it appears that this line may serve as a useful model for the study of human AD. Selective ablation of Ctip2 in the developing epidermis reduced epidermal proliferation and impaired terminal differentiation of keratinocytes (Golonzhka et al., 2008). Isolated Ctip2 null keratinocytes also exhibit severe proliferation and differentiation defects in culture (Zhang et al., submitted). To our surprise, in the postnatal stages, the epidermis of Ctip2ep-/- adult mice exhibited increased proliferation and differentiation compared with wild-type epidermis (Figure 3.1 C, 3.2, 3.3). A hyperplastic epidermis and a significantly higher percentage of Ki67-positive cells were observed in the mutants as early as 1 week after birth. Elevated proliferation and differentiation could be secondary to impaired epidermal barrier functions caused by Ctip2 ablation. Previous studies have established that hyperproliferation and acanthosis (thickened epidermis) are compensatory responses to impaired epidermal barrier (Proksch et al., 1991) A number of candidate genes have been identified in association with AD, however, may of these linkages have been called into question due to insufficiently powered studies and/or heterogeneity of the disease (Dubrac et al., 2010; Boguniewicz and Leung, 2011). In independent 107 studies, several genes such as Filaggrin (FLG), trans-acting T-cellspecific transcription factor (GATA3) and IL4 has been reported to be involved in AD pathogenesis (Barnes, 2010). FLG deficiency alone decreases stratum corneum hydration and leads to increased TEWL, and has been correlated with AD in the highest number of studies (Palmer et al., 2006; Barnes, 2010). It is worth mentioning that skin barrier deficiency has also been reported in AD patients without FLG mutation(s) or loss of expression (Jakasa et al., 2010) suggesting existence of different subtypes of AD in humans. We have shown previously that Ctip2 deficiency is associated with impaired terminal differentiation and decreased FLG expression during development (Golonzhka et al., 2008). Indeed, we have observed a ~50% reduction of FLG expression in the epidermis of Ctip2ep-/- skin compared to that of wildtype littermates around 1 week of age (Figure 3.13). However, no significant difference in level of FLG expression was observed between the wildtype and mutant epidermis at later timpoints from 2 weeks to 4 months of age possibly due to a compensatory upregulation of other related factors such as Ctip1 (Figure 3.3A, 3.13). The lack of loss-ofFLG expression observed in Ctip2ep-/- mice at later stages, indicates possible involvement of other factors to modulate barrier integrity. 108 TSLP, a keratinocyte-derived triad of cytokines, has been studied intensively recently in the pathogenesis of allergic inflammation and subsequent development of AD and other immune disorder (Soumelis et al., 2002; Yoo, 2005; Liu, 2006). It is recognized as the master switch for AD and other atopic diseases, such as asthma, as it plays a critical role to induce the Th2-type allergic inflammation in AD pathogenesis (Liu, 2006). Keratinocyte-derived TSLP participates in the inflammatory cascade by activation of other immune cells, such as dendritic cells and mast cells, and subsequent stimulation of release of Th2-related cytokines/chemokines from these immune cells, eventually initiate inflammatory a Th2 lymphocyte response (Soumelis et al., 2002; Liu, 2009; Comeau and Ziegler, 2010). In the present study, we have observed that TSLP was the earliest known cytokine induced in Ctip2ep/- mice and this induction was maintained in the mutants at all postnatal stages of development. Expression of CCL17, a downstream target of TSLP, was also significantly induced in the mutant mice at early timepoints. The induction of TSLP was mainly restricted to the epidermis, which correlates well with epithelial cells of the keratinocyte lineage as major producers of TSLP in AD patients (Soumelis et al., 2002). Hence, induction of TSLP following Ctip2 ablation in 109 keratinocytes could be a trigger factor that initiates and directs the Th2type inflammatory cascade observed in Ctip2ep-/- skin. Transgenic mice over-expressing TSLP in keratinocytes develop ADlike symptoms, indicating that TSLP expression is sufficient to initiate AD-like inflammatory responses (Yoo, 2005). Therefore, it remains critical to address upstream triggers of TSLP expression and how TSLP is regulated before and during AD pathogenesis. Transcriptional regulation of TSLP by transcription factors (NF-κB, AP-1, AP-2α) and nuclear receptors (RXR, RAR, VDR) were reported previously (Li, 2005, 2006; Lee and Ziegler, 2007; Lee et al., 2008; Takai, 2012). NF-κB and AP-1 was shown to directly regulate TSLP expression by binding to 3.8kb of transcription initiation site in mouse and human lung epithelium cells (Lee et al., 2008). Previous studies have identified presence of a RARE (DR2) around ~ 1kb of the TSLP promoter region that may bind to RXRα(β)-RAR heterodimers (Li, 2006). Since we have observed Ctip2 recruitment around that region on the mTSLP promoter, it is possible that NRs and Ctip2 could be a part of the same transcriptional complex to regulate TSLP gene expression in mouse keratinocytes. However, additional co-recruitment of Ctip2 along with NRs in further distal region of the mTSLP gene promoter cannot be ruled out. Here, we have identified a novel transcriptional regulatory mechanism(s) of 110 negative regulation of TSLP expression by Ctip2 in mouse epidermis. Ctip2 appears to interact directly with and repress expression of the TSLP promoter. We have demonstrated a predominant upregulation of Th2 cytokines in the Ctip2ep-/- skin at all timepoints, but a late surge of Th1 cytokines, including IFNγ and TNF α , was also observed in the mutant skin at 4 months (Figure 3.8 N-O). Indeed, it has been demonstrated that Th2and Th1 type cytokines both contribute to different stages of the development of skin lesions in AD patients (Grewe et al., 1998). Th2cell subsets are specifically activated during the initiation phase, followed by the Th1-cell subset to maintain the persistence of the chronic inflammatory response. Our work confirms that Th2- and Th1type immune responses are not mutually exclusive, especially at the chronic phage of the inflammatory disease. The interaction between different Th-cell subsets at the chronic stage may be a fruitful area for future investigation of AD pathogenesis. Systemic immune response related to elevated expression of IL-4, IL13, CCL17 and IgE, observed in Ctip2ep-/- mice is evidently Th2 celldependent and humoral in nature. IL-4, for example, induces differentiation of CD4+ T-cells into Th2 cells, and also inhibits production of Th1 cells (Rang, 2003). Th2 cells secrete IL-13, which 111 together with IL-4, initiates a B lymphocyte-directed, humoral response (Rang, 2003). IL-4 also upregulates MHC class II expression and induces B-cell class switching to IgE [41, 42]. Furthermore, TSLP stimulation of naïve CD4+ T-cells induces a specialized Th2 polarization, which results in production of downstream Th2-associated pro-inflammatory cytokines, such as IL-13, which carry out humoral immune processes (Ziegler and Artis, 2010). Simultaneously, CCL17 is known to attract Th2 cells, thereby favoring humoral responses (Nakayama et al., 2004). In summary, inflammation due to selective Ctip2 ablation in mice epidermis is not restricted to the site of deletion; rather Ctip2ep-/- mice show a chronic secondary Th2-dependent humoral systemic inflammatory response. In summary, our study reveals a novel role(s) of keratinocytic Ctip2 in epidermal barrier maintenance and homeostasis, and inflammation in adult mice. Ctip2 clearly modulates AD-like responses within the skin, as well as systemic inflammatory responses. TSLP was strongly induced in Ctip2ep-/- epidermis and likely causes increased dendritic cells infiltration into lymph nodes, in which these cells interact with CD4+ T cells to produce Th-2 inflammatory cytokines. Subsequently, other immune cells such as macrophages, leukocytes, mast cells and eosinophils could be attracted and recruited to the site of skin 112 inflammation, which become disseminated, leading to elevated circulating levels of inflammatory mediators. This study identifies a new mediator, Ctip2, that may be implicated in the etiology of AD. However, several issues need to be resolved in the future. For example, it is presently unclear if a defective skin barrier precedes inflammation and enhanced cellular proliferation in the early stages of AD development. Second, the contributory role of TSLP in AD and the mechanistic basis of regulation of TSLP expression in keratinocytes have not been identified. Finally, the role of Ctip1 in AD development in presence or absence of Ctip2 could shed further lights in AD pathogenesis and identify new avenues for therapeutic intervention. Our study highlights a central role of epidermal keratinocytes in initiating and shaping the immune response during the pathogenesis of AD-like skin diseases in mice. D epidermal thickness (µm) 113 WT Ctip2ep-/- 40 *** *** *** *** 30 20 10 0 2w 1m 2m 4m 114 Figure 3.1 Ctip2ep-/- mice develop a chronic skin lesions and epidermal hyperplasia. (A) Gross morphology of 4 months old wildtype (WT) and Ctip2ep-/mice. The yellow arrowheads indicate lesion and alopecia of Ctip2ep-/mice in the back, face and neck, to be compared with the normal appearance in a wildtype mouse. (B) Measurement of trans-epidermal water loss (TEWL) from dorsal skin of wildtype and Ctip2ep-/- mice at different time points. Statistical analyses were performed by student’s unpaired t-test using GraphPad Prism software; ** P< 0.01, *** P<0.001. (C) Hemotoxylin & Eosin stained 5µm thick paraffin sections from dorsal skin of WT and Ctip2ep-/- mice at 1 week (1W), 2 weeks (2w), 1month (1m), 2 months (2m) and 4 months (4m). Scale bar: 100 µm. Epidermis (E) and dermis (D) are indicated. (D) Measurement of epidermal thickness of WT and Ctip2ep-/- mice skin. Statistical analyses were performed by student’s unpaired t-test using GraphPad Prism software; * P<0.05, ** P< 0.005, *** P<0.0001. 115 116 Figure 3.2 Ctip2ep-/proliferation. mice develop enhanced epidermal Immunohistochemical staining of dorsal skin biopsies from WT and Ctip2ep-/- mice was performed with antibodies directed against (A) Ki67, (B) K14 and (C) K10 (all in red). All sections were counterstained with DAPI (blue). Scale bar: 100 µm. Epidermis (E) and dermis (D) are indicated. (D) Epidermal percent Ki67 positive cells in dorsal skin sections of WT and mutant mice. Statistical analyses were performed by student’s unpaired t-test using GraphPad Prism software; * P<0.05, ** P< 0.005. 117 Figure 3.3 Ablation of Ctip2 leads to increased epidermal terminal differentiation. Immunohistochemical staining of dorsal skin biopsies from WT and Ctip2ep-/- mice was performed with antibodies directed against (A) filaggrin and (B) loricrin (all in red). All sections were counterstained with DAPI (blue). Scale bar: 100 µm. Epidermis (E) and dermis (D) are indicated. 118 Figure 3.4 Enhanced eosinophil and mast cell infiltrates in dorsal skin of Ctip2ep-/- adult mice. (A) Combined eosinophil and mast cell (C.E.M) staining for eosinophils (pink) and mast cells (blue). Black arrows point to eosinophils. Scale bar: 50 µm. (B) Toluidine blue stained dorsal paraffin skin sections of WT and Ctip2ep-/- mice. Mast cells stain intensive blue color. Scale bar: 100 µm. 119 Figure 3.5 Characterization of inflammatory cell infiltrates in dorsal skin of WT and in Ctip2ep-/- adult mice. (A) Immunohistochemical staining of dorsal skin biopsies from WT and Ctip2ep-/- mice were performed with specific antibodies against CD3 (red). Yellow arrowhead indicates dermal infiltrates of CD3+ cells. Scale bar: 100 µm. (B) Percent CD3+ T cells at 2m and 4m. (C) Immunostaining of CD4+ T cells (red) in WT and mutant mice. Scale bar: 100 µm. (D) Percent CD4+ T cells at 2m and 4m. All sections were counterstained with DAPI (blue). Statistical analyses were performed by student’s unpaired t-test using GraphPad Prism software; ** P< 0.005. 120 121 Figure 3.6 Increased inflammatory cell infiltrate in Ctip2ep-/- adult mice. (A) Immunohistochemical staining of dorsal skin biopsies from WT and Ctip2ep-/- mice were performed with specific antibodies against F4/80 (green) to detect macrophage/dendritic cells. (B) Percent F4/80 positive cells at 2m and 4m. (C) Immunostaining of neutrophils (green) using anti-Ly6g antibody in WT and mutant mice skin. (D) Percent Ly6g positive cells at 2m and 4m. (E) Immunostaining of monocytes/macrophages (green) with anti-CD11b antibody in WT and mutant mice skin. (F) Percent CD11b positive cells at 2m and 4m. (G) Immunohistochemical staining of dorsal skin biopsies with antibody against CD45 (green). (H) Percent CD45 positive cells at 1m and 2m. All sections were counterstained with DAPI (blue). Scale bar (A, C, E and G): 100 µm. Statistical analyses were performed by student’s unpaired t-test using GraphPad Prism software; ** P< 0.005. 122 123 Figure 3.7 Th2-dependent cytokine and chemokine expression levels in WT and Ctip2ep-/- skin. (A-F) Quantitative RT-PCR (RT-qPCR) analyses of cytokines and chemokines in the dorsal skin of 1day, 1 week, 2 weeks, 1month, 2 months and 4 months old WT and mutant mice using specific primers as indicated in Supplementary Table1. Relative mRNA expression levels of (A) TSLP, (B) CCL17, (C) IL13, (D) IL4, and (E) IL6 in Ctip2ep-/skin was compared to wildtype (WT) skin (set as 1.0). (F) RT-qPCR analyses of TSLP mRNA levels in separated epidermis and dermis from tail skin. All values represent relative transcript level after normalization with HPRT transcripts. (G) Chromatin immunoprecipitation (ChIP) assay was performed on freshly isolated neonatal mouse skin keratinocytes using anti-Ctip2 antibody and results were analyzed by qPCR using primers indicated in table S1. Rat IgG was used as a control. Ctip2 was recruited to the distal promoter regions of TSLP. Statistical analyses were performed by student’s unpaired t-test using GraphPad Prism software; * P<0.05, ** P<0.01, *** P<0.001. 2 4m 0.0 1m 2 * 1 0 1m 4m 1.0 0.5 0.0 1m 4m CCL20 WT Ctip2ep-/- 500 *** 400 300 200 5 4 3 2 1 0 * 1m 4m CXCL2 Relative Expression Level ** 1.0 0.5 0.0 1m 1m 4m 4m WT Ctip2ep-/- 2.0 1.5 1.0 0.5 0.0 1m 4m CCL22 5 WT Ctip2ep-/- 4 * 3 2 1 0 1m 4m IFN 1.5 * 1.0 0.5 0.0 1.0 0.5 0.0 1m 4m 1.5 1.0 0.5 0.0 1m 4m CXCL10 O WT Ctip2ep-/- 5 4 * 3 2 1 0 1m 4m TNF Supplementary figure 3 4m WT Ctip2ep-/- 2.5 2.0 1.5 1.0 0.5 0.0 1m 4m IL23 L WT Ctip2ep-/- 1m H IL18 WT Ctip2ep-/- 2.0 WT Ctip2ep-/- IL12a 1.5 K N Relative Expression Level Relative Expression Level 1.5 0.0 IL17a WT Ctip2ep-/- 2.0 0.5 G * 1.5 J M Relative Expression Level WT Ctip2ep-/- 2.0 IL12b I 1.0 2.0 IL2 Relative Expression Level 3 Relative Expression Level Relative Expression Level F WT Ctip2ep-/- 4 * IL1 IL10 E 4m Relative Expression Level 1m Relative Expression Level 0 0.5 D Relative Expression Level ** ** 1.0 WT Ctip2ep-/- 1.5 Relative Expression Level 4 C Relative Expression Level ** WT Ctip2ep-/- 1.5 Relative Expression Level B WT Ctip2ep-/- 6 Relative Expression Level A Relative Expression Level 124 100 90 80 70 60 50 5 4 3 2 1 0 WT Ctip2ep-/- *** 1m 4m CCL3 125 Figure 3.8 Relative expression levels of cytokines and chemokines in WT and Ctip2ep-/- skin. The expression level of (A) IL10, (B) IL1α, (C) IL2, (D) IL12a, (E) IL12b, (F) IL17a, (G) IL18, (H) IL23, (I) CCL20, (J) CCL22, (K) CXCL10, (L) CCL3, (M) CXCL2, (N) IFNγ and (O) TNFα was studied with RT-qPCR in 1-month and 4-month-old wildtype and Ctip2ep-/- dorsal skin. Values represent relative transcript level after normalization with HPRT transcripts. Statistical analyses were performed by student’s unpaired ttest using GraphPad Prism software; * P<0.05, ** P<0.01, *** P<0.001. 126 Figure 3.9 Systemic immunological abnormalities in Ctip2ep-/- adult mice. Serum concentration of (A) IL4, (B) IL13, (C) CCL17, (D) TSLP, (E) TNFα and (F) IgE was measured by ELISA. Statistical analyses were performed by student’s unpaired t-test using GraphPad Prism software; * P<0.05, ** P< 0.01, *** P<0.005 127 Figure 3.10 Ctip2ep-/- adult mice exhibited systemic immune responses. (A) Gross morphological comparison of spleen, lymph node (LN) and liver in 4-months-old WT and Ctip2ep-/- mice. (B) Spleen weight of WT and Ctip2ep-/- mice at different months of age is indicated. Statistical analyses were performed by student’s unpaired t-test using GraphPad Prism software; * P<0.05, ** P< 0.01, *** P<0.005 128 Figure 3.11 Immunological abnormalities of spleen and lymph node in Ctip2ep-/- adult mice. (A, D) Hemotoxylin & Eosin stained 5µm thick paraffin sections from spleen of WT and Ctip2ep-/- mice at 4m. (B, C, E, F) C.E.M staining for eosinophils (pink) and mast cells (blue) in 4 month-old mice spleen sections. (G, J) Hemotoxylin & Eosin stained 5µm thick paraffin sections of WT and Ctip2ep-/- mice lymph node at 4m. (H, I, K, L) C.E.M staining for eosinophils (pink) and mast cells (blue) in 4 month-old mice lymph node. Black arrows point to eosinophils. Scale bar: 50 µm 129 Figure 3.12 Relative expression levels of RXRa and genes involved in Notch signaling pathway. The expression level of (A) Notch1, (B) Notch2, (C) Rbp-j and (D) RXRα was studied with RT-qPCR in 1-month and 4-month-old wildtype and Ctip2ep-/- dorsal skin. Values represent relative transcript level after normalization with GAPDH transcripts. Statistical analyses were performed by student’s unpaired t-test using GraphPad Prism software. (E) Immunohistochemical staining of dorsal skin biopsies with antibody against Notch1 (red) and (F) RXRα (red). Scale bar: 100 µm. All sections (in E & F) were counterstained with DAPI (blue). 130 Figure 3.13 Relative expression levels of FLG in 1w and 2w mice epidermis and dermis. The expression of FLG in (A) 1-week-old and (B) 2-week-old wildtype and Ctip2ep-/- skin by RT-qPCR. Values represent relative transcript level after normalization with HPRT transcripts. Statistical analyses were performed by student’s unpaired t-test using GraphPad Prism software; * P<0.05, ** P<0.01, *** P<0.001. 131 Table 3.1 List of primers used for RT-qPCR and ChIP assays Gene Sense Antisense CCL3 CTCCCAGCCAGGTGTCATTTT CTTGGACCCAGGTCTCTTTGG CCL11 TCCACAGCGCTTCTATTCCT CTATGGCTTTCAGGGTGCAT CCL17 TACCATGAGGTCACTTCAGATGC GCACTCTCGGCCTACATTGG CCL20 TCACCTCTGCAGCCAGGCAGA TCTTAGGCTGAGGAGGTTCACAGC CCL22 CCGTCACCCTCTGCCATCACG GACCTGCCTGGGATCGGCAC CXCL10 CCACGTGTTGAGATCATTGCCACG ATCCATCGCAGCACCGGGGT CXCL2 AGAAGTCATAGCCACTCTCAAGGGC AGCAGCCCAGGCTCCTCCTTT HPRT GTTAAGCAGTACAGCCCCAAA AGGGCATATCCAACAACAAACTT IL1a TCACCTTCAAGGAGAGCCG ATCTGGGTTGGATGGTCTCTT IL2 TGAGCAGGATGGAGAATTACAGG GTCCAAGTTCATCTTCTAGGCAC IL4 GAGCCATATCCACGGATGCGAC ATGCGAAGCACCTTGGAAGCCC IL5 CAGCTGTCCGCTCACCGAGCT TTTCCACAGTACCCCCACGGACAG IL6 ACAAAGCCAGAGTCCTTCAGAGAGA AGCCACTCCTTCTGTGACTCCAG IL10 GGCGCTGTCATCGATTTCTCCCC GGCCTTGTAGACACCTTGGTCTTGG IL12a CTGTGCCTTGGTAGCATCTATG GCAGAGTCTCGCCATTATGATTC IL12b AGTGTGAAGCACCAAATTACTCC CCCGAGAGTCAGGGGAACT IL13 TGCTTGCCTTGGTGGTCTCGC GCGGCCAGGTCCACACTCCA IL17a ACGCGCAAACATGAGTCCAGGG TGAGGGATGATCGCTGCTGCCT IL18 GTGAACCCCAGACCAGACTG CCTGGAACACGTTTCTGAAAGA IL23a GAACGCACATGCACCAGCGG TGCAAGCAGAACTGGCTGTTGTCC TNFa ACTTCGGGGTGATCGGTCCCC TGGTTTGCTACGACGTGGGCTAC TSLP ACGGATGGGGCTAACTTACAA AGTCCTCGATTTGCTCGAACT RXRa GATATCAAGCCGCCACTAGG TGTTGTCTCGGCAGGTGTAG RXRb CACCTCTTACCCCTTCAGCA GAGCGACACTGTGGAGTTGA Notch1 TCAATGCCGTGGATGACCTA CCTTGTTGGCTCCGTTCTTC Notch2 GAGAAAAACCGCTGTCAGAATGG GGTGGAGTATTGGCAGTCCTC Rbpj AGTTGCACAGAAGTCTTACGG CCTATTCCAATAAACGCACAGGG TSLP-ChIP proximal TSLP-ChIP distal TSLP-ChIP 3’-UTR ATCTTAACCCAACCCACCAT CTAGGGGAGGAACAGCTTCT CCGTAGGCGTTTAGGTGTTA CAAAGACTGTGCTCGGGTAT TATTGCAAATCCAGCTGTCA TTTCCAAAAGTGCTCACAAAA 132 3.6 References Argmann, C.A., and Auwerx, J. (2006). Collection of blood and plasma from the mouse. Curr Protoc Mol Biol Chapter 29, Unit 29A.3. Arlotta, P., Molyneaux, B.J., Chen, J., Inoue, J., Kominami, R., and Macklis, J.D. (2005). Neuronal Subtype-Specific Genes that Control Corticospinal Motor Neuron Development In Vivo. Neuron 45, 207–221. Avram, D., Fields, A., Pretty On Top, K., Nevrivy, D.J., Ishmael, J.E., and Leid, M. (2000). Isolation of a novel family of C(2)H(2) zinc finger proteins implicated in transcriptional repression mediated by chicken ovalbumin upstream promoter transcription factor (COUP-TF) orphan nuclear receptors. J Biol Chem 275, 10315–10322. B. Brandt, E., and Sivaprasad, U. (2011). Th2 Cytokines and Atopic Dermatitis. Journal of Clinical & Cellular Immunology 02,. Barnes, K.C. (2010). An update on the genetics of atopic dermatitis: Scratching the surface in 2009. Journal of Allergy and Clinical Immunology 125, 16–29.e11. Biedermann, T., Röcken, M., and Carballido, J.M. (2004). TH1 and TH2 lymphocyte development and regulation of TH cell-mediated immune responses of the skin. J. Investig. Dermatol. Symp. Proc. 9, 5–14. Boguniewicz, M., and Leung, D.Y.M. (2011). Atopic dermatitis: a disease of altered skin barrier and immune dysregulation. Immunol. Rev. 242, 233–246. Comeau, M.R., and Ziegler, S.F. (2010). The influence of TSLP on the allergic response. Mucosal Immunol 3, 138–147. Cork, M.J., Danby, S.G., Vasilopoulos, Y., Hadgraft, J., Lane, M.E., Moustafa, M., Guy, R.H., MacGowan, A.L., Tazi-Ahnini, R., and Ward, S.J. (2009). Epidermal Barrier Dysfunction in Atopic Dermatitis. Journal of Investigative Dermatology 129, 1892–1908. Dassule, H.R., Lewis, P., Bei, M., Maas, R., and McMahon, A.P. (2000). Sonic hedgehog regulates growth and morphogenesis of the tooth. Development 127, 4775–4785. Demehri, S., Liu, Z., Lee, J., Lin, M.-H., Crosby, S.D., Roberts, C.J., Grigsby, P.W., Miner, J.H., Farr, A.G., and Kopan, R. (2008). Notch- 133 Deficient Skin Induces a Lethal Systemic B-Lymphoproliferative Disorder by Secreting TSLP, a Sentinel for Epidermal Integrity. PLoS Biology 6, e123. Dubrac, S., Schmuth, M., and Ebner, S. (2010). Atopic dermatitis: the role of Langerhans cells in disease pathogenesis. Immunology and Cell Biology 88, 400–409. Dumortier, A., Durham, A.-D., Di Piazza, M., Vauclair, S., Koch, U., Ferrand, G., Ferrero, I., Demehri, S., Song, L.L., Farr, A.G., et al. (2010). Atopic Dermatitis-Like Disease and Associated Lethal Myeloproliferative Disorder Arise from Loss of Notch Signaling in the Murine Skin. PLoS ONE 5, e9258. Fuchs, E., and Raghavan, S. (2002). Getting under the skin of epidermal morphogenesis. Nat Rev Genet 3, 199–209. Ganguli-Indra, G., Liang, X., Hyter, S., Leid, M., Hanifin, J., and Indra, A.K. (2009). Expression of COUP-TF-interacting protein 2 (CTIP2) in human atopic dermatitis and allergic contact dermatitis skin. Experimental Dermatology 18, 994–996. Golonzhka, O., Leid, M., Indra, G., and Indra, A.K. (2007). Expression of COUP-TF-interacting protein 2 (CTIP2) in mouse skin during development and in adulthood. Gene Expr Patterns 7, 754–760. Golonzhka, O., Liang, X., Messaddeq, N., Bornert, J.-M., Campbell, A.L., Metzger, D., Chambon, P., Ganguli-Indra, G., Leid, M., and Indra, A.K. (2008). Dual Role of COUP-TF-Interacting Protein 2 in Epidermal Homeostasis and Permeability Barrier Formation. J Investig Dermatol 129, 1459–1470. Grewe, M., Bruijnzeel-Koomen, C.A., Schöpf, E., Thepen, T., Langeveld-Wildschut, A.G., Ruzicka, T., and Krutmann, J. (1998). A role for Th1 and Th2 cells in the immunopathogenesis of atopic dermatitis. Immunol. Today 19, 359–361. Hyter, S., Bajaj, G., Liang, X., Barbacid, M., Ganguli-Indra, G., and Indra, A.K. (2010). Loss of nuclear receptor RXRα in epidermal keratinocytes promotes the formation of Cdk4-activated invasive melanomas. Pigment Cell Melanoma Res 23, 635–648. Indra, A.K., Dupe, V., Bornert, J.M., Messaddeq, N., Yaniv, M., Mark, M., Chambon, P., and Metzger, D. (2005a). Temporally controlled 134 targeted somatic mutagenesis in embryonic surface ectoderm and fetal epidermal keratinocytes unveils two distinct developmental functions of BRG1 in limb morphogenesis and skin barrier formation. Development 132, 4533–4544. Indra, A.K., Mohan, W.S., Frontini, M., Scheer, E., Messaddeq, N., Metzger, D., and Tora, L. (2005b). TAF10 is required for the establishment of skin barrier function in foetal, but not in adult mouse epidermis. Dev Biol 285, 28–37. Indra, A.K., Warot, X., Brocard, J., Bornert, J.M., Xiao, J.H., Chambon, P., and Metzger, D. (1999). Temporally-controlled site-specific mutagenesis in the basal layer of the epidermis: comparison of the recombinase activity of the tamoxifen-inducible Cre-ER(T) and CreER(T2) recombinases. Nucleic Acids Res. 27, 4324–4327. Jakasa, I., Koster, E.S., Calkoen, F., McLean, W.H.I., Campbell, L.E., Bos, J.D., Verberk, M.M., and Kezic, S. (2010). Skin Barrier Function in Healthy Subjects and Patients with Atopic Dermatitis in Relation to Filaggrin Loss-of-Function Mutations. Journal of Investigative Dermatology 131, 540–542. Kashyap, M., Rochman, Y., Spolski, R., Samsel, L., and Leonard, W.J. (2011). Thymic stromal lymphopoietin is produced by dendritic cells. J. Immunol. 187, 1207–1211. Kastner, P., Chan, S., Vogel, W.K., Zhang, L.-J., Topark-Ngarm, A., Golonzhka, O., Jost, B., Le Gras, S., Gross, M.K., and Leid, M. (2010). Bcl11b represses a mature T-cell gene expression program in immature CD4(+)CD8(+) thymocytes. Eur. J. Immunol. 40, 2143–2154. Lee, H.-C., Headley, M.B., Iseki, M., Ikuta, K., and Ziegler, S.F. (2008). Cutting edge: Inhibition of NF-kappaB-mediated TSLP expression by retinoid X receptor. J. Immunol. 181, 5189–5193. Lee, H.-C., and Ziegler, S.F. (2007). Inducible expression of the proallergic cytokine thymic stromal lymphopoietin in airway epithelial cells is controlled by NF B. Proceedings of the National Academy of Sciences 104, 914–919. Leung, D.Y., and Bieber, T. (2003). Atopic dermatitis. The Lancet 361, 151–160. 135 Li, M. (2005). Retinoid X receptor ablation in adult mouse keratinocytes generates an atopic dermatitis triggered by thymic stromal lymphopoietin. Proceedings of the National Academy of Sciences 102, 14795–14800. Li, M. (2006). Topical vitamin D3 and low-calcemic analogs induce thymic stromal lymphopoietin in mouse keratinocytes and trigger an atopic dermatitis. Proceedings of the National Academy of Sciences 103, 11736–11741. Liang, X., Bhattacharya, S., Bajaj, G., Guha, G., Wang, Z., Jang, H.-S., Leid, M., Indra, A.K., and Ganguli-Indra, G. (2012). Delayed Cutaneous Wound Healing and Aberrant Expression of Hair Follicle Stem Cell Markers in Mice Selectively Lacking Ctip2 in Epidermis. PLoS ONE 7, e29999. Liu, Y. (2009). Chapter 1 TSLP in Epithelial Cell and Dendritic Cell Cross Talk. In Advances in Immunology, (Elsevier), pp. 1–25. Liu, Y.-J. (2006). Thymic stromal lymphopoietin: master switch for allergic inflammation. J. Exp. Med. 203, 269–273. Nakayama, T., Hieshima, K., Nagakubo, D., Sato, E., Nakayama, M., Kawa, K., and Yoshie, O. (2004). Selective induction of Th2-attracting chemokines CCL17 and CCL22 in human B cells by latent membrane protein 1 of Epstein-Barr virus. J. Virol. 78, 1665–1674. Novak, N., and Leung, D.Y. (2011). Advances in atopic dermatitis. Current Opinion in Immunology 23, 778–783. Palmer, C.N.A., Irvine, A.D., Terron-Kwiatkowski, A., Zhao, Y., Liao, H., Lee, S.P., Goudie, D.R., Sandilands, A., Campbell, L.E., Smith, F.J.D., et al. (2006). Common loss-of-function variants of the epidermal barrier protein filaggrin are a major predisposing factor for atopic dermatitis. Nat. Genet. 38, 441–446. Proksch, E., Brandner, J.M., and Jensen, J.M. (2008). The skin: an indispensable barrier. Exp Dermatol. Proksch, E., Feingold, K.R., Man, M.Q., and Elias, P.M. (1991). Barrier function regulates epidermal DNA synthesis. Journal of Clinical Investigation 87, 1668–1673. 136 Rang, H.P. (2003). Pharmacology (Edinburgh; New York: Churchill Livingstone). Scharschmidt, T.C., and Segre, J.A. (2008). Modeling atopic dermatitis with increasingly complex mouse models. J. Invest. Dermatol. 128, 1061–1064. Soumelis, V., Reche, P.A., Kanzler, H., Yuan, W., Edward, G., Homey, B., Gilliet, M., Ho, S., Antonenko, S., Lauerma, A., et al. (2002). Human epithelial cells trigger dendritic cell–mediated allergic inflammation by producing TSLP. Nature Immunology. Spergel, J. (2003). Atopic dermatitis and the atopic march. Journal of Allergy and Clinical Immunology 112, S118–S127. Takai, T. (2012). TSLP expression: cellular sources, triggers, and regulatory mechanisms. Allergol Int 61, 3–17. Tokura, Y. (2010). Extrinsic and intrinsic types of atopic dermatitis. Journal of Dermatological Science 58, 1–7. Topark-Ngarm, A., Golonzhka, O., Peterson, V.J., Barrett, B., Martinez, B., Crofoot, K., Filtz, T.M., and Leid, M. (2006). CTIP2 associates with the NuRD complex on the promoter of p57KIP2, a newly identified CTIP2 target gene. J Biol Chem 281, 32272–32283. van Vliet, E., Uhrberg, M., Stein, C., and Gleichmann, E. (1993). MHC control of IL-4-dependent enhancement of B cell Ia expression and Ig class switching in mice treated with mercuric chloride. Int. Arch. Allergy Immunol. 101, 392–401. Wang, Z., Coleman, D.J., Bajaj, G., Liang, X., Ganguli-Indra, G., and Indra, A.K. (2011). RXRα ablation in epidermal keratinocytes enhances UVR-induced DNA damage, apoptosis, and proliferation of keratinocytes and melanocytes. J. Invest. Dermatol. 131, 177–187. Yoo, J. (2005). Spontaneous atopic dermatitis in mice expressing an inducible thymic stromal lymphopoietin transgene specifically in the skin. Journal of Experimental Medicine 202, 541–549. Ziegler, S.F. (2010). The role of thymic stromal lymphopoietin (TSLP) in allergic disorders. Current Opinion in Immunology 22, 795–799. 137 Ziegler, S.F., and Artis, D. (2010). Sensing the outside world: TSLP regulates barrier immunity. Nature Immunology 11, 289–293. 138 Chapter 4 General Conclusion 139 Skin, the largest body organ, provides both a physical and a chemical barrier against the outside world (Proksch et al., 2006, 2008; Segre, 2006). The protective skin barrier is a very complicated system formed by a large number of inter-related components including proteins and lipids (Fuchs and Raghavan, 2002). The alteration of any of these components results in altered barrier function(s), which can lead to mild to lethal phenotype. Therefore, altered skin barrier function is a central event in various skin diseases (Segre, 2006). Our study revealed a novel role of transcription factor Ctip2 in skin barrier formation, maintenance and homeostasis. The establishment of an effective physical barrier protecting the living organism from its surrounding environment is the key function of mammalian skin (Hoffjan and Stemmler, 2007). Most of the transport of substances across the stratum corneum (SC) takes place through the lipid bilayer, suggesting its essential role for the barrier function (Jungersted et al., 2008). The major SC lipids, ceramides, cholesterol and fatty acids have distinct roles in maintaining normal barrier functions (Elias and Menon, 1991; Ishikawa et al., 2010). Ceramides as well as cholesterol were shown to have an influence on (and are influenced by) the integrity of the SC, while free fatty acids play a major 140 role in the lipid bilayer formation and pH maintenance (Baroni et al., 2012). We have recently identified Ctip2 as a key regulator of skin lipid metabolism, which plays an important role in establishment of epidermal permeability barrier (EPB). Compared with wildtype, uneven and thinner distribution of neutral lipids was observed in the Ctip2 null mice at E17.5 (Golonzhka et al., 2008). Furthermore, the ultrastructural analyses of the epidermis have revealed defects in lipid disc formation, loosely packed corneocytes, as well as disorganized intercellular lamellar body membranes in Ctip2 null mice (Golonzhka et al., 2008). Ablation of Ctip2 leads to altered lipid composition in the developing mouse epidermis by modulating the expression levels of key enzymes involved in lipid metabolism. We also demonstrated that Ctip2 was recruited to the promoter regions of several genes involved in the ceramide biosynthesis pathways and could directly regulate their expression. Besides stratum corneum, other important components contribute the formation of physical barrier, such as the nucleated epidermis, in particular the cell–cell junctions and associated cytoskeletal proteins (Proksch et al., 2008). Ctip2 null mice skin has fewer corneocytes, which are smaller, more fragile and less resistant to sonication 141 compared to wildtype skin. The linkage of Ctip2 to reduced expression of serine proteases such as channel-activating serine protease 1 (Cap1) and kallikrein-related peptidase (Klk5, Klk7 and Klk14) as well as tight junction proteins (Occludin, tight junction protein 1 and 2) in skin barrier deficiency remains to be addressed (Furuse et al., 2002; Segre, 2003; Leyvraz et al., 2005; Hachem et al., 2006). In addition to the physical barrier, the integrity of the skin is also maintained by the human immune system, which is equipped with a variety of tools that contribute to the establishment of an efficient defense system against various challenges including pathogenic infections. The immune defense system is composed of humoral (antibody mediated) and cellular (T-cell mediated) constituents (Proksch et al., 2008; Baroni et al., 2012). The wide range of invading microorganisms is fended by several immunological facets, including immediate, nonspecific mechanisms (innate immunity), and delayed, stimulus-specific responses (adaptive immunity) (Elias, 2007; Jung and Stingl, 2008; De Benedetto et al., 2012). Our data demonstrated an important role of Ctip2 in skin immune defense system. We demonstrated that keratinocytic ablation of Ctip2 142 leads to atopic dermatitis (AD)-like skin inflammation, characterized by alopecia, pruritus and scaling, as well as extensive infiltration of T lymphocytes and immune cells. We also observed increased expression of T-helper 2 (Th2)-type cytokines and chemokines in the mutant skin, as well as systemic immune responses that share similarity with human AD patients. Furthermore, we discovered that TSLP expression was significantly upregulated in the mutant epidermis as early as postnatal day 1 and ChIP assay revealed that TSLP is likely a direct transcriptional target of Ctip2 in epidermal keratinocytes. These results suggest that upregulation of TSLP expression in the Ctip2ep-/- epidermis could be due to a derepression of gene transcription in absence of Ctip2. Besides these diverse chemical/biochemical strategies, defense system, the skin mainly provides a composed of antimicrobial peptides (AMPs), also named host defense peptides, which kill a wide spectrum of bacteria, viruses and fungi, and some transformed or cancerous cells (Braff et al., 2005; Lai and Gallo, 2009; Borkowski and Gallo, 2011). AMPs build up an innate epithelial chemical shield with a rapid and direct first-line defense system termed as the anti-microbial barrier. AMPs have the ability to enhance immunity 143 by modulating the inflammatory response and activating cellular and adaptive immunity (Schauber and Gallo, 2009; Gallo and Nakatsuji, 2011). Reduced expression of AMPs such as cathelicidin and βdefensin has been observed in Ctip2 null skin epidermis at embryonic stage E18.5 (unpublished data), indicating potential role of Ctip2 in mediating formation of skin chemical/ anti-microbial barrier. The molecular mechanisms underlying Ctip2 mediated regulation of AMP expression need to be identified. Thus, Ctip2 could be involved in a diverse range of events in skin barrier formation, maintenance and homeostasis. Our study suggests that Ctip2 is a bi-functional transcription regulator that can either activate or repress gene expression in a cell and context specific manner. During sphingolipid biosynthesis, Ctip2 activate the transcription of Lass2 and Gba2 genes by binding to their promoter regions; however, in the fulfillment of immunological barrier functions, keratinocytic Ctip2 repress the expression of TSLP gene in a direct pattern by interacting with TSLP’s promoter region. The roles of Ctip2 in other aspects of skin function such as hair cycling and cancer pathogenesis are currently being pursued by Indra group. 144 Ctip1 and Ctip2 are highly related proteins which both interact to COUP-TF family members and have similar sequence-specific DNA binding activities, it is highly possible that they have some overlapping functions in vivo (Avram et al., 2000, 2002). Ctip1 has been identified as a myeloid or B cell proto-oncogene in mouse and human, and it’s required for the normal lymphoid development as well as human fatal hemoglobin expression ( Nakamura et al., 2000; Satterwhite, 2001; Uda et al., 2008; Liu et al., 2003; Sankaran et al., 2008). We have shown that the severity of the epidermal barrier defects in Ctip2 null mice is reduced at later timepoints of development (E18.5 compared to E17.5), which could be due to the compensatory mechanisms mediated by Ctip1 (Golonzhka et al., 2008). A modest increase of Ctip1 expression was observed in Ctip2 null mice at E18.5; however, the role of Ctip1 in epidermal barrier development and maintenance is still unclear (Golonzhka et al., 2008). Future investigation about the expression pattern and distribution of Ctip1 in mouse epidermis during development, as well as its role in cutaneous barrier formation will be performed utilizing Ctip1-null mice. In conclusion, our data suggested the critical role of Ctip2 in regulating epidermal permeability barrier formation and maintenance. On one 145 hand, in embryonic mouse skin, Ctip2 is required for the establishment of functional biosynthesis epidermal and barrier metabolism, by controlling keratinocyte epidermal lipid proliferation and differentiation, as well as the formation of structurally intact components of skin barrier such as lipid discs, cornified envelopes, and intercellular lamellar bodies. On the other hand, Ctip2 is crucial in adult mice epidermal barrier homeostasis. Loss of keratinocytic Ctip2 leads to dysregulated skin barrier characterized with AD-like phenotypes, such as epidermal hyperproliferation, immune cell infiltration, Th2-type skin inflammation, as well as systemic immunological syndrome. This study identifies a new mediator, Ctip2, transcriptionally regulating mouse epidermal barrier formation and homeostasis in both cell-autonomous and non cell-autonomous manner. 146 Bibliography Avram, D., Fields, A., Pretty On Top, K., Nevrivy, D.J., Ishmael, J.E., and Leid, M. (2000). Isolation of a novel family of C(2)H(2) zinc finger proteins implicated in transcriptional repression mediated by chicken ovalbumin upstream promoter transcription factor (COUP-TF) orphan nuclear receptors. J Biol Chem 275, 10315–10322. Avram, D., Fields, A., Senawong, T., Topark-Ngarm, A., and Leid, M. (2002). COUP-TF (chicken ovalbumin upstream promoter transcription factor)-interacting protein 1 (CTIP1) is a sequence-specific DNA binding protein. Biochem J 368, 555–563. Baroni, A., Buommino, E., De Gregorio, V., Ruocco, E., Ruocco, V., and Wolf, R. (2012). Structure and function of the epidermis related to barrier properties. Clinics in Dermatology 30, 257–262. De Benedetto, A., Kubo, A., and Beck, L.A. (2012). Skin Barrier Disruption: A Requirement for Allergen Sensitization? Journal of Investigative Dermatology 132, 949–963. Borkowski, A.W., and Gallo, R.L. (2011). The Coordinated Response of the Physical and Antimicrobial Peptide Barriers of the Skin. J Investig Dermatol 131, 285–287. Braff, M.H., Bardan, A., Nizet, V., and Gallo, R.L. (2005). Cutaneous defense mechanisms by antimicrobial peptides. J. Invest. Dermatol 125, 9–13. Elias, P.M. (2007). The skin barrier as an innate immune element. Seminars in Immunopathology 29, 3–14. Elias, P.M., and Menon, G.K. (1991). Structural and lipid biochemical correlates of the epidermal permeability barrier. Adv. Lipid Res 24, 1– 26. Fuchs, E., and Raghavan, S. (2002). Getting under the skin of epidermal morphogenesis. Nat Rev Genet 3, 199–209. Furuse, M., Hata, M., Furuse, K., Yoshida, Y., Haratake, A., Sugitani, Y., Noda, T., Kubo, A., and Tsukita, S. (2002). Claudin-based tight junctions are crucial for the mammalian epidermal barrier: a lesson from claudin-1-deficient mice. J. Cell Biol. 156, 1099–1111. 147 Gallo, R.L., and Nakatsuji, T. (2011). Microbial Symbiosis with the Innate Immune Defense System of the Skin. Journal of Investigative Dermatology 131, 1974–1980. Golonzhka, O., Liang, X., Messaddeq, N., Bornert, J.-M., Campbell, A.L., Metzger, D., Chambon, P., Ganguli-Indra, G., Leid, M., and Indra, A.K. (2008). Dual Role of COUP-TF-Interacting Protein 2 in Epidermal Homeostasis and Permeability Barrier Formation. J Investig Dermatol 129, 1459–1470. Hachem, J.-P., Houben, E., Crumrine, D., Man, M.-Q., Schurer, N., Roelandt, T., Choi, E.H., Uchida, Y., Brown, B.E., Feingold, K.R., et al. (2006). Serine Protease Signaling of Epidermal Permeability Barrier Homeostasis. Journal of Investigative Dermatology 126, 2074–2086. Hoffjan, S., and Stemmler, S. (2007). On the role of the epidermal differentiation complex in ichthyosis vulgaris, atopic dermatitis and psoriasis. British Journal of Dermatology 157, 441–449. Ishikawa, J., Narita, H., Kondo, N., Hotta, M., Takagi, Y., Masukawa, Y., Kitahara, T., Takema, Y., Koyano, S., Yamazaki, S., et al. (2010). Changes in the Ceramide Profile of Atopic Dermatitis Patients. Journal of Investigative Dermatology 130, 2511–2514. Jung, T., and Stingl, G. (2008). Atopic dermatitis: Therapeutic concepts evolving from new pathophysiologic insights. Journal of Allergy and Clinical Immunology 122, 1074–1081. Jungersted, J.M., Hellgren, L.I., Jemec, G.B.E., and Agner, T. (2008). Lipids and skin barrier function – a clinical perspective. Contact Dermatitis 58, 255–262. Lai, Y., and Gallo, R.L. (2009). AMPed up immunity: how antimicrobial peptides have multiple roles in immune defense. Trends in Immunology 30, 131–141. Leyvraz, C., Charles, R.-P., Rubera, I., Guitard, M., Rotman, S., Breiden, B., Sandhoff, K., and Hummler, E. (2005). The epidermal barrier function is dependent on the serine protease CAP1/Prss8. J. Cell Biol. 170, 487–496. Liu, P., Keller, J.R., Ortiz, M., Tessarollo, L., Rachel, R.A., Nakamura, T., Jenkins, N.A., and Copeland, N.G. (2003). Bcl11a is essential for normal lymphoid development. Nature Immunology 4, 525–532. 148 Proksch, E., Brandner, J.M., and Jensen, J.M. (2008). The skin: an indispensable barrier. Exp Dermatol. Proksch, E., Fölster-Holst, R., and Jensen, J.-M. (2006). Skin barrier function, epidermal proliferation and differentiation in eczema. Journal of Dermatological Science 43, 159–169. Sankaran, V.G., Menne, T.F., Xu, J., Akie, T.E., Lettre, G., Van Handel, B., Mikkola, H.K.A., Hirschhorn, J.N., Cantor, A.B., and Orkin, S.H. (2008). Human Fetal Hemoglobin Expression Is Regulated by the Developmental Stage-Specific Repressor BCL11A. Science 322, 1839– 1842. Schauber, J., and Gallo, R.L. (2009). Antimicrobial peptides and the skin immune defense system. J. Allergy Clin. Immunol. 124, R13–18. Segre, J. (2003). Complex redundancy to build a simple epidermal permeability barrier. Curr. Opin. Cell Biol. 15, 776–782. Segre, J.A. (2006). Epidermal barrier formation and recovery in skin disorders. J Clin Invest 116, 1150–1158.