£%. 3.. ‘114 I VII-p . . . .52...» .. u... . :1: 1.12:3: . . . A... Fairs .314. mil 7:5... 33.3.... k d. 1,... 1.2. .. .T . 2.. t..-) . 1.2.... 1 , $3.11 I: fix: . 3. 3.32:». .. {in .v ‘1: i .3 A ...§R.~.¢.I.uat.lvni& - .«~.Ln.1tp’ rlJr; 7...! .55.. .3) 3.. . 9 52.97 . :.\ .7 7y 8%... _\ v feta-Lulu». 4. .11 t I. .11.: . 2 V1 .1 .6... .rl) . . .1. r ‘. s: magma 300? This is to certify that the dissertation entitled FUNCTIONAL CHARACTERIZATION OF Drosophila C-TERMINAL BINDING PROTEIN presented by Priya Mani has been accepted towards fulfillment of the requirements for the degree in Cell and Molecular Biology MQMM Major Professor’s Signature Il/Ilj/Dé Date MSU is an Affimtative Action/Equal Opportunity Institution LIBRARY Michigan State University PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. i DATE DUE DATE DUE DATE DUE 2/05 p2/CIRCIDateDue.indd-p.1 FUNCTIONAL CHARACTERIZATION OF Drosophila C-TERMINAL BINDING PROTEIN By Priya Mani A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Program in Cell and Molecular Biology 2006 ABSTRACT FUNCTIONAL CHARACTERIZATION OF Drosophila C-TERMINAL BINDING PROTEIN BY Priya Mani Transcriptional repression results from the selective actions of negatively acting repressors and corepressors that function by multiple mechanisms. C-terminal Binding Protein (CtBP) is one such interesting corepressor important for development, oncogenesis and transcriptional regulation. Distinct forms of this protein have been uncovered in vertebrates that are capable of performing a variety of functions, both in the nucleus and cytoplasm. All CtBP isoforrns have a high degree of similarity to NAD- dependent hydroxy acid dehydrogenase enzymes, with some isoforrns carrying C-tenninal extensions (tail). Distinct CtBP proteins are produced in Drosophila that differ in the presence or absence of the C-terminal tail. The significance of the homology to metabolic dehydrogenase enzymes and the function of the C-tenninal extension is still largely unknown. Herein we demonstrate that the expression and coding information for distinct CtBP isoforrns differing in their C-terrnini is a conserved feature of phylogenetically divergent organisms. in many Drosophila species, the relative levels of isoforrns are altered during development with a significant drop in the tailed isoform following embryogenesis. This developmental shift in protein levels can be traced back to a difference in the abundance of individual spliceform levels and points to the tailed isoform contributing an evolutionarily conserved role. lntriguingly, all CtBP proteins contain dehydrogenase domains that include an nucleotide (NAD) binding Rossman fold, a putative catalytic core and a broad dimerization region. To investigate the significance of this dehydrogenase homology, mutants in these critical domains were misexpressed in the fly and analyzed for biological activity. Overexpressed wild type isoforms and catalytic mutants resulted in aberrant phenotypes while the NAD binding mutant and the dimerization mutants were completely disrupted for biological activity, with low protein expression. We show that the ability to dimerize is important for CtBP function and the biological activity of the catalytic mutant might be a consequence of dimerization with endogenous CtBP proteins. Our results also indicate that NAD binding is not needed for dimerization and we speculate that it may be required for association with cofactors. The experiments described here provide a broader understanding of CtBP function in the context of the whole organism, and should in turn facilitate our mechanistic understanding of CtBP function and eukaryotic gene regulation. To my Dad, Mr. Chandra Shekhar Mani, my Mom, Mrs. Lakshmi Mani, my husband, Dr. Adwait U. Telang AND To my mentor, Dr. David N. Amosti iv ACKNOWLEDGEMENTS This thesis is by far the most significant scientific accomplishment of my life and it would not exist without the people who supported me and believed in me. I am indebted to my mentor Dr. David Arnosti, for his guidance and his personal commitment towards my graduate training. In his own unique way, he challenged and inspired me to be a better thinker, a better orator and a better researcher. Thanks to him, science will always be fun for me! My experience in this lab taught me much more than just science, I learnt that the virtues of patience, dedication and tenacity can take you a long way and these are qualities that I take with me. I am very grateful to members of my thesis committee, Dr. R. William Henry, Dr. Laurie S. Kaguni, Dr. Karl Olson and Dr. Donald Jump for their insightful ideas and constructive comments that helped to shape my manuscript and this thesis. Many thanks to my colleagues in the Amosti laboratory, past (Meghana Kulkarni, Paolo Struffi, Scott Keller and Montserrat Sutrias-Grau) and present (Sandhya Payankaulam, Carlos Martinez, Walid Fakhouri, Yang Zhang, Li Li and Martin Buckley) for their friendship and scientific expertise. I am especially grateful to Meghana Kulkarni for getting me started in the Amosti lab and for her guidance through my formative years. Many thanks to Montserrat Sutrias-Grau whose work served as a starting point for this thesis. I am also very grateful to Sandhya Payankaulam for being a close friend and confidante, and for pushing me towards my goal. I am also very thankful for my friends and colleagues in various departments (Gauri Jawdekar, Dorothy Tappenden, Soumya Korrapati, Tejas Kadakia, Chetan Sukuru, Zakir Ullah and Deborah Yoder) for being there in good and bad times. To my family, thank you for being my family and for being there; my deepest gratitude to my mom, dad, sister, mother-in-law and brother-in-law who taught me to be tough by example. Finally, I don’t have enough words to thank my husband Adwait, who supported my ambition in getting a Ph.D. years ago, and from whom I received a daily dose of motivation to accomplish it. I thank him for his acceptance, love, and understanding of my elevated stress levels, especially in the last two years. He has my heartfelt gratitude for urging me to look at the bigger picture in life ...always. vi TABLE OF CONTENTS LIST OF FIGURES ................................................................................ ix LIST OF TABLES .................................................................................. xi KEY TO ABBREVIATIONS .................................................................... xii CHAPTER I Introduction ...................................................................................... 1-45 1.1 CtBP family of proteins ......................................................... 3 1.2 Domain structure — Implications for CtBP function ...................... 5 1.3 Regulation of CtBP proteins ................................................. 10 1.4 Mechanism of CtBP mediated transcriptional repression ............ 20 1.5 A distinct cytosolic role ascribed to CtBP ................................. 22 1.6 CtBP as a corepressor during Drosophila development .............. 24 1.7 Alternative splicing of Drosophila CtBP ................................... 34 CHAPTER II Developmental Expression and Phylogenetic Characterization of CtBP corepressor protein ......................................................................... 46-76 Abstract .................................................................................... 47 Introduction ............................................................................... 48 Materials and Methods ................................................................. 52 Results ..................................................................................... 57 Discussion ................................................................................. 71 References ................................................................................ 74 CHAPTER III NAD binding modulates CtBP biological activity .................................... 77-110 Abstract .................................................................................... 77 Introduction ................................................................................ 78 Materials and Methods ................................................................. 82 Results ..................................................................................... 87 Discussion ............................................................................... 103 References .............................................................................. 108 vii CHAPTER IV Conclusions and Future Directions ................................................... 111-123 APPENDIX A Rescue of CtBP function in the fly .................................................... 124-140 APPENDIX B An RNAi approach to depleting the CtBPL isoform ............................... 141-147 viii Figure l-1: Figure l-2: Figure I-3: Figure l-4: Figure "-1: Figure "-2: Figure "-3: Figure "-4: Figure III-1: Figure Ill-2: Figure III-3: Figure III-4: Figure Ill-5: LIST OF FIGURES Key domains of CtBP proteins ....................................... 9 CtBP genomic structure and splicing patterns .................. 14 Drosophila CtBP proteins are ubiquitously distributed during development ............................................................ 28 Alignment of different CtBP isoforms generated by alternative splicing in Drosophila ................................ 36 Developmental expression profile of CtBP isoforms in Drosophila melanogaster ............................................ 59 Conservation of coding information for CtBPL-specific C- terminus ............................................................. 63, 64 Conserved developmental regulation of CtBP protein expression in Drosophila moja vensis and Drosophila virilis ....................................................................... 67 Adult expression of CtBP proteins in four Drosophila species, Anopheles gambiae, Apis mellifera and Tribolium castaneum ............................................................... 69 Phenotypes induced by overexpression of wild type and mutant CtBP proteins ................................................. 88 Relative steady state levels of recombinant CtBP proteins: CtBPL (NAD) and CtBPL (DIM) mutants and CtBPs isoform are expressed at low levels ......................................... 93 CtBPL (CAT) protein is capable of dimerization .............................................................. 95 lmmunoprecipitation of endogenous CtBPL and CtBPs with FLAG-tagged CtBP proteins indicate CtBP homo— and heterodimerization .................................................... 98 CtBPL (NAD) and CtBPL (DIM) mutants are expressed at low steady state levels ..................................................... 99 ix Figure III-6: Figure A-1: Figure A-2: Figure A-3: Figure A-4: Figure 8-1: NAD binding mutant migrates in molecular mass consistent with dimers but not the dimerization mutant .................. 101 Genomic organization of the Drosophila CtBP locus ....... 126 Expression of CtBP isoforms under the control of endogenous promoter .............................................. 127 Expression of recombinant CtBPL assayed by Western blotting .................................................................. 128 Genetic crosses for whole animal rescue of CtBP mutant phenotype .............................................................. 131 A transgenic system to trigger RNAi and selectively eliminate CtBPL in Drosophila ................................................ 144 Images in this dissertation are presented in color. LIST OF TABLES Table Ill-1: Quantitation of CtBP misexpression phenotypes .............. 90 Table A-1: CtBP promoter driven transgenes do not rescue CtBP mutants ................................................................. 133 xi aa bp cDNA: Co-IP CtBP dCtBP DNA DTT Eco/i EDTA HAT HDAC HEPES HMT hsp Kb KDa NAD NLS PAGE PcG PCR Pol II Rb RNA RNAi RT-PCR TSA Wt KEY TO ABBREVIATIONS amino acid anterior-posterior base pair(s) complementary deoxyribonucleic acid Co-immunoprecipitation C-terrninal Binding Protein Drosophila C-tenninal Binding Protein Deoxyriboncleic acid Dithiothreitol Escherichia coli Ethylene dinitrilo tetra acetic acid Histone acetyltransferase Histone deacetylase N-2-hydroxyethylpiperazine—N’-2-ethanesulfonic acid Histone methyltransferases heat shock promoter Kilo base Kilo Dalton Nicotinamide adenine dinucleotide Nuclear localization signal Poly acrylamide gel electrophoresis Polycomb Polymerase chain reaction RNA Polymerase ll Retinoblastoma tumor suppressor protein Ribonucleic acid RNA interference Reverse transcriptase polymerase chain reaction Trichostatin A Wild type xii CHAPTER I INTRODUCTION Gene expression in eukaryotes is a highly elaborate process under tight regulation at several junctures. An ever-growing body of evidence shows that changes in transcriptional profiles form a vital part of the genetic basis of the evolution of development and contribute to animal diversity (Wray, 2003). Developmental processes are regulated by extensive protein: protein and protein: DNA interactions during the regulation of transcription. Appropriate transcriptional regulation is thus central to cellular homeostasis, and misregulation of gene expression has been implicated in several disease states. Mutations that disrupt developmental processes often map to loci encoding transcription factors, which emphasizes the importance of transcription in development. These regulatory factors are the key to spatio— temporal differential gene expression that must transpire for appropriate cellular differentiation and development to ensue (Lawrence, 1992). The specificity of differential gene expression in eukaryotes is controlled by the coordinated actions of sequence specific, context-dependent activator and repressor proteins. Early investigations have led to the identification of these coregulators and recent efforts are aimed at the precise characterization of their molecular function. Repressor proteins are aided by corepressor proteins that function by multiple mechanisms and may either help quantitatively by enhancing the overall repression activity (Struffi et al., 2004) or qualitatively, by providing a unique repression activity (Lunyak et al., 2002). Data from several sources suggests that such corepressors link sequence-specific repressors to the core RNA polymerase II machinery to accomplish direct repression (Austin and Biggin, 1995; Mannervik et al., 1999). In addition, corepressors are also aided by histone modifying activities like deacetylases, methyl transferases and newly-identified demethylases which act cooperatively and are documented to be important players of several repressor assemblies (Arnosti, 2004; Chinnadurai, 2006). An expanding array of post-translational modifications is being acknowledged as means of regulating protein localization and abundance. The ability to use various repression mechanisms in a combinatorial fashion may be exploited to provide unique patterns of gene expression. Current investigations are aimed at defining the precise mechanisms of cofactor function and regulation. C-terminal Binding Protein (CtBP) is a versatile prototype of an expanding family of corepressor proteins that aids in the repression of a diverse array of DNA binding transcriptional repressors in vertebrates and invertebrates alike. In this thesis, I describe studies that analyze the different isoforms of the Drosophila homolog of CtBP and reveal dynamic changes occurring in protein levels during development. Recently, the cellular redox status has also been suggested to influence gene transcription (lmai et al., 2000; Rutter et al., 2001). CtBP proteins resemble NAD-dependent dehydrogenases in structure but whether this similarity extends to function and whether such activity is linked to transcription is unclear. This study also extends to testing the significance of dehydrogenase-like conserved domains within CtBP in the context of a whole organism. These investigations are fundamental to understanding the molecular basis of CtBP’s function as a corepressor involved in development. 1.1 CtBP family of proteins The founding member of the C-terrninal Binding Protein (CtBP) family was discovered in 1993 as a 48KDa cellular phosphoprotein that associated with the C-terminal region of the adenoviral 2/5 E1A oncoprotein. This interaction was found to negatively modulate oncogenic transformation, tumorigenesis and metastasis by E1A. Binding was found to depend on a PLDLS (Pro-Leu-Asp-Leu-Ser) motif present near the C-terminal end of E1A protein, conserved among E1A proteins of primate adenoviruses (Boyd et al., 1993; Schaeper et al., 1995). The N-terminal exon of E1A is capable of cellular transformation, but the presence of this motif was shown to restrain exon 1 mediated transformation, suggesting that CtBP binding could influence E1A mediated gene expression (Sollerbrant et al., 1996). Deletion of the CtBP-binding motif abrogated the repressive activity of the C-terminal region. These studies provided early evidence of CtBP acting to antagonize transactivation and led to cloning of the cDNA for human CtBP1 (Schaeper et al., 1995). Shortly thereafter, a highly related human protein termed CtBP2 of 48KDa was identified by BLAST analysis of EST databank sequences. Coincidentally, CtBP2 was simultaneously discovered in a two-hybrid screen using the murine BKLF (Basic Kruppel-like factor) as prey (T umer and Crossley, 2001). Human CtBP1 and CtBP2 are present on different chromosomes and share over 83% amino acid similarity (Katsanis and Fisher, 1998). Other vertebrates such as mice and frogs also have two CtBP homologs, while the invertebrate genomes including those of C. elegans and Drosophila melanogaster contain a single gene for CtBP. It is in the fly system that an early role for CtBP in transcriptional repression was established (See Section 1.5). Since then, cell culture based assays and vertebrate models have conclusively shown that CtBP is an essential protein and acts as a bonafide corepressor in these systems. CtBP is essential for vertebrate development In mice, both Ctbp1 and Ctbp2 transcripts are expressed widely during development in unique and overlapping patterns (Furusawa et al., 1999). To address the in vivo significance of multiple CtBP functions, mice carrying mutations in both CtBP1 and 2 were generated. Ctbp1-null mice are reduced in size but viable. In contrast Ctbp2-null mice perish by embryonic day 10.5 and show defects in several tissues. This data demonstrates that there are spatial differences between expression patterns of the proteins and suggests that they might have unique functions. CtBP proteins have also been ascribed roles in regulating Golgi dynamics (See Section 1.4), however no Golgi defects in these mutants was noted. Compound mutants for Ctbp1-I- and Ctbp2-l- arrested at the head fold stage and a reduction in the dosage of one gene enhanced the severity of phenotype associated with the mutation in the other gene (Hildebrand and Soriano, 2002). These results indicate that CtBP1 and 2 genetically interact. A similar picture emerges from the recent avian expression study of Ctbp1 and Ctbp2, wherein the proteins are expressed in partly overlapping expression domains in the avian embryo (Van Hateren et al., 2006). This study suggests that avian Ctbp1 and Ctbp2 might show functional redundancy in some tissues and specific roles in others. 1.2 Domain structure - Implications for CtBP function From a structural standpoint, CtBP proteins can be divided into three distinct domains. The first is the substrate-binding cleft that is responsible for specific recognition of PXDLS motifs in binding partners. Co—crystallization of rat CtBP with NAD (H) and a PXDLS like peptide show the presence of this peptide bound to the N-terminus of CtBP. Structural comparisons between binary and ternary complexes reveal that binding of this short peptide is not associated with any major conformational changes in the protein and the peptide binding cleft has no contact with the nucleotide binding region (Nardini et al., 2003). The second and most conserved of all includes the central nucleotide binding regions. The initial cloning of human CtBP1 and subsequent homology searches revealed extensive homology with NAD-dependent D- isomer specific 2-hydroxy acid dehydrogenases (Schaeper et al., 1995). This homology extends over a nucleotide/NAD binding Rossman fold (GXGXXG) and a putative catalytic histidine residue (towards the C-terminus) in the active site of these enzymes. Dehydrogenase activities have not been previously associated with transcriptional repressors. The structural similarities prompted speculation that CtBP might possess an intrinsic dehydrogenase-like activity that assists in remodeling chromatin structure (Kumar et al., 2002). More importantly, it also pointed to a role for NAD as a modulator of gene expression. Cellular NAD levels have been linked to transcription before when the Sir2 histone deacetylase (HDAC) was identified as having an NAD- dependent HDAC activity (lmai et al., 2000). Early attempts to identify dehydrogenase-like activity associated with CtBP were unsuccessful, however a weak activity was reported for CtBP1 (Kumar et al., 2002; Schaeper et al., 1995). Here, Kumar and colleagues tested the dehydrogenase activity of CtBP by combining the reduction of pyruvate to lactate with the oxidation of NADH to NAD+. Recombinant CtBP was able to catalyze this in vitro enzymatic reaction in a dose dependent manner, albeit inefficiently, indicating it is a functional dehydrogenase. The crystal structure for CtBP’s minimal dehydrogenase domain revealed a CtBP monomer that forms extensive dimer contacts with another monomer, in the presence of NAD. The dehydrogenase-like domains provide the majority of the contacts needed for dimerization. NAD binds within a cleft at the confluence of two such monomers, inducing conformational changes that allow enhanced binding to transcription factors like E1A. When tested biochemically, mutations in key residues of the dinucleotide binding fold (G181V,G183V, D204A) or the dimerization interface (R141A, R142A, R163A, R171A) disrupt binding to E1A and impair repression by E1A (Kumar et al., 2002). Also conserved in CtBP is the His/Glu/Asp triad that constitutes the active center of dehydrogenases. These residues are conserved in CtBP proteins from vertebrates and invertebrates (See Figure l-1). Mutations in these residues were also shown to compromise the ability of CtBP to contact E1A, and the protein was non-functional when tested as a corepressor. Moreover, this putative catalytic mutant was unable to catalyze the dehydrogenase reaction, unlike the wild type CtBP (Kumar et al., 2002). These observations lend support to the PXDLS binding cleft making direct contacts with the dehydrogenase-like regions. The structural data described above agrees well with the crystal structure of CtBP3/BARS (a truncated form of rat CtBP1, discussed in Section 1.5). Overall, evidence from both reports concur that dinucleotide binding triggers a closed conformation, triggering closer contacts at the dimerization interface. However, there are some minor differences noted. BARS was co- crystallized was with a short PXDLS-like peptide and the peptide was found to associate exclusively with the N-terminal part of CtBP and binding of the peptide does not induce any noticeable structural alterations (Nardini et al., 2003). This is in contrast to the observations made by Kumar and colleagues for CtBP1, wherein the PXDLS motif is predicted to directly interact with the dehydrogenase-like regions, on the basis of structural data and mutagenesis experiments. Mutations in dehydrogenase-like regions (61720 in the nucleotide binding region or H304L in the catalytic center) were found to prevent NAD binding, but did not affect binding to transcription factor E1A. Lastly, the least well-conserved part of the protein is the variable C- terminal extension. This C-terminal tail harbors sites for various post- translational modifications and may play a regulatory role (discussed in Section1.3, See Figure l-1). Vertebrate CtBP proteins all possess C-terrninal extensions of ~90 residues that are predicted to be intrinsically unstructured in solution, being composed primarily of disorder promoting residues (56%) Figure M: Key domains of CtBP proteins CtBP proteins display striking similarity to alpha hydroxy acid dehydrogenases. Regions of highest homology between CtBP and hydroxy- acid dehydrogenases are shown in blue shading. The putative catalytic residue His-315 and the central nucleotide (NAD) binding domain are indicated (Chinnadurai, 2002). The PXDLS binding region has been mapped to the N-terminus of the vertebrate protein (Koipally and Georgopoulos, 2000; Nardini et al., 2003). Residues important for dimerization map within the dehydrogenase domains. CtBP is subject to a host of post-translational modifications, the majority of which have been identified to act on the C- terrninal tail of vertebrate CtBP proteins. Phosphorylation sites by Pak1, HlPK2 and c-Jun-NHZ kinase, sumoylation motif, PXDLS binding region and the nNOS-PDZ binding regions are indicated. wm100 fold) (Fjeld et al., 2003), however other studies find NAD+/NADH to be equally effective in enhancing CtBP binding E1A (Kumar et al., 2002). Thus, a 10 speculative model has been suggested for CtBP to play the part of a cellular redox sensor that interprets the cellular metabolic demands and accordingly directs transcription (Zhang et al., 2002). Binding of dinucleotide is thought to bring about conformational changes in protein structure that stimulate dimerization (Balasubramanian et al., 2003). This might be one mechanism by which CtBP proteins enter the nucleus and regulate target genes. In this manner, NAD can be considered a switch that assembles the CtBP complex. A good example of the impact of NAD on CtBP activity is a recent study describing CtBP to be a corepressor for neuronal genes such as those involved in drug-resistant epilepsy. CtBP was recruited by NRSF (neuronal restrictive silencing factor) and was found to enhance NRSF-dependent repression. The CtBP-NRSF direct interaction was disrupted upon increasing levels of NADH (by speeding up glycolysis) resulting in de-repression of neuronal targets. Conversely, the glycolytic inhibitor 2DG (2-deoxy-D-glucose) was found to enhance NRSF directed repression of target genes, that showed a marked reduction in levels of H3-K9 acetylation (mark of activation) and an increase in H3-K9 methylation (mark of repression). Repression was lost in CtBP null mouse embryonic fibroblasts (MEFs) (Garriga-Canut et al., 2006). This demonstration of small molecule regulation of energy metabolism directly impacting chromatin structure and gene expression in disease states is novel, but altering central processes like glycolysis is very likely to cause pleiotropic effects. 11 Multiple CtBP proteins are encoded by multifunctional genes In mammals, the CtBP family is encoded by two genes: Ctbp1 and Ctbp2. Both genes generate multiple isoforms by either alternative splicing or differential promoter utilization (See Figure l-2). Ctbp1 undergoes alternative splicing at the N-terminus to yield two splice variants: CtBP1-L (Long et al.) and CtBP1-S (short) that differ by the presence of 11aa additional at the N- terminus of CtBP1-L. Differential promoter usage and gene splicing from Ctbp 2 generates CtBP2 and a retinal-specific variant, RIBEYE (Chinnadurai, 2002). RIBEYE protein is expressed from a tissue specific promoter located within an intron in Ctbp2 that results in a large (~ 565 aa, also called the A domain) N-terminal domain being fused to CtBP2 except for the 20 aa from the N-terminus (Schmitz et al., 2000). CtBP2 uses an upstream promoter and alternative splicing to eliminate the exon that encodes the N-terminal domain of RIBEYE. CtBP1-L and CtBP2 have been deemed corepressors, while CtBP1-S has been ascribed a unique cytoplasmic role in Golgi fissioning. CtBP1—L has been found to associate with synaptic ribbons and its roles in the cytoplasm are speculative. The RIBEYE variant of CtBP2 is found in abundance in specialized photoreceptor ribbon synapses involved in vision, and may be involved in regulating neurotransmitter release (Schmitz et al., 2000). Interestingly, the exon encoding the RIBEYE specific region is absent in worm and fly genomes, suggesting it is a vertebrate novelty. A recent study also reported the identification of a splice variant of CtBP2 that is localized 12 Figure I-2: CtBP genomic structure and splicing patterns A. Alternative splicing of the vertebrate CtBP1 gene generates CtBP1-L and CtBP1-S possessing divergent functions. Exons exclusive to CtBP1-L (red) and CtBP1-S (blue) are color coded. B. Alternative promoter utilization and gene splicing create CtBP2 that includes an NLS (yellow box) while RIBEYE is produced from an intronic promoter that encodes a large A domain fused to the remainder of the CtBP2 coding sequence. RIBEYE hence lacks the NLS. CtBP2-S is a splice variant of CtBP2 that is devoid of the NLS and thus predominantly cytosolic. C. Drosophila melanogaster CtBP is spliced to produce CtBPL and CtBPs that differ by the presence of the C-terrninal extension encoded by exons 6 and 7 (numbered orange boxes), exclusive to CtBPL. Cartoons are not drawn to scale and are adapted from (Verger et al., 2006). 13 Cpr1 CtBP1-L LI--—: l—L ooreressorp a m 7 ~ ,Golgi Ll CtBP1-S [I-—-—---—---~~I T---- rim-"cram“u'i if“: fission 3- 5' UTR NLS 0‘pr l3 f3 “it. CtBP2 DWDMEmflE—U*fl§flfi- Corepressor CtBP2-S DmfI-m—Dm-WU-E-D-flfiflfifij Unknown my.-~--n---a-—a~clmi£:1Swap“ fibbons C. dCtbp ”dig/h CtBPs E-E—:3~Q~Ej Corepressor CtBPL E~E-flflij———~[:ID Corepressor specifically to the cytoplasm (See Figure l-2 and Section 1.4) (Verger et al., 2006). The Drosophila homolog of CtBP is also subject to alternative splicing and generates two major isoforms (See Figure l-2) as discussed in detail in Section 1.5 and Chapter 2. PXDLS interactions: Significance and determinants of binding Early studies with the E1A protein suggested that CtBP is recruited to promoters through interactions with a PXDLS motif (Pro-Leu-Asp-Leu-Ser in E1A)), where X is usually a bulky group like Leucine, lso-leucine or Valine. This has been found true for dozens of transcriptional effectors containing PXDLS-like motifs that bind CtBP. These include zinc-finger proteins like lkaros (Perdomo and Crossley, 2002), BKLF (van Vliet et al., 2000) , FOG, Snail, delta EF1 (Furusawa et al., 1999), Evi-1 (Palmer et al., 2001), bHLH group proteins like Hairy (Poortinga et al., 1998) and viral oncoproteins like E1A. Subsequent studies confirmed that a vast majority of DNA binding transcription factors that need CtBP contained variants of this motif, some in more than one copy. Mutation in this short stretch of residues has been tested in many proteins and has been found to abolish CtBP interaction and repression (Boyd et al., 1993; Keller et al., 2000; V0 et al., 2001). Most notable is a study describing a naturally occurring mutation in the PXDLS in human homeodomain protein, TGIF. As a consequence of this mutation, TGIF-CtBP interaction is disrupted, resulting in holoprosencephaly and craniofacial abnormalities (Melhuish and Wotton, 2000). In Xenopus, the 15 hematopoietic transcription factor FOG represses erythroid development by the recruitment of CtBP via a PXDLS motif. Mutations in this motif are seen to abrogate the interaction. These results suggest that CtBP plays an important role in hematopoiesis (Deconinck et al., 2000) . CtBPs also bind transcription factors that do not contain an identifiable PXDLS. Many of the vertebrate effectors like HDACs (HDAC1, 2 and 5) fall into this category. This is also true for the Drosophila HDAC de3 that does not have an discernible dCtBP binding motif but is a part of the Knirps repressive complex that includes CtBP, and interacts physically and functionally with Knirps (Struffi and Arnosti, 2005). CtBP physically interacts with several histone acetyltransferase (HAT) enzymes whose bromodomains contain a consensus PXDLS like motif. However, deletions of the bromodomain or mutations in the PXDLS within the bromodomain retain interaction with CtBP, suggesting that additional non-canonical sites must exist (Senyuk et al., 2005). Each unit of CtBP harbors a PXDLS binding cleft, suggesting that dimers can serve to bridge two PXDLS partners together. When fused to a heterologous DNA binding domain, disruption of the PXDLS responsive cleft in CtBP does not affect its repression activity. In contrast, repression is abolished when tested in the absence of the heterologous domain, suggesting that the cleft does not determine association with other effectors, but primarily functions to localize CtBP to repressors bound at target promoters (Quinlan et al., 2006). In the case of many transcription factors, it has been suggested that neighboring residues flanking the PXDLS motif might contribute to the folding 16 capacity of this region and consequently affect its interaction with CtBP. Structures of synthetic peptides examined using NMR spectroscopy suggest that peptides have different affinities for different CtBPs eg. the Drosophila Hairy protein binds Drosophila CtBP stronger than human CtBP suggesting alternative CtBPs have distinct binding specificities (Molloy et al., 2001). In E1A protein, acetylation of a lysine residue close to the PXDLS motif in the C- terminus disrupts binding to both CtBP1 and 2 when tested as a synthetic peptide. This interference could be through structural changes, charge neutralization or a change in the subcellular localization of CtBP (Molloy et al., 2006). Mechanisms dicta ting Localization and Stability of CtBP proteins CtBP1 and 2 are highly similar proteins and possess similar transcriptional repression activities, although new evidence suggests that their activities can be differentially regulated, by specific control of their subcellular location. CtBP proteins have been detected both in the nucleus and in the cytoplasm. Much of our current knowledge of CtBP localization is derived from studies centered on CtBP1-L, whose cellular location is actively regulated. CtBP1-L is predominantly cytoplasmic in Cos cells but when coexpressed with a PXDLS containing protein, such as Net or HIC1 transcription factors, it is relocated into the nucleus (Criqui-Filipe et al., 1999; Deltour et al., 2002). CtBP1-L has been shown to interact with the PDZ domain (protein-protein interaction domain; PSD-95, discs-large and zona 17 occludens-1) of nNOS (neuronal nitric oxide synthase) via a C-terrninal DXL sequence, which shuttles CtBP from the nucleus to the cytoplasm. This mechanism might also potentially serve to make CtBP available for its cytoplasmic roles as discussed in Section 1.5. Posttranslational modifications are also accountable for CtBP subcellular distribution. CtBP1-L is SUMOylated at Lys 428 via P02 (Polycomb group protein) and this modification has profound effects on its distribution into the nucleus, antagonistic to that of nNOS. Mutation of Lys 428 into Arg 428 shifts CtBP from the nucleus to the cytoplasm and consistent with its localization, restricts CtBP from acting as a corepressor (Lin et al., 2003). CtBP2 lacks this SUMOylation site and the PDZ binding domain and is likely to be regulated differently. Independent phosphorylation of CtBP by three different kinases also plays a regulatory role. The p21-activated kinase (Pak1) has been shown to phosphorylate CtBP1-L on Ser 158, a modification that relocates CtBP into the cytoplasm similar to nNOS activity (Barnes et al., 2003). Homeodomain Interacting Protein kinase 2 (HIPK2) is a serine/threonine kinase involved in transcription and apoptosis. HIPK2 targets Ser 422 in the C—terminus of CtBP for phosphorylation. HIPK2 has previously been shown to phosphorylate p53 in response to UV stress, thereby promoting apoptosis by induction of genes like Bax, PERP, p21 and Noxa which are also upregulated in CtBP knockout MEFs (Grooteclaes et al., 2003). HIPK2 modification marks CtBP for UV triggered ubiquitination and subsequent degradation via the proteasome providing an alternative pathway for apoptosis, by a p53 independent mechanism (Zhang et al., 2005). c-Jun-NH2 terminal kinase is the third 18 kinase that also targets Ser 422 for phosphorylation and degradation. C-jun- NH2 kinase plays important roles in inducing apoptosis under cellular stress conditions like UV irradiation or cisplatin treatment (Wang et al., 2006). Under both these conditions, CtBP levels were markedly reduced. These observations indicate Ser 422 to be a mark for CtBP proteasomal degradation and lead to the speculation that CtBP may respond to a wider network of signals than those currently known. Amongst the CtBP proteins, CtBP1-S and RIBEYE appear to have roles in the cytosol, CtBP2 is predominantly nuclear and CtBP1-L is both nuclear and cytoplasmic. A search for regions that serve as signals for localization within CtBP1-L and CtBP2 identified a nuclear localization signal (NLS) within CtBP2 (Verger et al., 2006). This ‘KRQR’ sequence is present in the first exon of CtBP2 and is critical for its nuclear distribution. This motif is not present in CtBP1-L. Within this unique CtBP2 NLS, mutagenesis data indicates that the Lys 10 and Arg 11 are critical for localization. Interestingly this region is conserved in the Dros0phila CtBP protein. This result is consistent with reports that describe CtBP2 as a nuclear protein (Chinnadurai, 2006). When cotransfected with other CtBP proteins, the NLS of CtBP2 is capable of shuttling the other CtBP proteins into the nucleus by virtue of dimerization. A splice isoform of CtBP2 that lacks this NLS (CtBP2-S) localizes to the cytoplasm. In contrast, a parallel study suggests that instead of functioning as a classical NLS, the Lysine 10 residue might be a site for acetylation of CtBP by p300 that directs it to the nucleus. Using a non- acetylatable version of K10R, this study demonstrated that this acetylation is required for nuclear retention of CtBP2 (Zhao et al., 2006). Further studies are 19 needed to distinguish whether the lysine 10 residue in CtBP2 plays a role in nuclear import or nuclear retention. This mode of regulation of CtBP proteins might have important ramifications on their roles in transcription, development and cellular differentiation. 1.4 Mechanism of CtBP mediated transcriptional repression CtBP is recruited to target promoters by DNA-binding repressors, but details of events that follow are unknown. The precise nature of CtBP’s mode of repression is a subject of intense investigation. Human CtBP1 associates with several HDACs and histone deacetylation might be a major contributor to repression (Sundqvist et al., 1998). However, CtBP mediated repression has been reported to be insensitive to the Class I HDAC inhibitor, Trichostatin A, signifying that HDAC-independent mechanisms might be at play (Ryu and Arnosti, 2003). A first step was taken at identifying these cofactors with the biochemical purification of vertebrate CtBP1 in HeLa cells. The associated proteins were: DNA binding proteins like ZEB1, histone modifiers like histone deacetylases and methyltransferases (HDACs 1 and 2, related HMTs like EuHMT1 and 69a), chromodomain containing protein like HPC2 (human polycomb), Co-REST and related proteins (corepressor for REST transcription factor) and NPAO (nuclear polyamine oxidase). CtBP2 was also found in the same complex, supporting the notion that CtBP proteins can heterodimerize. This core CtBP complex could efficiently cause H3K9 and H3K27 methylation of core histones, both of which are marks of repression. These modifications 20 were also enriched at a CtBP target, E-cadhen‘n. siRNA targeting CtBP not only reduced occupancy of CtBP and EuHMT1 on the E-cad promoter but also decreased methylation of H3 (Shi et al., 2003). Studies on the enzymatic activity of NPAO led to the identification and characterization of the histone demethylase, LSD1 (lysine specific demethylase) (Shi et al., 2005). LSD1 specifically demethylates H3K4 (a mark of active chromatin) via an amine oxidation reaction (Shi et al., 2004). Its presence in the CtBP complex suggests that CtBP proteins co-ordinate distinct enzymatic events that convert active chromatin to a repressed state. Another recently characterized mechanism of CtBP repression includes physical interactions with HAT coactivators like p300 and CBP that contain PXDLS motifs. CtBP was shown to specifically impair the HAT function of these proteins by suppressing H3 acetylation (K9, K14 and K18) in an NADH dependent manner (Kim et al., 2005; Senyuk et al., 2005). The association of CtBP with both coactivators and corepressors of transcription suggests that diverse CtBP complexes with distinct transcriptional properties might exist in vivo. In humans, CtBP also interacts with Cth (C-terminal Interacting Protein). Cth bridges CtBP to Retinoblastoma protein, human Polycomb and BRCA1 (breast cancer tumor suppressor) (Li et al., 1999; Meloni et al., 1999; Sewalt et al., 1999). CtBP proteins also interact with the Polycomb protein (Pc) in Xenopus (Sewalt et al., 1999). A functional role for this interaction has already been discussed with regard to the localization of CtBP proteins by SUMOylation. The Polycomb complex isolated from Drosophila does not appear to contain CtBP and a Cth-like protein is yet to be identified (Levine et 21 al., 2002). These observations postulate that there might be key differences in the complexes formed by human CtBP1 vs. Drosophila CtBP. CtBP proteins may act mostly as bridging molecules to recruit the above-described activities to the DNA binding repressor. As discussed above, CtBP itself has an inherent dehydrogenase activity that might partake in repression, but this remains unproven. Dehydrogenases have not yet been documented to possess repressive activities, however an Oct-1 transcription complex was recently found to utilize a dehydrogenase enzyme for the activation of the histone H2B promoter (Zheng et al., 2003). 1.5 A distinct cytosolic role ascribed to CtBP In a study of factors that regulate membrane tubulation, CtBP3 was identified as a protein that is ribosylated in rat kidney cells upon treatment with the fungal toxin Brefeldin A (Nardini et al., 2003; Spano et al., 1999). CtBP3 displays 97% identity to human and mouse CtBP1-L and 79% identity to CtBP2. CtBP3 lacks the first 11 aa encoded by CtBP1; whether this protein represents the rat version of CtBP1 or an N-terminal splice variant of CtBP1 is not certain. The current consensus in the field is that CtBP3 represents. an N- terminally truncated version of CtBP1. This protein is termed CtBP1-S or BARS (Brefeldin-A ribosylated substrate). Recombinant BARS has been shown to possess a weak acyl transferase enzymatic activity that catalyzes acylation of lysophosphatidic acid to phosphatidic acid (a lipid involved in membrane fissioning) (Weigert et al., 1999). This coincides with the onset of 22 mitosis that requires organelle partitioning between daughter cells. This activity of BARS is thought to lead to accumulation of phosphatidic acid in the Golgi membrane, enhancing the curvature of the phospholipid bilayer and thereby causing disassembly of the Golgi network (Corda et al., 2006). To test the role of BARS in this process, BARS activity was reduced in rat kidney cells using dominant negative mutants or by mRNA depletion using antisense oligos. These treatments prevented Golgi partitioning and arrested the cell cycle at 62. Both Golgi fragmentation and the onset of cell cycle were restored upon addition of recombinant BARS (Hidalgo Carcedo et al., 2004). In support of this data, another study found that in mitotic cells, human CtBP becomes associated with centrosomes during mitosis. Golgi morphogenesis is initiated and regulated within centrosomes, implicating CtBP in this process (Spyer and Allday, 2006). A more recent study re- evaluated the acyl transferase activity associated with BARS and claimed it to be a contaminant associated with the recombinant BARS protein (Gallop et al., 2005). lntriguingly, dual Ctbp1 and Ctbp2 mouse knockouts are embryonic lethal but no Golgi defects are observed and MEFs can be cultured from these animals with no visible Golgi defects. Clearly, much remains to be proven concerning the role of CtBP in Golgi fission. 23 1.6 CtBP as a corepressor during Drosophila development The Drosophila blastoderrn embryo serves as a premier model for the study of eukaryotic transcriptional regulation, providing an amenable background for the study of both cis- and trans-regulatory elements. Early embryonic development in Drosophila is controlled by a cascade of genes that encode for transcription factors. The cascade is turned on by preformed mRNAs and proteins that are synthesized by the mother and deposited in the egg prior to fertilization. The maternally derived proteins set up spatially distributed morphogenetic gradients that establish the antero-posterior (A-P) and dorso-ventral axes. These morphogen proteins can then activate or repress zygotic genes at specific positions along both axes and thereby specify patterning along the embryo (St Johnston and Nusslein-Volhard, 1992). Gap genes are the first category of zygotically transcribed genes, expressed in broad expression domains. They were identified based on phenotypes in a genetic screen looking for mutants lacking large sections of abdominal segments along the A-P axis (Nusslein-Volhard and Wieschaus, 1980). Gap genes encode well-characterized repressor proteins whose primary function is to refine the patterns of pair-rule genes, the next class of genes in the segmentation cascade. Their repression activity is described as short-range, acting at distances of up to 100-150 bp to inhibit the basal promoter or upstream bound activators (Gray and Levine, 1996; Nibu et al., 2003). This is in sharp contrast to long-range repressors that globally suppress transcription over several kilobases (Cai et al., 1996). Short-range 24 repressors employ CtBP as a corepressor, while long-range repressors use the Groucho corepressor. It has been long believed that Groucho and CtBP represent two distinct pathways of repression that form the basis for this so called ‘range' of repressor activity. Pair-rule genes are expressed in seven transverse stripes along the anterior-posterior axis and in turn, they control the fourteen-stripe pattern of the subsequently expressed segment polarity genes that delineate the future segments of the fly. Identification of these zygotic gene products involved in body plan determination of a Drosophila embryo is the outcome of large genetic screens that searched for embryonic lethal mutations. CtBP plays a vital role in this hierarchical organization by contributing to the repression abilities of short-range repressors encoded by gap genes. The first thread of evidence linking CtBP to short-range repression came from yeast two-hybrid assays, in which Knirps and Snail, two short-range repressors were used as bait to screen an embryonic cDNA library (Nibu et al., 1998b). Both proteins selected Drosophila CtBP cDNA, specifying a putative protein of 383 residues and exhibiting high sequence similarity with the human CtBP. The CtBP gene is expressed during early oogenensis and the message is deposited in the egg prior to fertilization. Following zygotic induction, CtBP proteins can be traced throughout development to be ubiquitously and uniformly expressed in the fly (See Figure l-3). It has been demonstrated that CtBP is essential for short-range transcription during development by analyzing embryos lacking maternal CtBP. dCtBP is encoded by a single gene whose locus has been mapped cytologically to 87D8—9 on the third chromosome. P1590 is a homozygous 25 lethal P-element insertion within the CtBP coding region that results in homozygotes dying as pharate adults (i.e. still within pupal case). Mutant embryos containing reduced CtBP levels demonstrate a host of patterning defects that can be attributed to impaired activity of short-range repressor proteins like Knirps, Kriippel and Giant. In situ hybridization assays show that the expression of these repressors is mostly normal in CtBP mutants, but the expression of their pair-rule targets such as even-skipped and fushi-tarazu is severely disturbed (Nibu et al., 1998a). Like vertebrate repressors, short- range repressors in Drosophila recruit CtBP via a PXDLS motif. Mutations in this motif compromise the activity of these proteins, indicating that CtBP- dependent repression represents a major form of transcriptional regulation during development. A direct transcriptional repression activity of dCtBP has also been shown by Gal4-CtBP in a tethering assay wherein CtBP can repress nearby transcriptional activators but not the basal promoter elements (Nibu et al., 1998b; Sutrias-Grau and Arnosti, 2004). This tethering assay has been exploited further to question the relevance of CtBP’s homology to dehydrogenases. Mutations made in the putative catalytic histidine are found not to compromise CtBP corepressor function while mutations in the nucleotide-binding fold destroy its ability to obstruct repression on integrated reporter genes (Sutrias-Grau and Arnosti, 2004). A protein interaction map in Drosophila emphasizes that CtBP forms a major transcriptional regulatory 26 Figure l-3: Drosophila CtBP proteins are ubiquitously distributed during development Stage specific embryos were collected and imaginal discs were harvested from third instar larvae. Samples were fixed in a heptane-formaldehyde solution as per lab protocol and immunostained with anti-CtBP rabbit polyclonal serum (1:500) (F) embryo stained with rabbit preimmune, (B-E) embryos from progressive stages during development, (G-l) wing, eye and leg imaginal discs extracted from late third instar larvae (P.Mani, unpublished). 27 28 network where each interactor contains a discernible P-DLS like motif (Giot et aL,2003) CtBP and long-range repression by Hairy As typified by the Hairy protein in Drosophila, long-range repressors can function over many kilobases to block transcription, in sharp contrast to short-range repressors. The mechanisms underlying these modes of repression are unclear (Courey and Jia, 2001 ). Possible clues have arisen from the observations that short-range repressors mediate repression by the recruitment of CtBP, while long-range repressors recruit a different corepressor, Groucho. These corepressor may mediate repression by distinct pathways (Chen and Courey, 2000). However, both CtBP and Groucho have been proposed to utilize chromatin remodeling mechanisms through the recruitment of histone deacetylases like de3 (Chen et al., 1999; Struffi and Arnosti, 2005). The long-range repressor Hairy also shows a weak interaction with CtBP and has a canonical CtBP binding motif. The removal of the weak dCtBP interaction motif (PLSLV) does not impair Hairy-mediated repression of its targets. Instead, removal of this motif augments Hairy function. CtBP and Groucho binding to Hairy may be antagonistic because just nine amino acid residues separate the CtBP binding P-SLV-K and Groucho binding WRPW motifs. Mechanistically speaking, when dCtBP and Groucho both bind, they might be unable to interact with additional corepressors or contact their target proteins in the core transcription complex (Zhang and Levine, 1999). Such antagonism is supported by genetic studies, which suggest that lowering the 29 dose of maternal dCtBP products can partially suppress the embryonic phenotypes of hairy mutants while reducing groucho can enhance it (Phippen et al., 2000; Poortinga et al., 1998). The simplest interpretation of these observations is that the dCtBP and Groucho interfere with one another when both are bound to Hairy, but the rules governing the choice of cofactor recruited are undetermined. Another cofactor of Hairy is the Sir2 protein that encodes an NAD+ -dependent histone deacetylase. A global chromatin profiling study looking at genomic recruitment of Hairy and its cofactors demonstrates that while Groucho is believed to be the major cofactor, it is found to be associated with only subsets of Hairy targets. Surprisingly, CtBP and Sir2 are predominantly found at Hairy targets with their association being largely overlapping and distinct from Groucho (Bianchi-Frias et al., 2004). CtBP as a corepressor for signaling pathways in the fly Numerous signaling pathways are seen to converge upon general transcription coregulators. Decapentaplegic (dpp) encodes a TGF-fl homolog that functions as a long-range signaling morphogen, specifying cell fates, in a dose-dependent manner. A response to thresholds of Dpp proteins is regulated by an opposing gradient of Brinker, which is a dpp-target and a transcriptional repressor. Brinker’s repression domain contains binding sites for both CtBP and Groucho corepressors and physical interactions have been established. For Brinker to silence its endogenous targets, Groucho alone is 30 sufficient. For simpler targets such as Brinker autoregulation, both corepressors can function interchangeably and adequately. Like Hairy, in the absence of Groucho, Brinker’s repressive capacity is impaired and unlike Hairy, CtBP’s absence also seems to affect Brinker output to a smaller extent (Hasson et al., 2001). These results suggest that Brinker uses multiple modes of repression (provided by CtBP and Groucho) that provide it with a flexibility to silence a variety of genes in response to a gradation in Dpp activity during development. The Notch signaling pathway is employed in a variety of cell fate decisions during development. The Notch signal is transduced via Su(H) [Suppressor of Hairless] that acts as a repressor in the absence of Notch signal and repression is critical for appropriate cell fate specification. In the presence of Notch signaling, Su(H) is an activator. Hairless (H) acts as a corepressor for Su(H) by antagonizing its activation via the recruitment of CtBP and Groucho. Reduction in levels of either CtBP or Groucho enhances Hairless loss of function phenotypes, and mutating Hairless’s CtBP binding motif debilitates its activity, suggesting that both corepressors are vital components of this signaling pathway (Barolo and Posakony, 2002; Barolo et al., 2002). Regulation of the Wnt/ ,B-catenin signaling cascade is also utilized in cell fate determination and oncogenesis. Constitutive induction of this pathway is observed in many cancers, especially in colorectal carcinoma. In the absence of Wnt signal, TCFs (T-cell factor) function as repressors of Wnt target genes, similar to the functionings of the Notch pathway. In Xenopus, thBP is known to act as a corepressor for xTCF-3 (Brannon et al., 1999). In 31 the fly, TCF’s are thought to recruit Groucho for repression and when Wnt signaling is on, binding of B-catenin displaces Groucho and repression is relieved. CtBP can bind to vertebrate TCF’s, and CtBP overexpression inhibits TCF mediated activation of Wnt targets, but no direct interaction of TCF and CtBP has been shown. One model suggests that CtBP antagonizes Wnt pathway by binding to APO tumor suppressor (Adenomatous polyposis coli) that causes it to sequester B-catenin away from TCFs (Hamada and Bienz, 2004). Some support for this model comes from a study where APC and CtBP can be traced on a Wnt target and this coincides with a loss of B-catenin from TCFs (Sierra et al., 2006). As colorectal cancer cells all have mutations in APC proteins, the functional interaction of APC and CtBP suggests a role for CtBP as a tumor suppressor. In contrast, another model suggests CtBP to be recruited by an unknown DNA binding protein and act in parallel to TCF to repress Wnt targets (Fang et al., 2006). CtBP- independent repression activity Multiple repression activities may assist quantitatively or qualitatively in regulating gene expression. Three general mechanisms of transcriptional repression in eukaryotes include competition, where repressors directly compete with activators for a common cis-element, and blocking the basal transcriptional machinery or direct repression. The third is by quenching, whereby repressors repress activators co-occupying adjacent sites. Chromatin modifications that reduce accessibility of transcription factors to the 32 DNA template may involve covalent modifications of histones or DNA (Strahl and Allis, 2000). In addition, cis—acting elements named silencers or boundary elements may prevent transcription of a gene by preventing crosstalk when located between a promoter and enhancer (Gerasimova and Corces, 2001 ). Embryo repression assays performed with short-range repressors Knirps, Giant and Kriippel all provide basis for CtBP-dependent and CtBP- independent modes of repression. Knirps has a repression domain towards the C-terminus that encodes a PXDLS like motif important to bind CtBP. In addition, an N-tenninal region has also been mapped that is active for repression and does not contain any canonical CtBP binding motifs. This activity is evident in experiments in which the CtBP corepressor is absent. In transgenic embryos containing integrated reporters and lacking CtBP, Knirps can still repress its endogenous target, the eve stripe 3 enhancer (Keller et al., 2000) In case of Krilppel, when its binding sites do not overlap but are adjacent to activator binding sites, repression is dependent on CtBP by quenching. Quenching might involve hindering activator-basal machinery crosstalk or it might invoke the intrinsic dehydrogenase activity ascribed to CtBP to remodel chromatin. However, Krinpel can repress by competition without any help from CtBP, when the activator sites overlap with Kriippel repressor binding sites, indicating a CtBP-independent mode of repression (Nibu et al., 2003). Giant is also able to effectively regulate hunchback while its other targets like eve stripe 2 are derepressed in a CtBP mutant background proposing that Giant too can function via CtBP-independent pathways (Strunk et al., 2001). The relevance of these multiple activities was 33 put to test in the case of Knirps. It was found that supplying CtBP-independent activity at higher than regular levels could repress a target that normally requires CtBP, suggesting that CtBP contributes in a quantitative fashion to boost repressor output (Struffi et al., 2004). 1.7 Alternative splicing of Drosophila CtBP In the fly embryo, multiple CtBP transcripts are expressed ubiquitously throughout development that encode polypeptides of 383, 386, 476 and 479 amino acids (See Figure M). The first 376 amino acids required for interaction with short-range repressors are common to all four isoforms (essentially composed of only the dehydrogenase-like domain). Any distinctions in the function of individual isoforms is yet unidentified. These distinct isoforms are produced from alternative splicing that use alternative donor / acceptor sites and differ largely in the presence or absence of a C- ten'ninal tail of ~90 amino acids of unidentified significance. The 476 and 479 aa isoforms both include the C-terminal tail and differ in the alternate use of the splice acceptor that results in the presence/absence of an ‘LNGGYYT’ motif at the start of exon 6 and a ‘VSSQS' motif at the start of exon 7 (Also see Chapter II). Both the 383 and 386 aa isoforms lack the C-terminal extension and differ in the inclusion of a ’VFQ’ motif by resorting to different splice acceptors at the start of exon 5. It is very likely that these different isoforms might function in a tissue or stage specific manner with 34 Figure l-4: Alignment of different CtBP Isoforrns generated by alternative splicing in Drosophila The Drosophila CtBP gene encodes differentially spliced products that are divergent at their COOH-terminal regions (conserved residues are shaded in yellow). Two isoforms [AY060646 (479aa) and NM_001014617 (476 3a)] include an unstructured C-terminal extension of ~90 aa of unknown function and differ in the alternative use of a splice donor that results in the inclusion of ‘LNGGYYT’ motif in the 479 aa isoform and an inclusion of a ‘VSSQS’ motif in the 476 aa isoform. The two shorter isoforms [AY069170 and ABO11840] are essentially composed of just the dehydrogenase-like domains and do not include the C-terrninal tail. These also differ in the use of a splice donor that results in the inclusion of a ‘VFQ' motif in AY069170. The red arrow indicates a conserved splice donor that results in the generation of the longer isoforms. This splice donor represented by a conserved glycine codon within exon 5 that is not subject to silent mutations in several organisms, supporting the notion that isoforms with tails are functionally important. The experiments in Chapter II, III and Appendix A were performed using the 479 and 383 aa isoforms obtained from Yutaka Nibu, Weill Medical College of Cornell University, New York. 35 EEHEEESH§ESH§3§=§>§ a . I . N. :30 gaugHflnEngfi .. .. .. .. .. H3A§§Efl§>§ o coouohd H2BgagmfisgfigmH6>HSHE§HHH6¢2§BH§ 3:32 HoneHggafiggfissaugfigmH6>H§HHHH¢§HHH662§aBHHz 2.2.82 «engage—H6835; ........ asgggfiasgfigaH6>H6~Huagfla662§a85z 7...». £38 aggufiofimfiugflgasfigggmggfigH6>§HHH§HE662§EHE .3322 H 9356-- -zuggggfiggzsa ans—2H HHHLE. :32? Egg 6.8662 .53 3.324 goém Haggis—Hm H:eo§§am 85 8:633 26.. ..HHBG 5223:2263 262 92H .82 :E 5322 3:23 2.22:2 226959.. :2 22H :8 326299662? :H6HBBH H:quHBEwmz.HHb= 6:2 as: 3:5 .HSBHon 6:3 1...: .836 22462.55 655366 :23 agaifig :H6aaaH u§:§§ :2 :0 flgfia H33. 23.. .58 333.2 2HE§>66630>§¢§0§2HE§§Hiya—«562553663366 3.22: 2Engages6326285603226265655HHoEQEnBBHEEHSuogHEo 2.22:2 22225236HHSoSoHSeHHfiuegHu—Sfifignggg HHoEQBEEHEZHSHogHzgo 7...: £30 EH§§§66H¢3§6§¢H§§2§§§§HgguogiHofieaBHfi—Bme $335 HgHHxOHgHgan—MHBHHH HHH§§§ ”Mam §>B§H3Hu§>a gagngdagn—l ONE—Hon“ HgHH “OH gH Haifa: :5 EH” BOO‘ flethGEAHiH :UGMOn—dqd .323 AOENBOEH Haggai a; HgHHu Hon—“Hg :gggnHHggg §:‘ :EHgHmoanna4 :mmdg mOahmegHumfimI-aanl : h. ‘30 HgHH :OH gH :flgggaH H§>§ HHMHOO‘BH‘EEAHSHaUn—MOQAA‘» :3 aOEHIOEH “nag ovoouo: 36 the splicing machinery favoring certain splice sites. CtBPL is generated from splicing originating within exon5 that joins core CtBP sequences (exons 1-5) to that of exon 6 and 7. In this thesis, we study the expression and activities of two of these isoforms that we designate as CtBPL (479 aa) and CtBPs (383 aa) respectively. These two splice variants are found to mediate effective repression on reporter genes when assayed as Gal4 fusion proteins both in the cultured cells and in the embryo (Ryu and Arnosti, 2003; Sutrias-Grau and Amosti, 2004). This prompts an understanding of whether CtBP isoforms in Drosophila are functionally equivalent or have distinct roles in vivo. Chordate genomes like those of human, mouse and Ciona are seen to contain only CtBPL-Iike isoforms (not shown). CtBPs isoforms have only been described thus far in Drosophila, raising significant questions concerning the function of forms of this protein that do not contain the unstructured C-terminal extension. 37 REFERENCES Arnosti, D. N. (2004). Multiple Mechanisms of Transcriptional Repression in Eukaryotes. In Handbook of Experimental Pharmacology, vol. 166 (ed. K. J. a. T. S. J. Gossen M.): Springer(Berlin). Austin, R. J. and Biggin, M. D. (1995). A domain of the even-skipped protein represses transcription by preventing TFIID binding to a promoter: repression by cooperative blocking. Mol Cell Biol 15, 4683-93. Balasubramanian, P., Zhao, L. J. and Chinnadurai, G. (2003). Nicotinamide adenine dinucleotide stimulates oligomerization, interaction with adenovirus E1A and an intrinsic dehydrogenase activity of CtBP. FEBS Lett 537, 157-60. Barnes, C. J., Vadlamudl, R. K., Mishra, S. K., Jacobson, R. H., Li, F. and Kumar, R. (2003). Functional inactivation of a transcriptional corepressor by a signaling kinase. Nat Struct Biol 10, 622-8. Barolo, S. and Posakony, J. W. (2002). Three habits of highly effective signaling pathways: principles of transcriptional control by developmental cell signaling. Genes Dev 16, 1167-81. Barolo, 8., Stone, T., Bang, A. G. and Posakony, J. W. (2002). Default repression and Notch signaling: Hairless acts as an adaptor to recruit the corepressors Groucho and dCtBP to Suppressor of Hairless. Genes Dev 16, 1964-76. Bianchl-Frias, D., Orian, A., Delrow, J. J., Vazquez, J., Rosales-Nieves, A. E. and Parkhurst, S. M. (2004). Hairy transcriptional repression targets and cofactor recruitment in Drosophila. PLoS Biol 2, E178. Boyd, J. M., Subramanian, T., Schaeper, U., La Regina, M., Bayley, S. and Chinnadurai, G. (1993). A region in the C-terminus of adenovirus 2/5 E1a protein is required for association with a cellular phosphoprotein and important for the negative modulation of T24-ras mediated transformation, tumorigenesis and metastasis. Embo J 12, 469-78. Brannon, M., Brown, J. D., Bates, R., Kimelman, D. and Moon, R. T. (1999). XCtBP is a Xch-3 oo-repressor with roles throughout Xenopus development. Development 126, 3159-70. Cal, H. N., Arnosti, D. N. and Levine, M. (1996). Long-range repression in the Drosophila embryo. Proc Natl Acad Sci U S A 93, 9309-14. Chen, G. and Courey, A. J. (2000). Groucho/1' LE family proteins and transcriptional repression. Gene 249, 1-16. 38 Chen, G., Fernandez, J., Mische, S. and Courey, A. J. (1999). A functional interaction between the histone deacetylase de3 and the corepressor groucho in DrOSOphila development. Genes Dev 13, 2218-30. Chinnadurai, G. (2002). CtBP, an unconventional transcriptional corepressor in development and oncogenesis. Mol Cell 9, 213-24. Chinnadurai, G. (2006). CtBP family proteinszunique transcriptional regulators in the nucleus with diverse cytosolic functions: Landes Bioscience. Corda, D., Colanzl, A. and Luinl, A. (2006). The multiple activities of CtBP/BARS proteins: the Golgi view. Trends Cell Biol 16, 167-73. Courey, A. J. and Jia, S. (2001). Transcriptional repression: the long and the short of it. Genes Dev 15, 2786-96. Criqui-Filipe, P., Ducret, C., Maira, S. M. and Wasylyk, B. (1999). Net, a negative Ras-switchable TCF, contains a second inhibition domain, the CID, that mediates repression through interactions with CtBP and de-acetylation. Embo J 18, 3392-403. Deconinck, A. E., Mead, P. E., Tevosian, S. G., Crlsplno, J. D., Katz, S. G., Zon, L. I. and Orkln, S. H. (2000). FOG acts as a repressor of red blood cell development in Xenopus. Development 127, 2031 -40. Deltour, S., Pinte, S., Guerardel, G., Wasylyk, B. and Leprince, D. (2002). The human candidate tumor suppressor gene HIC1 recruits CtBP through a degenerate GLDLSKK motif. Mol Cell Biol 22, 4890-901. Fang, M., Li, J., Blauwkamp, T., Bhambhani, C., Campbell, N. and Cadlgan, K. M. (2006). C-terrninal-binding protein directly activates and represses Wnt transcriptional targets in Drosophila. Embo J 25, 2735-45. Field, C. C., Birdsong, W. T. and Goodman, R. H. (2003). Differential binding of NAD+ and NADH allows the transcriptional corepressor carboxyl- terminal binding protein to serve as a metabolic sensor. Proc Natl Acad Sci U S A 100, 9202-7. Furusawa, T., Moribe, H., Kondoh, H. and Higashl, Y. (1999). Identification of CtBP1 and CtBP2 as corepressors of zinc finger-homeodomain factor deltaEF1. Mol Cell Biol 19, 8581-90. Gallop, J. L., Butler, P. J. and McMahon, H. T. (2005). Endophilin and CtBP/BARS are not acyl transferases in endocytosis or Golgi fission. Nature 438, 675-8. Garrlga-Canut, M., Schoenlke, B., Qazl, R., Bergendahl, K., Daley, T. J., Pfender, R. M., Morrison, J. F., Ockuly, J., Stafstrom, C., Sutula, T. et al. (2006). 2-Deoxy-D-glucose reduces epilepsy progression by NRSF-CtBP- dependent metabolic regulation of chromatin structure. Nat Neurosci. 39 Gerasimova, T. I. and Corces, V. G. (2001). Chromatin insulators and boundaries: effects on transcription and nuclear organization. Annu Rev Genet 35, 193-208. Glot, L., Bader, J. S., Brouwer, C., Chaudhurl, A., Kuang, 8., Li, Y., Hao, Y. L., Ool, C. E., Godwin, B., Vltols, E. et al. (2003). A protein interaction map of Drosophila melanogaster. Science 302, 1727-36. Gray, S. and Levine, M. (1996). Transcriptional repression in development. Curr Opin Cell Biol 8, 358-64. Grooteclaes, M., Deveraux, G., Hildebrand, J., Zhang, 0., Goodman, R. H. and Frlsch, S. M. (2003). C-terminal-binding protein corepresses epithelial and proapoptotic gene expression programs. Proc Natl Acad Sci U S A 100, 4568-73. Hamada, F. and Bienz, M. (2004). The APC tumor suppressor binds to C- terrninal binding protein to divert nuclear beta-catenin from TCF. Dev Cell 7, 677-85. Hasson, P., Muller, B., Basler, K. and Paroush, Z. (2001). Brinker requires two corepressors for maximal and versatile repression in Dpp signalling. Embo J 20. 5725-36. Hldalgo Carcedo, C., Bonazzi, M., Spano, S., Turacchlo, G., Colanzl, A., Luinl, A. and Corda, D. (2004). Mitotic Golgi partitioning is driven by the membrane-fissioning protein CtBP3/BARS. Science 305, 93-6. Hildebrand, J. D. and Soriano, P. (2002). Overlapping and unique roles for C-terrninal binding protein 1 (CtBP1) and CtBP2 during mouse development. Mol Cell Biol 22, 5296-307. Imal, 8., Armstrong, C. M., Kaeberlein, M. and Guarente, L. (2000). Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 403, 795-800. Katsanls, N. and Fisher, E. M. (1998). A novel C-terrninal binding protein (CTBP2) is closely related to CTBP1, an adenovirus E1A-binding protein, and maps to human chromosome 21q21.3. Genomics 47, 294-9. Keller, S. A., Mao, Y., Struffi, P., Margulies, C., Yurk, C. E., Anderson, A. R., Amey, R. L., Moore, 8., Ebels, J. M., Foley, K. et al. (2000). dCtBP- dependent and -independent repression activities of the Drosophila Knirps protein. Mol Cell Biol 20, 7247-58. Kim, J. H., Cho, E. J., Kim, S. T. and Youn, H. D. (2005). CtBP represses p300-mediated transcriptional activation by direct association with its bromodomain. Nat Struct Mol Biol 12, 423-8. Koipally, J. and Georgopoulos, K. (2000). lkaros interactions with CtBP reveal a repression mechanism that is independent of histone deacetylase activity. J Biol Chem 275, 19594-602. 40 Kumar, V., Carlson, J. E., Ohgl, K. A., Edwards, T. A., Rose, D. W., Escalante, C. R., Rosenfeld, M. G. and Aggarwal, A. K. (2002). Transcription corepressor CtBP is an NAD(+)-regulated dehydrogenase. Mol Cell 10, 857-69. Lawrence, P. A. (1992). The Making of a Fly: Blackwell Sci. Pub. (Oxford). Levine, S. 8., Weiss, A., Erdjument-Bromage, H., Shao, Z., Tempst, P. and Kingston, R. E. (2002). The core of the polycomb repressive complex is compositionally and functionally conserved in flies and humans. Mol Cell Biol 22, 6070-8. Li, S., Chen, P. L., Subramanian, T., Chinnadurai, G., Tomlinson, G., Osborne, C. K., Sharp, 2. D. and Lee, W. H. (1999). Binding of Cth to the BRCT repeats of BRCA1 involved in the transcription regulation of p21 is disrupted upon DNA damage. J Biol Chem 274, 11334-8. Lin, X., Sun, 8., Liang, M., Liang, Y. Y., Gast, A., Hlldebrand, J., Brunlcardi, F. C., Melchior, F. and Feng, X. H. (2003). Opposed regulation of corepressor CtBP by SUMOylation and PDZ binding. Mol Cal/11, 1389-96. Long, J., Zuo, D. and Park, M. (2005). Pc2-mediated sumoylation of Smad- interacting protein 1 attenuates transcriptional repression of E-cadherin. J Biol Chem 280, 35477-89. Lunyak, V. V., Burgess, R., Prefontaine, G. G., Nelson, C., Sze, S. H., Chenoweth, J., Schwartz, P., Pevzner, P. A., Glass, C., Mandel, G. et al. (2002). Corepressor-dependent silencing of chromosomal regions encoding neuronal genes. Science 298, 1747-52. Mannervik, M., Nibu, Y., Zhang, H. and Levine, M. (1999). Transcriptional coregulators in development. Science 284, 606-9. Melhulsh, T. A. and Wotton, D. (2000). The interaction of the carboxyl terminus-binding protein with the Smad corepressor TGIF is disrupted by a holoprosencephaly mutation in TGIF. J Biol Chem 275, 39762-6. Meloni, A. R., Smith, E. J. and Nevins, J. R. (1999). A mechanism for Rb/p130-mediated transcription repression involving recruitment of the CtBP corepressor. Proc Natl Acad Sci U S A 96, 9574-9. Molloy, D., Mapp, K. L., Webster, R., Galllmore, P. H. and Grand, R. J. (2006). Acetylation at a lysine residue adjacent to the CtBP binding motif within adenovirus 12 E1A causes structural disruption and limited reduction of CtBP binding. Virology. Molloy, D. P., Barral, P. M., Bremner, K. H., Galllmore, P. H. and Grand, R. J. (2001). Structural determinants outside the PXDLS sequence affect the interaction of adenovirus E1A, C-terrninal interacting protein and Drosophila repressors with C-terminal binding protein. Biochim Biophys Acta 1546, 55- 70. 41 Nardini, M., Spano, S., Cericola, C., Pesce, A., Massaro, A., Millo, E., Luinl, A., Corda, D. and Bolognesl, M. (2003). CtBP/BARS: a dual-function protein involved in transcription co-repression and Golgi membrane fission. Embo J 22, 3122-30. Nardini, M., Svergun, D., Konarev, P. V., Spano, S., Fasano, M., Bracco, C., Pesce, A., Donadini, A., Cericola, C., Secundo, F. et al. (2006). The C- terrninal domain of the transcriptional corepressor CtBP is intrinsically unstructured. Protein Sci 15, 1042-50. ' Nibu, Y., Senger, K. and Levine, M. (2003). CtBP-independent repression in the Drosophila embryo. Mol Cell Biol 23, 3990-9. Nibu, Y., Zhang, H., Bajor, E., Barolo, 8., Small, S. and Levine, M. (19983). dCtBP mediates transcriptional repression by Knirps, Kruppel and Snail in the Drosophila embryo. Embo J 17, 7009-20. Nibu, Y., Zhang, H. and Levine, M. (1998b). Interaction of short-range repressors with Drosophila CtBP in the embryo. Science 280, 101-4. Nusslein-Volhard, C. and Wieschaus, E. (1980). Mutations affecting segment number and polarity in Drosophila. Nature 287, 795-801. Palmer, 5., Broulllet, J. P., Kilbey, A., Fulton, R., Walker, M., Crossley, M. and Bartholomew, C. (2001). Evi-1 transforming and repressor activities are mediated by CtBP co-repressor proteins. J Biol Chem 276, 25834-40. Perdomo, J. and Crossley, M. (2002). The lkaros family protein Eos associates with C-terminal-binding protein corepressors. Eur J Biochem 269, 5885-92. Phippen, T. M., Swelgart, A. L., Moniwa, M., Krumm, A., Davie, J. R. and Parkhurst, S. M. (2000). Drosophila C-terminal binding protein functions as a context-dependent transcriptional co-factor and interferes with both mad and groucho transcriptional repression. J Biol Chem 275, 37628-37. Poortinga, G., Watanabe, M. and Parkhurst, S. M. (1998). Drosophila CtBP: a Hairy-interacting protein required for embryonic segmentation and hairy- mediated transcriptional repression. Embo J 17, 2067-78. Quinlan, K. G., Verger, A., Kwok, A., Lee, S. H., Perdomo, J., Nardini, M., Bolognesl, M. and Crossley, M. (2006). The role of the C-terminal binding protein PXDLS motif binding cleft in protein interactions and transcriptional repression. Mol Cell Biol. Rutter, J., Relck, M., Wu, L. C. and McKnight, S. L. (2001). Regulation of clock and NPASZ DNA binding by the redox state of NAD cofactors. Science 293, 510-4. Ryu, J. R. and Arnosti, D. N. (2003). Functional similarity of Knirps CtBP- dependent and CtBP-independent transcriptional repressor activities. Nucleic Acids Res 31, 4654-62. 42 Schaeper, U., Boyd, J. M., Verma, S., Uhlmann, E., Subramanian, T. and Chinnadurai, G. (1995). Molecular cloning and characterization of a cellular phosphoprotein that interacts with a conserved C-terminal domain of adenovirus E1A involved in negative modulation of oncogenic transformation. Proc Natl Acad Sci U S A 92, 10467-71. Schmitz, F., Konlgstorfer, A. and Sudhof, T. C. (2000). RIBEYE, a component of synaptic ribbons: a protein's journey through evolution provides insight into synaptic ribbon function. Neuron 28, 857-72. Senyuk, V., Sinha, K. K. and Nucifora, G. (2005). Corepressor CtBP1 interacts with and specifically inhibits CBP activity. Arch Biochem Biophys 441, 168-73. Sewalt, R. G., Gunster, M. J., van der Vlag, J., Satijn, D. P. and Otte, A. P. (1999). C-Terminal binding protein is a transcriptional repressor that interacts with a specific class of vertebrate Polycomb proteins. Mol Cell Biol 19, 777- 87. Shl, Y., Lan, F., Matson, C., Mulligan, P., Whetstlne, J. R., Cole, P. A., Casero, R. A. and Shl, Y. (2004). Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cal/119, 941-53. Shl, Y., Sawada, J., Sui, G., Affar el, 8., Whetstlne, J. R., Lan, F., Ogawa, H., Luke, M. P., Nakatani, Y. and Shl, Y. (2003). Coordinated histone modifications mediated by a CtBP co-repressor complex. Nature 422, 735-8. Shl, Y. J., Matson, C., Lan, F., lwase, S., Baba, T. and Shl, Y. (2005). Regulation of LSD1 histone demethylase activity by its associated factors. Mol Cell 19, 857-64. Sierra, J., Yoshida, T., Joazeiro, C. A. and Jones, K. A. (2006). The APC tumor suppressor counteracts beta-catenin activation and H3K4 methylation at Wnt target genes. Genes Dev 20. 586-600. Sollerbrant, K., Chinnadurai, G. and Svensson, C. (1996). The CtBP binding domain in the adenovirus E1A protein controls CR1-dependent transactivation. Nucleic Acids Res 24, 2578-84. Spano, S., Silletta, M. G., Colanzl, A., Alberti, S., Fiuccl, G., Valente, C., Fusella, A., Salmona, M., Mironov, A., Luinl, A. et al. (1999). Molecular cloning and functional characterization of brefeldin A-ADP-ribosylated substrate. A novel protein involved in the maintenance of the Golgi structure. J Biol Chem 274, 17705-10. Spyer, M. and Allday, M. J. (2006). The transcriptional co-repressor C- terrninal binding protein (CtBP) associates with centrosomes during mitosis. Cell Cycle 5, 530-7. St Johnston, D. and Nussleln-Volhard, C. (1992). The origin of pattern and polarity in the Drosophila embryo. Cell 68, 201-19. 43 Strahl, B. D. and Allis, C. D. (2000). The language of covalent histone modifications. Nature 403, 41-5. Struffl, P. and Arnosti, D. N. (2005). Functional interaction between the Drosophila knirps short range transcriptional repressor and RPD3 histone deacetylase. J Biol Chem 280, 40757-65. Struffl, P., Corado, M., Kulkarni, M. and Arnosti, D. N. (2004). Quantitative contributions of CtBP-dependent and -independent repression activities of Knirps. Development 131, 2419-29. Strunk, B., Struffl, P., Wright, K., Pabst, B., Thomas, J., an, L. and Arnosti, D. N. (2001). Role of CtBP in transcriptional repression by the Drosophila giant protein. Dev Biol 239, 229-40. Sundqvist, A., Sollerbrant, K. and Svensson, C. (1998). The carboxy- terrninal region of adenovirus E1A activates transcription through targeting of a C-terrninal binding protein-histone deacetylase complex. FEBS Lett 429, 183-8. Sutrias-Grau, M. and Arnosti, D. N. (2004). CtBP contributes quantitatively to Knirps repression activity in an NAD binding-dependent manner. Mol Cell Biol 24, 5953-66. Turner, J. and Crossley, M. (2001). The CtBP family: enigmatic and enzymatic transcriptional co-repressors. Bioessays 23, 683-90. Van Hateren, N., Shenton, T. and Borycki, A. G. (2006). Expression of avian C-terminal binding proteins (Ctbp1 and Ctbp2) during embryonic development. Dev Dyn 235, 490-5. van Vliet, J., Turner, J. and Crossley, M. (2000). Human Kruppel-like factor 8: a CACCC-box binding protein that associates with CtBP and represses transcription. Nucleic Acids Res 28, 1955-62. Verger, A., Quinlan, K. G., Crofts, L. A., Spano, S., Corda, D., Kable, E. P., Braet, F. and Crossley, M. (2006). Mechanisms directing the nuclear localization of the CtBP family proteins. Mol Cell Biol 26, 4882-94. Vo, N., Field, C. and Goodman, R. H. (2001). Acetylation of nuclear hormone receptor-interacting protein RlP140 regulates binding of the transcriptional corepressor CtBP. Mol Cell Biol 21, 6181-8. Wang, S. Y., Iordanov, M. and Zhang, Q. (2006). c—Jun NH2-terminal kinase promotes apoptosis by down-regulating the transcriptional co-repressor CtBP. J Biol Chem. Welgert, R., Silletta, M. G., Spano, S., Turacchlo, G., Cericola, C., Colanzl, A., Senatore, S., Mancini, R., Polishchuk, E. V., Salmona, M. et al. (1999). CtBP/BARS induces fission of Golgi membranes by acylating lysophosphatidic acid. Nature 402, 429-33. 44 Wray, G. A. (2003). Transcriptional regulation and the evolution of development. Int J Dev Biol 47, 675-84. Zhang, H. and Levine, M. (1999). Groucho and dCtBP mediate separate pathways of transcriptional repression in the Drosophila embryo. Proc Natl Acad Sci U S A 96, 535-40. Zhang, G., Nottke, A. and Goodman, R. H. (2005). Homeodomain- interacting protein kinase-2 mediates CtBP phosphorylation and degradation in UV-triggered apoptosis. Proc Natl Acad Sci U S A 102, 2802-7. Zhang, Q., Piston, D. W. and Goodman, R. H. (2002). Regulation of corepressor function by nuclear NADH. Science 295, 1895-7. Zhao, L. J., Subramanian, T., Zhou, Y. and Chinnadurai, G. (2006). Acetylation by p300 regulates nuclear localization and function of the transcriptional corepressor CtBP2. J Biol Chem 281, 4183-9. Zheng, L., Roeder, R. G. and Luo, Y. (2003). S phase activation of the histone H28 promoter by OCA-S, a coactivator complex that contains GAPDH as a key component. Cell 114, 255-66. 45 CHAPTER II Developmental Expression and Phylogenetic Conservation of Alternatively Spliced Forms of the C-termlnal Binding Protein Corepressor Mani-Telang, P. and Amosti D. N. (2006). Development, Genes and Evolution In Press 46 ABSTRACT The C-terrninal binding protein (CtBP) is an evolutionarily conserved transcriptional corepressor found in multicellular eukaryotes. Multiple forms of the protein are typically found in animal cells, produced from separate genes and by alternative splicing. CtBP isoforms have also been implicated in cytoplasmic functions, including Golgi fission and vesicular trafficking. All forms of CtBP contain a conserved core domain that is homologous to a- hydroxyacid dehydrogenases, and a subset of isoforms (CtBPL) contain extensions at the C-terminus. Despite distinct developmental profiles and knockout phenotypes in the mouse, the properties of different isoforms of the protein are found to be similar in many transcriptional assays. We have investigated the expression and conservation of distinct isoforms of the CtBP protein in insects, and found that the expression of multiple, developmentally regulated isoforms is widely conserved. In a variety of Drosophila species, the relative abundance of CtBPL to CtBPs drops sharply after embryogenesis. revealing a conserved developmental shift. Despite the overall lower levels of this isoform, bioinforrnatic analysis reveals that exons encoding the C-terminal extension in CtBPL are conserved from Diptera to Coleoptera, suggesting that the CtBPL isoform contributes an important, evolutionarily conserved function. 47 Introduction The C-terminal binding protein (CtBP) is an evolutionarily conserved factor that has been implicated in a variety of cellular processes, including transcriptional repression, Golgi function, and vertebrate retinal synapse activity (Chinnadurai, 2003). Originally identified by its ability to interact with the C-terminus of the adenovirus E1A protein, CtBP has been shown to directly bind to a variety of transcription factors in vertebrate cells and in Drosophila, and recruit chromatin-modifying factors including histone deacetylases and histone demethylases (reviewed in Turner and Crossley, 2001; Chinnadurai, 2005). CtBP proteins share a high degree of similarity within a central domain comprised of an NAD-binding domain and a substrate-binding fold. The proteins form dimers, and demonstrate extensive structural similarity to NAD-dependent dehydrogenases (Kumar et al., 2002; Nardini et al., 2003). CtBP proteins also possess C-terminal sequences of variable lengths that are likely to be unstructured (Nardini et al., 2006). CtBP proteins exhibit a weak NAD-dependent catalytic activity in vitro, however the physiological relevance of this activity is unknown (Kumar et al., 2002; Balasubramanian et al., 2003; Barnes et al., 2003). NAD binding has also been suggested to regulate CtBP allostery, permitting the interaction of the protein with binding partners. In vitro, association of CtBP with E1A proteins is stimulated by NAD and NADH, suggesting a possible molecular switch that might regulate CtBP activity (Kumar et al., 2002; Barnes et al., 2003). Differential affinity of the protein for NADH relative to NAD has been suggested to endow CtBP with the potential to respond to differing 48 intracellular levels of these cofactors, possibly linking gene regulation to cellular redox states (Zhang et al., 2002). A possible lysophosphatidic acid acyl transferase activity relevant to membrane trafficking has also been ascribed to one form of CtBP (CtBP3/CtBP1-S/BARS), however this result has been disputed (Weigert et al., 1999; Gallop et al., 2005). Distinct CtBP isoforms are expressed as a result of alternative splicing, alternative promoter usage, and different genes. In vertebrates, the ctbp1 and ctbp2 genes are expressed in overlapping patterns during development and exhibit distinct functions. ctbp1 knockout mice are viable, but are smaller and show increased postnatal mortality, while the ctbp2 mutation is embryonic lethal (Hildebrand and Soriano, 2002). Similar to the expression pattern in mice, avian orthologs of Ctbp1 and Ctbp2 are expressed in overlapping regions and distinct domains suggesting that Ctbp1 and Ctbp2 might have unique roles in certain tissues (Van Hateren et al., 2006). The rat CtBP1 isoform termed CtBP1-S/CtBP3/BARS lacks a short region at the N terminus; this protein has been implicated in membrane fission events that are required for Golgi trafficking and Golgi fragmentation during mitosis (reviewed in Corda et al., 2006). In vertebrates, the RIBEYE splice form of CtBP2 produces a protein containing CtBP residues fused to a unique N terminus; this protein is localized to synaptic vesicles of the retina (Schmitz et al., 2000). , It is unknown whether the distinct developmental phenotypes seen with vertebrate CtBP mutants are largely a reflection of the genes' unique promoter activities, or whether differences in the proteins themselves play a large role. Posttranslational modifications and association with other binding proteins have been shown to regulate the stability, activity and localization of 49 CtBP proteins in vertebrates. Some of these modifications target the central conserved region of the protein; the Pak1 kinase phosphorylates CtBP1 at Ser158, stimulating nuclear to cytoplasmic translocation and downregulating CtBP repression activity (Barnes et al., 2003). Other modifications are targeted to the C-terminal nonconserved portion of the protein; phosphorylation of CtBP1 ser422 by the HIPK2 kinase promotes degradation of the protein, whereas SUMOylation of the C-terminus is required for nuclear localization of CtBP1 (Kagey et al., 2003; Lin et al., 2003; Zhang et al., 2005). In addition to being covalently modified, the C-terminus can also serve as the binding target for a PDZ-domain containing protein, neuronal nitric oxide synthase, that drives cytoplasmic localization of the CtBP1 (Riefler and Firestein, 2001). Drosophila CtBP is vital for embryonic development and embryos lacking dCtBP function exhibit grave defects in segmentation. In contrast to vertebrates, distinct Drosophila CtBP proteins are produced from a single gene. Two major isoforms, termed CtBPL and CtBPs, differ by the presence or absence of a ~90 amino acid extension at the C-terminus, which, although of similar size and amino acid composition, is not homologous to C-terminal extensions found in vertebrate CtBP proteins (Poortinga et al., 1998; Nibu et al., 1998a). In light of the fact that the unstructured C-terminus can play a regulatory role in vertebrates, it seems possible that Drosophila CtBPL may be subject to similar covalent modifications as those found in vertebrates, but currently there is little understanding of the biological importance of the different isoforms. In vitro, both CtBPL and CtBPs are able to bind to short- range transcriptional repressors such as Knirps and Kriippel, and in 50 transcriptional assays, both isoforms exhibit similar activities (Sutrias-Grau and Arnosti, 2004; Fang et al., 2006). Therefore, we have utilized biochemical and phylogenetic analysis to study expression of the protein in disparate orders to gain more insight into the significance of distinct isoforms of this widely conserved protein. Biochemical and phylogenetic evidence indicates that the alternatively spliced CtBPL isoform represents a conserved, developmentally regulated form of the protein, suggesting a specific functional role for this protein. 51 MATERIALS AND METHODS Insect stocks and Iysate preparation The fly stocks used in this study were: Drosophila melanogaster yW" (Amosti lab), Drosophila sechellia, Drosophila moja vensis (Tucson Drosophila Stock Center), Drosophila virilis (Dr. Scott Pitnick). Tn'bolium castaneum was a gift from Dr. Susan Brown (Kansas State University), Anopheles gambiae from Dr. Ned Walker (Michigan State University) and Apis mellifera from Dr. Zachary Huang (Michigan State University). All files were maintained on standard cornmeal/molasses food and embryos collected at 25°C on apple juice/agar. For developmental expression analysis, staged embryos were collected, dechorionated and resuspended in lysis buffer (150mM NaCl, 50mM Hepes, pH 7.9, 10% glycerol, 0.1mM EDTA with Complete mini-EDTA free protease inhibitor cocktail tablet, Roche) and sonicated using a Branson-250 sonifier. Larvae, pupae and adults were homogenized in lysis buffer with a steel pestle and then sonicated under the same conditions. Lysates were cleared by centrifugation and total protein concentration of the supernatant was measured by a Bradford assay with bovine serum albumin as the standard. The supernatant was mixed with Laemmli sample buffer for sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SOS-PAGE) analysis. For identification of CtBP isoforms in bee, beetle, and mosquito and flies, whole adult animals were homogenized in Laemmli sample buffer for SOS-PAGE analysis. 52 Western Blot Analysis Immunoblotting was performed using 10% SOS-PAGE gels in a tank transfer system (Bio-Rad Mini Trans-Blot® Cell) and proteins were transferred to lmmuno—BlotTM PVDF membrane (Bio-Rad). Antibody incubation was performed in TBST (20mM Tris-HCI, pH 7.5, 120mM NaCl, 0.1% Tween-20) supplemented with 5% non-fat dry milk as a blocking agent. Rabbit polyclonal antibodies used to detect CtBP (1:10.000) and monoclonal mouse antibody for tubulin (1:6000, Iowa Hybridoma Bank) were visualized using HRP- conjugated secondary antibodies (Pierce) and SuperSignal® West Pico chemiluminiscent substrate (Pierce). Western blots shown are representative of at least three biological replicates for each experiment. Antibodies and recombinant CtBP proteins Polyclonal anti-CtBP antibodies were generated as described in Struffi et al. (2005). For the production of recombinant proteins, the cDNAs for CtBPL and CtBPs bearing two Flag epitope tags at the C-terminal end was cloned into the pET15b expression vector and used to transform E.coli BL-21 cells. Expression of bacterial proteins was induced by treating log-phase cultures with 0.4mM IPTG. The cells were then sonicated in lysis buffer, centrifuged and supernatant was dissolved in Laemmli sample buffer for western analysis. 53 RT-PCR measurements of splice form abundances In embryos and adufls. Total RNA of D. melanogaster embryos (stage 0-12) and adults was isolated by tissue homogenization in Trizol reagent (Sigma) according to the manufacturer's protocol. RNA was treated with Rnase-free DNase (RQ1, Promega) to remove contaminating genomic DNA. RT-PCR was performed using AccessQuickTM RT—PCR System from Promega. Transcripts for CtBPL were amplified using primer pairs DA1 147 — 5' CCCCACAGTACAACCAACCT 3' and DA1 148 - 5' TCCGTTTTTATGCTCGATGA 3', CtBPs using primer pairs DA 1146 — 5' CTCAACGAGCACAACCATCA'ITI’AATC 3' and DA 1150 — 5' CTCTACTTTTCTTGATTTGATATCATTTGTAG 3' and total CtBP was amplified using primer pairs DA 1 146 - 5' CTCAACGAGCACAACCATCATTI’AATC 3' and DA 1151 - 5'GCACGTCTGGAATATTGCCGAC 3’. The transcript for tubulin was amplified using primers BWH 763 - 5' CCGCCACCTTCATCGGCAAC 3' and BWH 764 - 5' TTAGTTCTCGTCGACCACAG 3'. All primer pairs spanned an intron such that amplification of contaminating genomic DNA could be distinguished from the RT-PCR amplified products. The RT step. was performed at 45°C for 45 minutes followed by 30 cycles of PCR in a 25 pl reaction mix for 94°C for 1min, 56°C for 1 min and 72°C for 1 min for CtBP transcripts and 25 cycles for the tubulin transcript. PCR products were visualized by agarose gel electrophoresis and quantitated using BioRad Quantity One software Version 4.4.1. . Three separate RNA preparations for 54 embryos and three for adults were quantitated and a scatter plot was generated in Excel. Linear fits to the data were used to determine relative amounts of CtBPL and CtBPs mRNA for each RNA preparation, and these amounts were compared to levels of total CtBP and to tubulin. Relative levels of CtBPs mRNA increase from embryo to adult by 3-8 fold, when normalized to either total CtBP or tubulin controls, whereas normalized CtBPL mRNA levels do not change significantly. The data shown in Figure “-10 is a representative result of RT-PCR analysis of biological triplicates that were each analyzed at least two times. In this particular dataset, CtBPs was upregulated 6 fold in adults when normalized to both total CtBP and tubulin controls. Sequence alignments To determine the conservation of CtBP exons in diverse insect genomes we searched the Flybase database (Release 4.2) using FLYBASE BLAST for the assembled genomes of Drosophila melanogaster, D. sechellia, D. persimilis, D. mojavensis, D. virilis, D. grimshawi, Anopheles gambiae, Aedes aegypti, Apis mellifera and Tribolium castaneum. Matches to conserved exons 1-4 of CtBP were obtained for D. sechellia (AAKOO1000254.1), D. persimilis (AAIZ01000471), D. mojavensis (contig_8705), D. virilis (contig_15233), D. gn'mshawi (contig_21987), A.gambiae (AAABO1008805), Aedes aegypti (supercontig_1.155), A.mellifera (AADGOSOO6060) and 7'. castaneum (CM000284.1). These automated alignments generally did not identify exons 6 and 7 however, therefore sequences 3’ to the conserved exons were searched in all three reading frames for conserved coding information and 55 aligned using Clustal W. Predicted gene sequences for A. mellifera (XM_392682) and T. castaneum (XP_972241) were included in these alignments. Relative similarities were calculated based on the pairwise alignment with the D. melanogaster peptide sequence for exons 6+7 as a reference. 56 RESULTS Expression of CtBP Isoforrns In Drosophila Four major CtBP transcripts are detected ubiquitously during development and are predicted to produce proteins of 383, 386, 476 and 479 amino acids (Poortinga et al., 1998; Nibu et al., 1998b; Sutrias-Grau and Arnosti, 2004). To analyze endogenous CtBP proteins in Drosophila, we generated polyclonal rabbit antibodies against CtBPL protein (aa1-479) expressed in E.coli. The antibodies are specific and detect proteins of the expected sizes in embryonic extracts, approximately 42 (CtBPs) and 50 KDa (CtBPL ) (Figure lI-1A). lmmunostaining revealed that CtBP proteins are ubiquitously present in the nuclei of pre- and post-blastoderrn embryos and imaginal discs from third Instar larvae (data not shown). To analyze the developmental expression profile of CtBP isoforms, we analyzed soluble extracts from different developmental stages of the fly (Figure ll-1B). Both CtBPL and CtBPs isoforms are detected throughout the first 15 hours of embryogenesis. with relatively higher levels of CtBPs than CtBPL (antibody recognition of CtBPL is expected to be equal or better than that of CtBPs because the two proteins are virtually identical in the central domain, and the antibody was raised against CtBPL). The relative levels of CtBPL to CtBPs drop further after embryogenesis. showing weak expression of CtBPL in the larva, pupa, and adult (Figure II-1B). The lower abundance of CtBPL in postembryonic stages is not simply due to sequestration of the protein in an insoluble form, because similar low levels of CtBPL were observed in whole animal extracts prepared in boiling SDS (discussed below). 57 Figure "-1: Developmental expression profile of CtBP isoforms In Drosophila melanogaster. A. Specificity of anti-CtBP antibody tested in Western blot with Drosophila melanogaster embryonic extract (lanes 1,2) or bacterial extracts containing recombinant CtBPL (lane 3) or CtBPs (lane 4). Preimmune serum did not cross react with any proteins in embryo extract, while anti-CtBP recognized two isoforms of approximately 42 and 50 kDa in embryonic extracts. Recombinant proteins migrate slower than endogenous counterparts due the presence of an N-terminal hexahistidine tag and a C-terminal Flag tag. Markers (kDa) are indicated to the left. B. Expression of CtBP isoforms in embryos, larvae, pupae, and adults. 50 pg of total soluble protein was loaded on 10% SOS-PAGE and analyzed by immunoblotting with anti-CtBP. Relative CtBPL and CtBPs levels were unchanged during embryogenesis. A marked reduction in the relative level of CtBPL was observed from the larval through adult stages. CtBPs levels remained relatively unchanged throughout the developmental time course. The bottom panel shows B-tubulin as a loading control. C. Steady-state levels of CtBP mRNAs measured by RT-PCR analysis. Total mRNA from embryos and adults was reverse transcribed and PCR amplified using primers specific to CtBPL exons, CtBPs regions, or a region common to both isoforms as indicated. Tubulin mRNA was measured as a control. Reverse transcription reactions were primed with 60, 30, or 15 ng of total RNA, as indicated by triangular symbol. The —RT control reactions were primed with 60 ng of RNA. In this experiment, CtBPs levels increased from embryo to adult six-fold relative to either total CtBP or tubulin, while normalized CtBPL levels were unchanged. 58 A 8” 83V 83 \{9 KO k(.- 0 0° 90 ,bo 3“ e \o \o 6‘ 0° 6‘8 69 0‘ <9 <9 <26” <28“ 4“ Q9 5°“ "' . o 36 - 4 ’5 0 A B. <9 0 \09 (<3, «3’ 69 $89 989 e“ vs” v“ >9 '0 s s 3* 6° 65" '9 V26 Q°Q v8 v8 <10th . <10th IB :anti-CtBP 55i - - - _ - - ‘ IB :anti-tubulin 1 2 3 4 5 6 7 l Embryos Adults 59 We measured the relative levels of specific CtBP mRNA splice forms in embryonic and adult stages to determine if this developmental switch reflects a change in alternative mRNA isoform abundance. Primer pairs specific to the CtBPs, CtBPL, and to a region of the gene common to both isoforms were used in RT-PCR reactions. The absolute amounts of CtBPs and CtBPL RT- PCR products are not directly comparable because different primer sets were used, however the relative ratios in different stages of development are informative. The levels of CtBPs transcripts relative to total CtBP or tubulin mRNAs undergo a marked shift between these two stages, with CtBPs increasing 3-8 fold in the adults, while little change is seen in CtBPL levels (Figure ll-1C). This change suggests that the protein profile favoring CtBPs later in development may be dictated by changes in the abundance of distinct splice forms of the mRNA. Additional post-transcriptional effects may also contribute to the decreased CtBPL protein levels observed. Identification of conserved CtBPL-specific coding information We examined genomic sequences of 10 different insects representing >300 million years of evolutionary divergence - the fruit flies D. melanogaster, D. sechellia, D. persimilis, D. moja vensis, D. grimshawi and D. virilis, the mosquitoes Anopheles gambiae and Aedes aegypti (Diptera), the honey bee Apis mellifera (Hymenoptera) and the red flour beetle Tribolium castaneum (Coleoptera) - to determine if these organisms might also express diverse isoforms of CtBP. Analysis of putative open reading frames 3' of core 60 conserved CtBP sequences identified regions homologous to D. melanogaster exons 6 and 7, which encode the C-terminal extension of CtBPL (Figure ll-2A). In Drosophila species, the sequences of exon 6 appear to be separated from an upstream exon by a ~3 kbp intron, while the intron is of smaller size in mosquito and beetle. In the honey bee, this intron appears to have been entirely eliminated. The overall similarity among putative C- terminal coding regions is clearly lower than that observed for the core CtBP sequences (>92%), suggesting a lower level of evolutionary constraint (Figure Il-2B). However, some motifs are conserved; the similarities include several distinctive motifs involving less abundant amino acids, not simply tracts of repeating residues that would show similarities by chance. Splice signals following the terminal codons for exon 5 (YPEG), are conserved in all Drosophila, as well as lower Diptera and T. castaneum, suggesting that the downstream coding information is likely to be incorporated into mRNAs (Figure lI-ZC). Splice acceptor sites are present immediately 5' of the conserved LNGGYYT coding region of exon 6 in Drosophila species. A conserved splice acceptor sequence is not found directly 5' of INNGGY coding sequences present in T. castaneum and A. gambiae, raising the possibility that acceptor sites in alternative locations may be used (Figure ll- 20). In the bee, the open reading frame for the C-terminal extension is fused to the core sequences, 61 Figure "-2: Conservation of coding infonnatlon for CtBPL-speclflc C- terminus. A. Peptide coding information present in dipterans, bee, and beetle genomic sequences homologous to alternatively spliced exon 6 and 7 in Drosophila melanogaster encoding CtBP “tail” region. Conceptual translations of genomic sequences are shown below sequence of CtBPL, in which YPEG represents the end of the exon 5 coding sequence for the CtBPL isoform. Predicted intron size in nucleotides is indicated between exons. lntrons in Apis mellifera have apparently been eliminated. Dark gray shading indicates widely conserved sequences; light gray shading partially conserved sequences. Possible sumoylation sites (IN K X E) are indicated by gray bars above exon 7 residues. An alternative splice acceptor site 5' of the junction shown for exon 7 would produce an mRNA encoding an additional VSSQS motif at the beginning of exon 7 (not shown); this sequence is not conserved outside of Drosophila, unlike the residues 5' of exon 6 shown in ZC. B. Phylogenetic tree of species analyzed, displaying relative similarities of the C-terrninal ‘tail’ in different species compared to Drosophila melanogaster. Pairwise comparisons were performed using D. melanogaster exons 6 and 7 (103 amino acids) as a reference. Percentage indicates identical amino acids or a conserved substitution. Residues present in expansions in more divergent species such as D. mojavensis and D. virilis were not scored. C. The cDNA sequences reported for D. melanogaster CtBPL contain alternative splice acceptor sites for the 5' end of exon 6; a sequence isolated from adult head uses a downstream acceptor site (NP_001014617), while a different sequence isolated from embryo uses a more upstream acceptor (Sutrias-Grau and Arnosti, 2004) incorporating the residues LNGGYYT. This portion of the protein is evolutionarily conserved and contains appropriate splice acceptor sequences both 5' and 3' of this motif, thus alternative splicing may be a conserved feature here as well. 62 mm B a $4 _ E32330 h m m ”Ewan—mama --o ..... .. p-406Em---wdmm> 6mm? moo muu mmm um_muoumouuwuucuommommummmumummuommfiuuommmmmmummo ..... um _m mom mo... om» 3Q>mwm .< oawmeMMOUZHU‘NHAxKQ Ummw umu muo mom om Tmaumumuumuommommummmuummme ....................... um F mom 000 any hmuwmmocflme .D m a d U .H. w m. U U z A O m m M. o zOXw m ZOxm .0 Ezccflmmog. .x. hm 93:38.1 fix. on aq>mmm.< .X. ..v mmEEmm< o\a :4 _>>mcmE_._m.o .x. no m___.__> D o\o mm ,|. m_wco>m_oE d in m___E_9_8 o Slum 9.350090 3. v0 , .m chmmmocsoE d o\o oow supporting the notion that these are indeed coding sequences. Similar to the case with vertebrate CtBP proteins, the predicted C-terminal extensions of these CtBP isoforms are probably unstructured in solution. The sequences are rich in disorder promoting amino acids (ala, gly, pro, ser) and are predicted to not assume a globular structure by the GlobPlot program (not shown) (Linding et al., 2003). Developmental expression of alternative Isoforrns in D. mojavensis and D. virilis The presence of the regions correlating to D. melanogaster exons 6 and 7 does not in itself reveal whether distinct CtBP isoforms are produced, therefore we measured CtBP protein levels in embryos, larvae, pupae and adults from D. mojavensis and D. virilis, which are estimated to have shared the last common ancestor with D. melanogaster about 40-60 million years ago. Western blot analysis revealed that two major bands of sizes similar to CtBPs and CtBPL were present in these species (Figure "-3). The relative abundance of the CtBPL isoform decreases in larval and pupal stages, staying low in D. mojavensis in the adult, but increasing again in adult D. virilis. While differing in details, these changes suggest that developmental changes in relative abundances of CtBP isoforms are a conserved feature in Drosophilids. 65 Figure "-3: Conserved developmental regulation of CtBP protein expression in D. mojavensis and D. virilis. Expression of CtBP isoforms in embryos, larvae, pupae, and adults of D. mojavensis (A.) and D. virilis (B.). As in D. melanogaster, two predominant species were observed in both species, but the CtBPL isoform has a lower mobility (~60kDa vs. 50kDa in D. melanogaster). The relative levels of CtBPL to CtBPs in the embryo was greater in these species than in D. melanogaster, but just as in that species there is a pronounced decrease in relative levels of CtBPL in the larva and pupa. Adult levels of CtBPL remain low in D. mojavensis, but recover in D. virilis. 50 pg of total soluble protein was loaded on 10% SOS-PAGE and analyzed by immunoblotting with anti-CtBP. The bottom panels shows B-tubulin as a loading control. 66 50- 36- - -- , ‘ o cmaom mcmmmcmc. mmctozmzm wwmtbzmzc 02:5 m>m 90 of CtBP (H3150 mutation) induced wing blisters and rough eyes when misexpressed in the wing using an en-Gal4 driver and in the eye using the GMR-Gal4 driver. In contrast, misexpression of the NAD binding mutant, dimerization mutant, or the short isoform of CtBP induced no obvious phenotype in this assay (Figure Ill-1, Table Ill-1). To test if the differences in induced phenotypes reflected intrinsic differences in protein activity or rather in levels of protein expression, adult flies were assayed for CtBP protein levels by immunoblotting and larvae were assayed by immunostaining of eye imaginal discs. The CtBP proteins carry a double FLAG-tag at the C-terminus that permits detection of the recombinant protein by an anti-M2 antibody. As shown in Figure III-2, the steady state protein levels CtBPL (NAD), CtBPL (DIM) and CtBPs were significantly lower than the wild type CtBPL or CtBPL (CAT). The low expression of CtBPL (DIM) suggests that lack of dimerization may destabilize the protein, which raises the possibility that the CtBPL (NAD) mutant protein may also be poorly expressed due to lack of dimerization. The expression of CtBPL (CAT) protein was consistently observed to be comparable to the wild type protein for several independent lines tested. 91 Figure III-2: Relative steady state levels of recombinant CtBP proteins: CtBPL (NAD) and CtBPL (DIM) mutants and CtBPs isoform are expressed at low levels. A. Single adult flies misexpressing the transgene were solubilized in Laemmli sample buffer and extracts were probed with anti-M2 antibody to detect the recombinant FLAG-tagged proteins. The CtBPL (NAD) (lanes 5.6), or CtBPL (DIM) (lanes 7.8) were detected at low levels compared to CtBPL (lanes 1,2) and CtBPL (CAT) (lanes 9,10). CtBPs isoform was practically undetectable, although upon darker exposure a faint band could be observed (lanes 3. 4; data not shown). The lower panel shows the same blot stripped and reprobed with B-tubulin antibody as a loading control. B-tubulin was reduced in lanes 1- 3, indicating these lanes may be underloaded. Two independent lines per transgene are shown. B. Third instar wing imaginal discs were dissected from third instar larval progeny misexpressing the transgenes described above. Discs were immunostained with anti-M2 (121500). The eye driver GMR-Gal4 expresses CtBP in the posterior half of the imaginal disc. CtBPL and CtBPL (CAT) showed comparable expression (Panels 1, 2) while the CtBPL (NAD) (Panel 3). CtBPL (DIM) (Panel 4) and CtBPs (Panel 5) were expressed at low levels. Discs from GMR-Gal4 crossed to non-transgenic flies served as negative control (Panel 6). 92 GMR (eye driver) crosses IB :anti-M2 v-r * 50- g. lB:anti-tubulin 1 2 3 4 5 6 7 8 9 10 11 B. CtBPL CtBPL-CAT CtBPL-NAD CtBPL-DIM CtBPs yw ‘1 93 DroSOphiIa CtBPL presumptive catalytic mutant is competent for dimerization Consistent with previous cell-based and embryo repression assays, mutation of the well-conserved histidine residue does not measurably affect CtBP biological activity. The mutated histidine is important for catalysis in dehydrogenase enzymes and reported to be critical for the weak in vitro dehydrogenase activity of CtBPL (Balasubramanian et al.. 2003; Kumar et al., 2002). It is possible that this presumptive catalytic residue in CtBP might function in a different biological context not tested in our assay. In addition, the mutant protein may form heterodimers, recruiting one wild-type CtBP that may provide enough function to mask the effect of the mutation (Figure Ill-3). To test this possibility, co-immunoprecipitations were performed on embryo extracts expressing recombinant, epitope-tagged mutants ubiquitously. lmmunoprecipitation using the anti-M2 FLAG antibody recovered the epitope tagged CtBPL (CAT) protein. as well as endogenous CtBPL and CtBPs proteins (Figure Ill-3). The association of CtBPL (CAT) with endogenous CtBP proteins sheds new light on our previous embryo repression assays in which Gal4-CtBPL (CAT) was assayed (Sutrias-Grau and Arnosti, 2004). This result suggests that the repression activity of the catalytic mutant might be attributable to its dimerization with wild-type endogenous proteins. To better characterize the impact of this specific mutation. a CtBP null background is desired where possible heterodimerization can be ruled out (See Appendix A). 94 Input cg as , Q/ Q o8 e“ QK {‘3 * CtBPL-FLAG - - fiCtBPL "' .- C®DOCQ 8%.? . aw . . . _ dew . -..eo ._ fig. . | .. am m5 96 Sm Q 8 ms: :6 Sm .. 89¢ .35 X , as. ,. _....n.m8. 133 A heteroallelic CtBP mutation combination was tested to avoid mortality resulting from other uncharacterized recessive mutations that may be present on these chromosomes. All chromosomes contained identifiable markers to keep track of transgenes. Neither one nor two copies of CtBPL were able to rescue the lethality of homozygous CtBP mutations. Similar results were obtained when complementation was attempted with the short isoform alone, suggesting that maybe both forms of the protein are needed to sustain development. To test this possibility, CtBPL and CtBPs were recombined on the second chromosome and checked for rescue. However, lethality could not be rescued by simultaneous expression of both isoforms (See Table A-1). The developmental profile of endogenous CtBP proteins shows CtBPs to be present in relatively higher amounts than the long isoform (Mani-Telang and Arnosti, 2006). However, it was noted that the expression from promoter driven transgenic lines was the opposite, with higher CtBPL levels relative to CtBPs (Figure A-2). The relative abundances of the two isoforms may be critical for proper function. A developmental profile for transgenics expressing the recombinant protein showed a detectable decrease in levels of CtBPL- FLAG, mirroring the decrease in endogenous CtBPL levels, suggesting that the flag-tagged transgene is subject to the same regulation as the native proteins (Figure A-3). This decrease was more apparent with anti-CtBP antiserum (second panel) while the anti-M2 FLAG antibody did not reveal a detectable decrease; this decrease might reflect modified forms of CtBPL that do not cross-react well with the CtBP antiserum. CtBPL and CtBPs transcripts also differ in coding sequence and have different 3’UTRs. The 3’UTR is known to be important for regulating levels of protein expression either 134 temporally and/or spatially (Fujioka et al.. 1999; Kosman and Small, 1997). It is also conceivable that we failed to include other genomic regions that carry necessary transcriptional information. for example this construct lacks a large ~3 kb intron located between exons 5 and 6 (Figure A-1) that is conserved in Drosophilids (data not shown) which might contain regulatory regions that determine expression levels of the transgene. Finally, there are at least four CtBP transcripts have been detected by northern analysis using an embryonic cDNA library that might be functionally relevant to the fly (Poortinga et al., 1998). Correspondingly. four cDNAs have been recovered (Chapter l-Figure l-4). Although the differences appear to be minor, expression of only one or two of the isoforms might not suffice in this setting to provide complete biological function. These initial rescue experiments were attempted with cDNAs because of the ease of expressing different CtBP isoforms and mutants. An alternate approach will be to attempt rescue with the entire genomic locus contained within ~15 Kb. Such an endeavor has been successfully demonstrated for even-skipped, where a 16 Kb genomic region was sufficient and capable for rescuing lethality of an a null mutant (Fujioka et al., 1999). Alternative forms of the protein can then be tested in this context. A successful rescue with the full length CtBPL wbuld set the stage for a more definitive analysis of dehydrogenase mutants and C-terrninal extensions. This whole animal rescue approach has been challenging and has not been demonstrated in vertebrate models, thus these studies can add to our general understanding of CtBP in development. 135 MATERIALS AND METHODS Construction of transgenes For rescue tests, cDNA fragments for CtBPL (1.5Kb) and CtBPs (1.2Kb) were double flag-tagged at the C-terrnini and were inserted into the unique Kpnl and Xbal sites of pCaspeR-tubulin vector. This vector additionally provides a hexahistidine fusion at the N-terminus of CtBP. For rescue using CtBP endogenous promoter (PC-CtBP), cDNA fragments for both isoforms were introduced into the Kpnl and Xbal sites in the polylinker of the pUAST vector (Brand and Perrimon, 1993). The original vector has a basal hsp70 promoter and 5X UAS sites that were removed by restriction digestion with Sphl and Kpnl and replaced with an adaptor that containing Sphl, a new Ascl and Kpnl restriction sites and an optimal Kozak sequence (DA 885 5’- CACCGGCGCGCCACCAAAATGGGTAC-3’; DA 886 5’- CCATTI'TGGTGGCGCGCCGGTGCATG-3’). The promoter region of CtBP (~8054 bp) was amplified from embryo genomic DNA in two halves: the first 4000 bp fragment containing one of the predicted start sites was amplified as an Sphl-Ascl fragment (CG8841302-8845092) with DA 926 (5’- GTGCATGCGAAATGG‘I'I'AGCCAGCGTGGTG-3’) and DA 927 (5’- CGGGCGCGCCTTGAAATCGAGAATCCTGCAATGG-3’) and inserted immediately upstream of the cDNA. The second fragment (~4054 bp) with the other putative transcriptional start site was amplified as an Sphl-Sphl fragment (CG8837323-8841301) with primer sets DA 924 (5’- CTGCATGCATACCATAAT‘I’C‘ITGCAGT‘ITGCC-B’) and DA 925 (5’- CGGCATGCAGCTI'TCTGTTTCATGCATATGCAC -3') and introduced into 136 the Sphl site, upstream of the first fragment. Another version of this promoter driven construct (PH-CtBP) was built to retain the basal hsp70 promoter and the 5X UAS sites, in order to achieve drive higher levels of transgene using a combination of the CtBP regulatory region and the inducible pUAS promoter but was not used in rescue. In this case, the Sphl site was used to insert an Sphl-AscI-Sphl adaptor (DA 964 5’- CACCTCAGGCGCGCCACCTGCGCATG-3’; DA 965 5’- CGCAGGTGGCGCGCCTGAGGTGCATG-3’). The 8Kb promoter sequence was then cloned in two steps like that described for PC-CtBP. Western Blot Analysis Total extracts from adult flies were immunoblotted using 10% SOS-PAGE gels in a tank transfer system (Bio-Rad Mini Trans-Blot® Cell) and proteins were transferred to lmmuno-BlotTM PVDF membrane (Bio-Rad). Antibody incubation was performed in TBST (20mM Tris-HCI. pH 7.5, 120mM NaCl, 0.1% Tween-20) supplemented with 5% non-fat dry milk as a blocking agent. Mouse monoclonal antibodies include anti-M2 used to detect FLAG epitope- tagged CtBP (1:10.000), anti-CtBP (polyclonal, 1:10.000) (Struffi and Arnosti, 2005) and B-tubulin (1:6000, Iowa Hybridoma Bank). These were visualized using HRP-conjugated secondary antibodies (Pierce) and SuperSignal® West Pico chemiluminiscent substrate (Pierce). 137 CtBP fly stocks Flies carrying CtBP promoter driven transgenes were introduced into files by injection procedures described previously (Rubin and Spradling, 1982). Transgenic lines were checked for protein expression and flies homozygous for the transgene on the second chromosome were used for rescue. To test if rescue relied on both transgenes, flies expressing PC-CtBPL and PC-CtBPs carried over balancers were crossed and allowed to recombine on the second chromosome. Progeny were screened by performing an anti-M2 western to detect both FLAG-tagged forms of CtBPL and CtBPs from single flies still carrying balancer, which is indicative of a recombined chromosome. Flies carrying a P-element insertion in CtBP (Stock #P-1590, described in (Poortinga et al., 1998) or a deletion (Stock #1663) were obtained from Bloomington Stock Center. Mutant chromosomes for CtBP were maintained over balancer chromosomes i.e. TM3. Sb. All genetic crosses were maintained at 25 °C on standard cornmeal/molasses medium. 138 REFERENCES Basler. K. and Struhl. G. (1994). Compartment boundaries and the control of Drosophila limb pattern by hedgehog protein. Nature 368, 208-14. Brand, A. H. and Perrimon, N. (1993). Targeted gene expression as a means of altering cell fates and generating dominant phenotypes. Development 118, 401-15. Donaldson, T. D., Noureddine. M. A., Reynolds, P. J., Bradford, W. and Duronlo, R. J. (2004). Targeted disruption of Drosophila Roc1b reveals functional differences in the Roc subunit of Cullin-dependent E3 ubiquitin ligases. Mol Biol Cal/15, 4892-903. Fujioka, M., EmI-Sarker, Y., Yuslbova, G. L., Goto, T. and Jaynes, J. B. (1999). Analysis of an even-skipped rescue transgene reveals both composite and discrete neuronal and early blastoderm enhancers, and multi-stripe positioning by gap gene repressor gradients. Development 126, 2527-38. Crumbling. G., Strelets, V. and Consortium, T. F. (2006). FlyBase: anatomical data, images and queries. Nucleic Acids Research 34 D484-D488; doi:10.1 093/nar/gkj068. http://flmaseorg/ Kosman, D. and Small, S. (1997). Concentration-dependent patterning by an ectopic expression domain of the Drosophila gap gene knirps. Development 124. 1343-54. Manl-Telang, P. and Arnosti, D. N. (2006). Developmental expression and phylogenetic conservation of alternatively spliced forms of the C-terrninal Binding Protein corepressor. Development. Genes and Evolution In Press. Moran, E. and Jimenez, G. (2006). The tailless nuclear receptor acts as a dedicated repressor in the early Drosophila embryo. Mol Cell Biol 26, 3446- 54. Poortinga, G., Watanabe, M. and Parkhurst, S. M. (1998). Drosophila CtBP: a Hairy-interacting protein required for embryonic segmentation and hairy- mediated transcriptional repression. Embo J 17, 2067-78. Rubin, G. M. and Spradling, A. C. (1982). Genetic transformation of Drosophila with transposable element vectors. Science 218, 348-53. Struffi, P. and Arnosti, D. N. (2005). Functional interaction between the Drosophila knirps short range transcriptional repressor and RPDB histone deacetylase. J Biol Chem 280, 40757-65. Sutrias-Grau, M. and Arnosti, D. N. (2004). CtBP contributes quantitatively to Knirps repression activity in an NAD binding-dependent manner. Mol Cell Biol 24, 5953-66. 139 Wagner, C. R., Mahowald, A. P. and Miller, K. G. (2002). One of the two cytoplasmic actin isoforms in Drosophila is essential. Proc Natl Acad Sci U S A 99, 8037-42. 140 APPENDIX B AN RNAi APPROACH TO DEPLETING THE CtBPL ISOFORM The CtBP gene encodes two major splice variants (CtBPL and CtBPs), both of which can be detected through all developing stages of the fly (Chapter I - Figure I-4). CtBP plays vital roles during fly embryogenesis, emphasized by mutations in the gene which are homozygous lethal. It is not known whether the existence of multiple isoforms imparts functional specificity during development. however expression of single isoforms is not sufficient to rescue lethality. In other systems, there are several examples of distinctions between CtBP isoforms. Human CtBP1, but not CtBP2, is modified by sumoylation that results in its nuclear translocation. CtBP2 but not CtBP1 contains an NLS that directs it to the nucleus, suggesting that CtBP1 and CtBP2 are regulated differently. Mouse knockouts show distinct phenotypes, lending support to the hypothesis of different isoforms having specific roles. Initial attempts for a transgene-based rescue experiment do not provide any clues regarding the functional equivalency of the two isoforms in Drosophila (Appendix A). Both isoforms when tethered at the promoter have been documented to display comparable repression activities (Sutrias-Grau and Arnosti, 2004). In vivo studies assaying biological activity of CtBPs have been of limited value due to the low expression levels of this protein. However, we observe a decline in CtBPL protein levels from the larval to adult stages (Mani-Telang and Arnosti, 2006). Whether this translates to CtBPs supplying CtBPL like functions or whether CtBPL is no longer required during these developmental stages and is hence down regulated, remains to be 141 determined. This downregulation might transpire by post-translational modifications that occur within the C-tenninal tail (Chinnadurai, 2006) (Also see Chapter 1). As an independent yet complementary approach, we tried to selectively deplete levels of CtBPL isoform to gain insights into isoform-specific roles. To inhibit CtBPL levels. we used RNA interference (RNAi) in combination with the inducible UAS-Gal4 system in a fly model. RNAi by double stranded RNA was first described in worms and the mechanism has since been found to be conserved in vertebrates, invertebrates and plants (Reichhart et al., 2002). In Drosophila, the UAS-Gal4 system has been successfully applied to study gain-of-function and more recently, loss-of-function phenotypes; the latter resulting from gene specific perturbations by RNAi (Duffy, 2002; Roman, 2004). In Drosophila, The C-terrninal region of CtBPL is absent in CtBPs (includes exon 6+7 and a ~700 bp 3’UTR) and represents a unique sequence by which this particular isoform can be targeted. A responder line consisting of tandem inverted repeats to the C-tenninal tail region of CtBPL (See Materials and Methods) were cloned downstream of the UAS sites in pUAST and were expressed ubiquitously using a constitutive ActinSC-Gal4 driver line (See Figure B-1). This cross tests the efficacy of the hairpins to knockdown CtBPL by inducing the expression of hairpins in a ubiquitous fashion early in development. If the function of CtBPL is mandatory during these times, then lethality among progeny is expected. If lethality were to be observed, it would validate that interference with CtBP function is possible, following which the next step would be test the specificity of knockdown. RNAi has been demonstrated to be gene specific, but it is uncertain if the inhibitory effects 142 can spread to related family members (like splice variants). It is possible that the inverted repeats will also cause a depletion of CtBPs levels. This possibility will be tested by crossing the responder line to a tissue specific driver. and testing CtBP levels in this tissue by a western blot. These experiments are currently in progress. If specific knockdown of CtBPL levels are observed, I shall analyze outcomes arising from depletion of this isoform at various stages and in different tissues during development. Another possibility is that partial knockdown of CtBPL is observed, in which case it would still be interesting to analyze phenotypes from temporal and spatial reductions of the long isoform. 143 Actin SC promoter 5X UAS CtBP IRs d.s. hairpins crap. I—I—I—I~EE stem mica-{aw 3' UTR Figure B-1: A transgenic system to trigger RNAi and selectively eliminate CtBPL in Drosophila A transgenic driver line expressing Gal4 activator under the control of a ubiquitous driver (Actin SC) is crossed with a transgenic responder line expressing tandem inverted repeats against CtBPL, downstream of UAS- regulatory sites. The responder line was made homozygous for the P-element carrying the inverted repeats and the driver line is maintained over a balanced chromosome. The results of CtBPL knockdown will be judged by screening progeny lacking the balancer-marker resulting from this cross. 144 MATERIALS AND METHODS Construction of transgenes Tandem inverted repeats were cloned into the pUAST vector that allows expression of dsRNA hairpins in vivo, facilitating inducible knockdown of CtBPL (Brand and Perrimon, 1993). Two types of double stranded hairpin constructs were generated. The first construct was designed to target coding sequence for exons 6+7 in CtBPL (221 bp+81 bp at the C-termini). Inverted repeats (le) of this ~300 bp sequence were amplified using CtBPL cDNA as a Bglll-Notl fragment using primer pairs DA 1219 (5'- CTGGATAGATCTGCACTGCATCACCGGGCACAC-3’) and DA1220 (5'- ATAAGATGCGGCCGCCGGCGCCTCCG‘I‘I’GACTC-3’) for the forward repeat .The reverse repeat was amplified from as a Kpnl-Xhol fragment using DA 1221 ( 5’-GATGGTACCGCACTGCATCACCGGGCACAC-3’) and DA 1222 (5’-GATCTCGAGCGGCGCCTCCGTTGACTCGG-3’) from CtBPL cDNA. The second construct was designed to target unique 3’ exon regions encoding CtBPL ‘tail’, along with ~700 bp of 3’ UTR sequence specific to this isoform. Inverted repeat sequences (~1.3 Kb) were amplified as a Bglll-Notl fragment using primer pairs DA 1219 (see above) and DA 1223 (5’- ATAAGATCAGCGGCCGCG'ITI’CTCGTAATTAAAATTTI’CCAAC-S') for the forward repeat and primer pairs DA 1261 (5’- TATTCTAGAGCACTGCATCACCGGGCAC-3') and DA 1262 (5’- TCAGGTACCGTTTCTCGTAATTAAAATT'I'I’CCAAC-3’) for the reverse repeat amplified as a Xbal-Kpnl insert. In both constructs. the inverted repeats were separated from each other using an ~800 bp region containing the 145 second intron of the mub transcript (CG7437) amplified with primers DA 753 (5’- ATAAGATCAGCGGCCGCCAGGACGTCCAATCAAAGTGGTCAAACCCG-B’) and DA 754 (5’- CTGGATCTCGAGGGCTGGAG'I'TCAATAAATATACCATCGCTCTTTGGC-B’) (Used in Reichhart et al., 2002). Fly Stocks and Expression System Transgenic fly lines carrying the UAS-RNAi vectors were obtained using standard techniques of P-element mediated germline transformation. Multiple independent lines for each construct were made homozygous and all crosses were performed at 25°C. Knockdown efficacy was tested by crossing to a strong ubiquitous actin50-Gal4 driver line (Bloomington Stock Center, Stock # 4414) that is carried over a marked balancer chromosome. The progeny of this cross will first be screened for any flies that carry non-marked chromosomes and if such progeny emerge, will be validated for CtBP protein levels by western blotting with an anti-CtBP antisera (as described in Chapter ll- Materials and Methods). 146 REFERENCES Brand, A. H. and Perrimon, N. (1993). Targeted gene expression as a means of altering cell fates and generating dominant phenotypes. Development 11 8, 401-15. Chlnnadural, G. (2006). CtBP family proteins:unique transcriptional regulators in the nucleus with diverse cytosolic functions: Landes Bioscience. Duffy, J. B. (2002). GAL4 system in Drosophila: a fly geneticist's Swiss army knife. Genesis 34, 1-15. Mani-Telang, P. and Arnosti, D. N. (2006). Developmental expression and phylogenetic conservation of alternatively spliced forms of the C-terminal Binding Protein corepressor. Development, Genes and Evolution In Press. Reichhart. J. M., ngoxygakls, P., Naltza, S., Woerfel, G., Imler, J. L. and Gubb, D. (2002). Splice-activated UAS hairpin vector gives complete RNAi knockout of single or double target transcripts in Drosophila melanogaster. Genesis 34, 160-4. Roman, G. (2004). The genetics of Drosophila transgenics. Bioessays 26, 1243-53. Sutrias-Grau, M. and Arnosti, D. N. (2004). CtBP contributes quantitatively to Knirps repression activity in an NAD binding-dependent manner. Mol Cell Biol 24, 5953-66. 147 llllllllllliilllllll