T. :6” . y... . . §w..am.!;,§umuv. {My )4 vhfhrtun . . .. 3 : LP $32 if.“ u % v1 Shun: .mw . a .21. 1?. fit , .. I... 2... 7“»? . 16.)..1» ”as. 2.. ... a. . nJ. ‘ . $3 . u} v n.1, ... .3: m n . Mum: ”£qu J.Lu4.”Mm.. o. a. 3.. . mmnumfk fifiw‘qn... 2. 1 ‘. i . 2... . f... #4.. Wyatt“; .3 in", I . I . .91.. 1 V L: ‘4. (.30! I) Iva: x, in“. L nu. (Gr. 29!}; ' .u-i. fink? V . 13%.; :u , 5.22:..fiunvflii E......um.‘_nmmm£. a ausfih E ELE.‘ , . . #63“? ,a l ' 3%... 1%.... 45.1.??? : x ‘ 1; r .3 . 2 LIBRARY 3 005 Michigan State ‘ University This is to certify that the dissertation entitled Regulation of human small nuclear RNA gene transcription by the oncogneic protein kinase CK2 presented by Liping Gu has been accepted towards fulfillment of the requirements for the Doctoral degree in Department of Biochemistry and Molecular Biology Ago/114% Major fidfessor’s Signature pg” (/0! Date MSU is an Affirmative Action/Equal Opportunity Institution PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 2/05 ammonium-ms REGULATION OF HUMAN SMALL NUCLEAR RNA GENE TRANSCRIPTION BY THE ONCOGENIC PROTEIN KINASE CK2 By Liping On A DISSERTATION Submitted to Michigan State University for the Degree of DOCTORAL OF PHILOSOPHY Department of Biochemistry and Molecular Biology 2005 ABSTRACT HUMAN SMALL NUCLEAR RNA GENE TRANSCRIPTION REGULATED BY THE ONCOGENIC PROTEIN KINASE CK2 By Liping Gu Protein kinase CK2 participates in many cellular processes including cell cycle, cell differentiation, stress response, and apoptosis. One important role of CK2 is to transduce extracellular signals into the nucleus to control gene expression. CK2 appears to globally regulate protein-coding gene transcription by RNA polymerase II and non- coding gene transcription by RNA polymerases I and III. Human small nuclear RNA genes are transcribed by either RNA polymerase II or RNA polymerase III depending upon their promoter architectures. CK2 regulates RNA polymerase III-specific U6 snRNA gene transcription, but whether CK2 plays a role in regulating the closely related polymerase II-specific U1 snRNA gene transcription has been uncertain. Herein, I report that CK2 inhibits U1 snRNA gene transcription by RNA polymerase II. CK2 associates with U1 gene promoters, and U1 snRNA expression negatively correlates with endogenous CK2 levels. Thus, CK2 plays a direct role in U1 gene regulation in living cells. CK2 phosphorylates the general transcription factor snRNA activating protein complex (SNAPC) that is required for human snRNA gene transcription. SNAPC phosphorylation reduces its DNA binding affinity as supported by the observations that CK2 phosphorylation of a recombinant partial SNAPC can inhibit its PSE-specific binding before and after DNA association has occurred. Importantly, CK2 phosphorylation of mini-SNAPc restricts promoter recognition by SNAPC on a U1 promoter and permits higher order complex formation with TBP on a U6 promoter, indicating that the cooperation between SNAPc and TBP can counteract CK2 inhibition of DNA binding by SNAPC for RNA polymerase III-specific preinitiation complex assembly. Therefore, CK2 may differentially regulate snRNA gene transcription by selectively influencing pre-initiation complex assembly for RNA polymerase II and polymerase III transcription of human snRNA genes depending upon the promoter architectures. Further investigation showed that SNAP190, the largest subunit of SNAPC, is phosphorylated in vivo and can be phosphorylated in vitro. CK2 phosphorylates the N- terminal half of SNAP19O at two regions that contain multiple CK2 consensus sites (amino acid 20-63 and 514-545) as determined by mass spectrometric analysis. The region within SNAP19O that is required for CK2 inhibition is contained within amino acids 506-719 encompassing a serine rich region with multiple CK2 consensus sites. The downregulation of SNAPC DNA binding activity by CK2 phosphorylation may contribute to reduced U1 snRNA gene transcription by RNA polymerase II. iv T o my family ACKNOWLEDGEMENTS I gratefully acknowledge my mentor R. William Henry for his encouragement, support, patience, and understanding. It was a great fortune for me to meet Bill and to join his laboratory when I arrived in Michigan several years ago. Thank you very much for mentoring me during these years in the Ph. D. program and teaching me how to become a scientist. I would also like to thank the members of my thesis committee, Zachary F. Burton, Kathleen A. Gallo, James H. Geiger, John L. Wang, and R. William Henry for their time and effort in my guidance during these years. I would like to thank the members of the Henry lab including Craig Hinkley, Heather Hirsch, Zakir Ullah, Gauri Jawdekar, Anastasia Gridasova, Tharakeswari Selvakumar, and Xianzhou Song for sharing their expertise and friendship. I would also like to thank my friends and colleagues in this department and other departments. Special thanks to Rhonda Hussain, Susanne Hoffmann-Benning, Keith Ray, and Walter Esselman and his student Weiqin Chen for technique assistance. I would like to thank my husband Ruanbao Zhou and my son Kai Ruan for their tremendous support throughout all these years. I would also like to thank my parents and siblings in China for their love and understanding. TABLE OF CONTENTS List of Figures ....................................................................................... ix Key to symbols and abbreviations ............................................................... xi Chapter One Introduction and literature review 1. Human small nuclear RNA gene transcription ................................................ 1 1.1 The functional significance of snRNAs 1.2 Human snRNA gene transcription 1.2.1 The structures of human snRNA gene promoters 1.2.2 Trans-acting factors required for snRNA gene transcription 1.2.2.1 The DSE binding factors 1.2.2.2 The PSE binding factor 1.2.2.3 The TATA box binding factor 1.2.3 Composition of RNA pol II and pol 1H transcription initiation complexes 1.2.3.1 The basal transcription machinery for pol II-specific snRNA genes 1.2.3.2 The basal transcription machinery for pol III-specific snRN A genes 1.2.4 Other factors involved in regulating human snRN A gene transcription 2. Protein Kinase CK2 .............................................................................. 15 2.1 CK2 structure and chemical properties 2.1.1 CK2 holoenzyme 2.1.2 Regulation of CK2 enzymatic activity 2.1.3 Dual-substrate and dual-cosubstrate specificity of CK2 vi 2.2. CK2 participates in various cellular processes 2.2.1 CK2 localization 2.2.2 CK2 in cell proliferation 2.2.3 CK2 in cell survival and apoptosis 2.2.4 CK2 and cancer 2.2.5 CK2 in stress response 2.3. The role of CK2 in transcription regulation 2.3.1 CK2 phosphorylation controls transcription at different levels 2.3.2 The role of CK2 in protein-coding gene expression 2.3.3 The role of CK2 in non-coding gene transcription 3. Main hypothesis and experimental design .................................................... 33 4. References ......................................................................................... 35 Chapter Two Cooperation between SNAPC and TBP antagonizes protein kinase CK2 inhibition of DNA binding by SNAPC Abstract ....................................................................................... 55 Introduction .................................................................................. 56 Materials and Methods ..................................................................... 60 Results ........................................................................................ 65 Discussion ..................................................................................... 90 References ................................................................................... 93 Chapter Three CK2 phosphorylation of SNAP190 negatively regulates SNAPC function Abstract ...................................................................................... 98 Introduction .................................................................................. 99 Materials and Methods ................................................................... 102 vii Results ...................................................................................... 107 Discussion .................................................................................. 127 References ................................................................................. 130 Chapter Four Summary and Future Plan Summary and future plan ................................................................ 134 References ..................................................................................... 141 viii LIST OF FIGURES Figure 1-1. Schematic representation of cis-elements of human snRNA promoters ..... 3 Figure 1-2. SNAP190 acts as a scaffold protein at snRNA promoters ..................... 7 Figure [-3. Composition of snRN A transcription initiation complexes for transcription by RNA polymerases II and III ................................... 12 Figure 11-1. CK2 inhibits U1 snRNA in vivo gene expression ............................ 67 Figure 11-2. CK2 represses U1 in vitro transcription ......................................... 71 Figure 11-3. Endogenous CK2 associates with SNAPC ..................................... 74 Figure 11-4. Endogenous SNAPC is phosphorylated ........................................ 79 Figure 11-5. Endogenous CK2 phosphorylates the N-terminal region of SNAP190 ........................................................................... 82 Figure 11-6. TBP and SNAPC cooperate at U6 but not at U1 promoter probes to overcome CK2 inhibition of DNA binding by SNAPC . . . . . . . . ....87 Figure 111-1. SNAPC function is modulated by protein kinase CK2 ...................... 108 Figure 111-2. Endogenous CK2 phosphorylates the N-terminus of SNAP190 in the region within amino acids 20-63 containing multiple CK2 consensus sites .............................................................. 111 Figure III-3. Endogenous CK2 phosphorylates the N-terminus of SNAP190 ix in the serine rich region within amino acids 514-545 containing multiple CK2 consensus sites ................................................... 114 Figure III-4. Deletion of CK2 phosphorylation regions within SNAP190 did not affect the transcription properties of SNAPC ........................ 118 Figure III-5. A functional comparison between mini-SNAP complexes m8 and mSAC ......................................................... 123 2, 3-DPG 6-TG APE/Ref- 1 ARC ATFl bPrP Brfl -TFIIIB Brfl-TFIIIB ChIPs Chk2 CHOP/GADDI 53 CK2 CO-IP CoREST CPE CREB CTCF CTD C-terminal Da DEK DHN l DPE DRB DSB KEY TO SYMBOLS AND ABBREVIATIONS 2, 3-diphosphoglycerate 6-thioguanine apurinic endonuclease apoptosis repressor with caspase recruitment domain activating transcription factor-l bovine prion protein TFIIIB complex composed of TBP, Brfl and del TFIIIB complex composed of TBP, Bra and del chromatin immunoprecipitations checkpoint kinase 2 C/EBP homologous protein transcription factor/growth arrest and DNA damage inducible protein casein kinase 2 or casein kinase H co-immunoprecipitation corepressor for the repressor element 1 silencing transcription factor core promoter element CAMP-dependent response element binding protein CCCTC-binding factor carboxy terminal domain of RNA polymerase 11 largest subunit carboxy-terminal Dalton proto-oncogene protein transcribed from a gene named dek dehydrin protein 1 downstream promoter element 5, 6-dichloro- l -B-D-ribofl.1ranosylbenzimidazole double-strand breaks xi DSE d-siRNA dsRNA ecto-CKZ EMSA FACT FCPl FGF-Z GAPDH GTFs HDAC HEXIM 1 HP] HS 1 HSFl HTH ICB90 1P IR IKBu MALDI-TOF MS MEFZC mini- SNAPC mRNA MS mS mSA(N+C) mSAC mSAN MT A-2 distal sequence element Dicer generated small interfering RNA double stranded RNA CK2 released from the surface of intact cells electrophoretic mobility shift assay facilitating chromatin-mediated transcription TFIIF-associated CTD phosphatase 1 fibroblast growth factor-2 glyceraldehyde-3-phosphate dehydrogenase the general transcription factors histone deacetylase hexamethylene bisacetamide-inducible protein 1 heterochromatin-associated protein 1 haematopoietic lineage cell-specific protein 1 heat shock factor-1 helix- turn-helix motif Inverted CCAAT box Binding Protein of 90 kDa immunoprecipitation ionizing radiation Inhibitor of kappaB alpha matrix-assisted laser desorption/ionization—time of flight mass spectrometry myocyte enhancer factor-2C complex composed of SNAP43, SNAPSO, SNAP190 (1-719) messenger RNA mass spectrometry mini- SNAPC complex composed of SNAP43, SNAPSO, SNAP190 (63-505) complex composed of SNAP43, SNAPSO, SNAP190 (1-505) complex composed of SNAP43, SNAPSO, SNAP190 (63-719) multithreaded architecture xii muTATA NAP-2 NE NFATc NFKB N-terminal OIR PA-PLA l or PBP PEST domain PIC PK6OS PKA pol 13131386 PSE pSer P-TEF b PTF pThr PTP PTyr Q-TOF MS/MS quercetin RB RbAp48 RNAi RNPS RNPS 1 rRN A RRR mutant TATA box sequence nucleosome assembly protein 2 nuclear extract nuclear factor of activated T cells nuclear factor kappa B amino-terminal Oct-1 interact region phosphatidic acid-preferring phospholipase 1A proximal sequence element binding protein proline-glutamic acid-serine-threonine domain Pre-initiation complex protein kinase 608 protein kinase A RNA polymerase phosphatase proximal sequence element phosphoserine positive transcription elongation factor-b proximal sequence element transcription factor phosphothreonine phosphotyrosine phosphatase phosphotyrosine quadrupole time of flight mass spectrometry 3, 3’, 4’, 5, 7-pentahyroxyflavone the retinoblastoma tumor suppressor protein the Rb-associated protein RNA interference ribonucleoprotein particles RNA-binding protein prevalent during S phase ribosomal RNA arginine rich region xiii RT-PCR SANT SBF SL 1 /T IF -IB SNAPC snRNAs snRNPs SOD SPH SRR SSB SSRPl Staf SV 40 TAF TBB TBP TGE running buffer TLC TLE TLR-3 tRNA TRR uPA UPE UV VSMC WCE thSE thATA XRCCl reverse transcriptase polymerase chain reaction DNA-binding domain in SWI-SNF, ADA, N-CoR and TFIIIB the SPH binding factor promoter selectivity factor or transcription initiation factor 1B the snRNA activating protein complex small nuclear RN As small nuclear ribonucleoprotein particles superoxide dismutase SphI postoctamer homology serine rich region single-strand breaks structure-specific recognition protein 1 the selenocysteine tRN A gene transcription activating factor Simian virus 40 large T antigen TBP-associated factor 4, 5, 6, 7-tetrabromobenzotriazole TATA binding protein buffer containing 50 mM Tris, 380 mM glycine, 2 mM EDTA thin layer chromatography thin layer electrophoresis Toll-like receptor 3 transfer RNA TBP recruitment region urokinase upstream promoter element ultraviolet light vascular smooth muscle cell whole cell extract wild type PSE sequence wild type TATA box sequence the X-Ray cross-complementing group 1 protein xiv CHAPTER 1 INTRODUCTION AND LITERATURE REVIEW 1. Human small nuclear RNA gene transcription 1.]. The functional significance of snRNAs The final products of human small nuclear RNA genes are small nuclear RNAs (snRNAs), which play fundamental roles in processing of other RNAs. For example, U1, U2, and U6 snRN As are essential components of small nuclear ribonucleoprotein particles (snRNPs) that control pre-messenger (m) RNA splicing (Burge et al., 1999). U3 snRNA is involved in ribosomal (r) RNA processing (Fatica, et al., 2002; Grandi, et al., 2002), while H1 snRNA is the catalytic subunit of RNase P responsible for transfer (t) RNA maturation (Bartkiewicz et al., 1989). In addition, snRNA functions in regulating mRNA gene transcription. For example, U1 snRNA associates with TFHH and stimulates TFIIH-dependent RNA polymerase (pol) H transcriptional initiation (Kwek etal., 2002). 78K snRNA, on the other hand, associates with a positive transcription elongation factor- b (P-TEFb) (Peng et al., 1998; Wei et al., 1998) and the hexamethylene bisacetamide- inducible protein 1 (HEXIMl) (Ouchida et al., 2003; Michels et al., 2003). HEXIMl sequesters the 7 SK-P-TEFb ribonucleoprotein particle (RNP) and represses P-TEFb kinase and transcriptional activities by inhibiting P-TEFb phosphorylation of the carboxy terminal domain (CTD) of RNA pol II (Nguyen et al., 2001; Yang et al., 2001; Yik et al., 2003; Chen et al., 2004; Yik et al., 2005). Futhermore, snRNA can mediate a preimmune signal. For example, U1 snRNA, a component of U1-70-kDalton (kDa) RNP, is capable of inducing innate immunity signaling through activation of Toll-like receptor 3 (TLR-3) (Hoffman et al., 2004). Human snRNAs fimction in diverse cellular processes, the levels of snRNA gene transcription need to be well regulated when cells undergo cell proliferation, differentiation and embryo development (Caceres et al., 1992; Cheng etal., 1997; Glibetic et al., 1992; Lund et al., 1987; Meissner et al., 1995; Nash et al., 1987; Robert et al., 2002). 1.2 Human snRNA gene transcription Human snRNA genes are transcribed by either RNA pol II or RNA pol III. The polymerase specificity depends upon their promoter architectures (for a review, see Hernandez, 2001). 1.2.1 The structures of human snRNA gene promoters U1 and U2 snRNA gene promoters are representatives of pol H-specific snRNA promoters, whereas U6 and 78K snRNA gene promoters are representatives of pol III- specific snRNA promoters. As illustrated in Figure I-l, two distinct and conserved cis- acting regulatory regions that are important for snRNA gene transcription are present in both human RNA pol II and pol III snRNA gene promoters. A proximal sequence element (PSE) is located in the core promoter region centered near position -55 relative to the transcription initiation start site and a distal sequence element (DSE) is located in the regulatory region near position —220, and serves as a transcriptional enhancer. The PSE is Figure 1-1. Schematic representation of cis-elements of human snRNA promoters. The cis-elements on RNA pol II transcribed snRNA gene promoters, such as the U1 snRNA promoter, include a proximal sequence element (PSE) centered near position -55 relative to the transcription initiation start site and a distal sequence element (DSE) near position -200 at the regulatory region. RNA pol III-transcribed snRN A genes, represented by the U6 snRNA promoter, contain an additional TATA-box near position -25 adjacent to the PSE at the core promoter. * Images in this dissertation are presented in color. .90 E05 28 869 E08. zoo. e \\ 5.5. T mmm mmo AIL \\ P+ mm- mm. com- A... .8. .20an 9.3 A) induced a marked inhibition of cell proliferation in both NIH3T3 and CCL39 cells (Lebrin et al., 2001). These results indicate that CK2 plays an important role in controlling cell division and cell proliferation. 2.2.3. CK2 in cell survival and apoptosis Biochemical and genetic evidence indicates that CK2 participates in the maintenance of cell viability. CK2 appears to exert an anti-apoptotic role by protecting regulatory proteins from caspase-mediated degradation (Litchfield, 2003). Inhibition of CK2 by its specific inhibitor TBB induces apoptosis in Jurkat cells (Ruzzene etal., 2002). Similarly, depletion of endogenous CKZa and/or CKZa ' catalytic subunits by RNAi also causes marked apoptosis when HeLa cells have been exposed to 6-thioguanine (6-TG) (Yamane and Kinsella, 2005a) or ionizing radiation (IR) (Yamane and Kinsella, 2005b). However, these responses can be suppressed by introducing ARC (apoptosis repressor with caspase recruitment domain) or by blocking caspase enzymatic activity (Yamane and Kinsella, 2005a; Yamane and Kinsella, 2005b), suggesting that CK2 participates in inhibition of apoptosis by negatively regulating caspase activity. A number of antiapoptotic proteins, such as Bid (Desagher et al., 2001), Max (Krippner-Heidenreich et al., 2001), ICB90 (Inverted CCAAT box Binding Protein of 90 kDa) (Bronner et al., 2004), and HSI (haematopoietic lineage cell-specific protein 1) (Ruzzene et al., 2002) are 23 targeted by CK2 leading to downregulation of caspase-dependent degradation. Moreover, increased expression of CK2 protects cells from drug-induced apoptosis (Guo et al., 2001). Taken together, these observations indicate that CK2 plays a critical role in maintaining cell survival. 2.2.4. CK2 and cancer The growth-promoting and anti-apoptotic properties of CK2 may contribute to its ability to participate in transformation. CK2 is abnormally active in a variety of human cancers including leukemias and solid tumors (Munsterrnann et al., 1990; Landesman- Bollag et al., 2001). The detection of higher CK2 nuclear translocation in tumor cells suggests that CK2 nuclear association plays an important role in tumorigenesis (Faust et al., 1999; Guo et al., 2001). 2.2.5. CK2 in stress response Another important function of CK2 is that it acts as a downstream effector responsible for transducing genotoxic stress signals to either the transcriptional machinery or DNA-damage repair system. For example, in yeast, CK2 targets the TBP component of Brfl -TFIIIB for regulation of RNA pol III-specific tRNAs and SS rRNA gene transcription. Transcription of these genes is repressed in cells that have experienced DNA damage, which causes the dissociation of the CK20L from the CK2/TFIIIB complex (Ghavidel and Schultz, 2001; Schultz, 2003). Thus, CK2 acts as a terminal effector in the signaling pathway that transduces DNA-damage signals to the RNA pol III 24 transcriptional machinery. In addition, disruption of the CKZB gene causes yeast cells to permanently arrest in G2/M at the DNA damage checkpoint when double-strand breaks (DSB) occur, while normal cells can override this block, suggesting a role of CK2 in controlling adaptation to the DNA-damage checkpoint (Toczyski et al., 1997). CK2 phosphorylation of histone H4 at serine 1 appears to promote DNA DSB repair (Cheung et al., 2005). Furthermore, in mammalian cells, CK2 facilitates DNA single-strand breaks (SSB) repair mediated by the X-Ray cross-complementing group 1 protein (XRCCI) (Loizou et al., 2004). CK2 can also control the activity of the DNA repair protein apurinic endonuclease (APE/Ref-l) (Fritz and Kaina, 1999). APE/Ref-l phosphorylation by CK2 stimulates its redox capability towards the transcription factor AP-l, thus promoting AP-l DNA binding activity and activating its target genes. Upon CK2 inhibition, cells become more sensitive to DNA-damage reagents (Fritz and Kaina, 1999). Moreover, CK2 has been shown to translocate to the nuclear matrix in response to heat shock (Gerber et al., 2000). In addition, CK2 regulates p53 activity (Keller et al., 2001), and p53 phosphorylation by CK2 is increased in response to UV irradiation (Blaydes and Hupp, 1998). In the case of nuclear factor kappa B (NFKB), its activation is dependent upon CK2 phosphorylation of IKBa, an inhibitory partner that associates with and sequesteres the NFKB in the cytoplasm. Upon oxidative stress, CK2 targets IKBab for degradation, thereby allowing NFKB proteins to translocate to the nucleus and induce transcription (Schoonbroodt et al., 2000). These data indicate that CK2 is a key participant in the cellular response to various stress conditions. 25 2.3. The role of CK2 in transcription regulation Protein phosphorylation appears to be one the most important post-translational modifications involved in the regulation of transcription factor activity. Protein phosphorylation also impinges upon most of the signal transduction pathways. Therefore protein phosphorylation provides a link between signal transduction and gene expression (Karin, 1994; Bohmann, 1990; Litchfield, 2003). Phosphorylation or dephosphorylation of transcription factors and/or other related proteins either positively or negatively regulate their activity to control gene expression. 2.3.1. CK2 phosphorylation controls transcription at different levels 2.3.1.1. Several mechanisms have been proposed for a role of CK2 phosphorylation of transcription factors in gene regulation. (a) Phosphorylation by CK2 influences transcription factor subcellular localization. For example, the nuclear import of SV40 large T antigen is enhanced by CK2 phosphorylation at serine 112 (Xiao et al., 1998); while phosphorylation of muscle- specific transcription factor Myf-S at serine 49 by CK2 reduces its nuclear localization (Winter et al., 1997). In contrast, CK2 phosphorylation of NFATc (nuclear factor of activated T cells) promotes NFATc nuclear export (Porter et al., 2000). (b) Phosphorylation of transcription factors by CK2 governs their DNA-binding activities both positively and negatively. For example, CK2 phosphorylation of the myocyte enhancer factor-2C (MEF2C) at serine 59 enhances the DNA binding and 26 transcriptional activity of MEF2C (Molkentin et al., 1996). Heat shock factor-l (HSF 1) phosphorylation by CK2 at threonine 142 promotes it binding to heat shock elements and trans-activation of the HSP70 gene (Soncin et al., 2003). In addition, the high-mobility group domain protein SSRPl (structure-specific recognition protein 1), which plays a role in transcription and DNA replication in the chromatin context, is targeted by CK2. SSRPl phosphorylation at serine 510 by CK2 prevents the nonspecific DNA-binding activity of SSRPl and FACT (facilitating chromatin-mediated transcription) complex (Li et al., 2005). CK2 phosphorylation of the c-Myb nuclear oncoprotein at serines 11 and 12 results in reduced DNA binding (Liischer et al., 1990). (c) CK2 phosphorylation can modulate protein-protein interaction. CK2 phosphorylation of TFIIF-associated CTD phosphatase (FCPl) enhances the interaction between TFIIF subunit RAP74 and FCPl (Abbott et al., 2005). The trans-activation properties of some transcription factors are also regulated by CK2-mediated phosphorylation through influencing the recruitment of coregulators and/or RNA polymerase to the promoter. For instance, CK2 also targets the N-terminal transcriptional activation domain of CHOP/GADD153 (C/EBP homologous protein transcription factor/growth arrest and DNA damage inducible protein) for phosphorylation and downregulates its transcription activity (Ubeda and Habener, 2003). Normally, the zinc- finger transcription factor CTCF (CCCTC-binding factor) acts as a transcriptional repressor, but CK2 phosphorylation at serine 612 switches CTCF to an activator (El- Kady and Klenova, 2005). Phosphorylation by CK2 on threonine 142 of HSFl promotes trans-activation of the HSP70 gene (Soncin et al., 2003). However, CK2 phosphorylation 27 at the C-terrninal serine 386 of p53 is critical for p53-dependent repression (Hall et al., 1 996). (e) CK2 phosphorylation is also important for regulating the level of a transcription factor by either stabilizing it or marking it for destruction. For example, CK2 phosphorylation of Max at serine ll protects Max from caspase-5 mediated degradation (Krippner-Heidenreich et al., 2001). Ubiquitin-directed proteasomal degradation of the c-Myc oncoprotein is protected by CK2 phosphorylation in its C- terminal proline-glutamic acid-serine-threonine (PEST) domain (Channavajhala and Seldin, 2002). In contrast, CK2 phosphorylation of IKBG. at serine 283 and threonines 291 and 299 in its C-terrninal PEST domain accelerates its degradation, which is important for NFKB nuclear translocation and transcription activation (Lin et al., 1996; McElhinny et al., 1996). 2.3.1.2. CK2 in chromatin remodeling processes Evidence for CK2 function in chromatin remodeling processes is increasing. Firstly, histone cytoplasmic-nuclear translocation is influenced by CK2 phosphorylation. CK2 targets a histone chaperone protein called nucleosome assembly protein 2 (NAP-2) for phosphorylation and prevents histones/NAP-Z complex transport from the cytoplasm into the nucleus (Rodriguez et al., 2000). Secondly, chromatin associations and/or chromatin-related protein-protein interactions are affected by CK2-mediated phosphorylation. For example, a chromatin-associated phosphotyrosine phosphatase 28 PTP-SZ phosphorylation by CK2 results in its dissociation from chromatin (Nambirajan et al., 2000). DNA binding activity of the human chromatin protein DEK is also downregulated by CK2 phosphorylation (Kappes et al., 2004). Histone deacetylases (HDAC) 1 and HDACZ phosphorylation by CK2 increases its binding affinity to interacting partners, including the Rb-associated protein (RbAp48), MTA-2 (multithreaded architecture), mSin3A, and CoREST (a corepressor for the repressor element 1 silencing transcription factor) (Pflum et al., 2001; Tsai and Seto, 2002; Sun et al., 2002). In addition, HDACI phosphorylation at serine 421 and 423 by CK2 promotes its enzymatic activity (Pflum et al., 2001), which is proposed to alter the balance of histone (dc-)acetylation of its target genes. Furthermore, several high mobility group nonhistone chromosomal proteins (HMGs) and the heterochromatin-associated protein 1 (HPl) are phosphorylated by CK2, which then affect their DNA binding and their interaction with specific transcription factors (Wisniewski et al., 1999; Zhao et al., 2001; Krohn et al., 2002). Thirdly, CK2 function in chromatin remodeling processes is fiirther supported by the observation that the level of CK2 is higher in the active than in inactive nucleosomes from normal prostate (Guo et al., 1998), and that CK2 co-localizes with productively transcribing RNA pol II on polytene chromosomes of Chironomus salivary gland cells (Egyhazi et al., 1999). CK2 can be recruited to the promoter by either complexing with histones and general chromatin remodeling factors (Gavin et al., 2002; Ho et al., 2002; Krogan et al., 2002), or interacting with other transcription factors such as ATFl, CREB, c-Fos or c-Jun through the basic leucine-zipper domains (Yamaguchi et al., 1998). Thus, CK2 may participate in promoting the conformational transition of 29 inactive nucleosomes to the active form and/or in the function of transcriptionally active nucleosomes. 2.3.2. The role of CK2 in protein-coding gene expression Numerous lines of evidence have established that CK2 plays an important role in regulating protein-coding gene transcription by RNA pol II. A comparative genome-wide expression analysis of Saccharomyces cerevisiae CK2 deletion strains and corresponding wild types were performed by Barz and coworkers (2003). Their studies showed that of roughly 900 gene products proposed to be involved in cell cycle regulation, the expression of 283 protein-coding genes depend on or are affected by CK2 (Barz et al. 2003). CK2 not only phosphorylates a large number of transcription factors or effectors of DNA/RNA structure or translational elements (Meggio and Pinna, 2003), which are important for regulating protein-coding gene expression, but also directly targets the basal transcriptional machinery. For example, CK2 phosphorylates two subunits of RNA pol 11 [214,000 and 20,500 Daltons (Da)], and these modifications are required for transcriptional activity (Dahmus, 1981). The general transcription factors TFIIA, TFIIE, and TFIIF are also phosphorylated by CK2. Pre-initiation complex (PIC) formation on the Ad-MLP promoter was stimulated by CK2 phosphorylation of TFIIA, TFIIF, and RNA pol 11. However, the mRNA synthesis was dramatically inhibited by CK2 phosphorylation of RNA pol II (Cabrejos et al., 2004). One possibility is that CK2 phosphorylation of RNA pol 11 increases the formation of a PIC but inhibits RNA pol [1 recycling. Indeed, CK2 also fimctions to recruit a CTD-specific phosphatase FCPl to the transcriptional machinery to facilitate RNA pol II recycling. CK2 phosphorylation of 30 F CPI stimulates its phosphatase activity and enhances the interaction between TFIIF subunit RAP74 and PCP] (Egyhazi et al., 1999; Abbott et al., 2005). CK2 has been shown to associate with the RNA pol II elongation complex (Krogan et al., 2002). Also, a study with TATA-less DPE (downstream promoter element)-dependent transcription indicates that CK2 along with the coactivator PC4 is required for DPE-specific transcription (Lewis et al., 2005). Moreover, activation of pre-mRNA splicing by human RNPS] (RNA-binding protein prevalent during S phase) is regulated by CK2 phosphorylation (Trembley et al., 2005). Taken together, these data strongly suggest that CK2 plays a role in regulating protein coding gene expression. 2.3.3. The role of CK2 in non-coding gene transcription Non-coding RNAs trancribed by RNA polymerases I, II, and III play important catalytic and structural roles in the RNA processing and protein synthesis. Research from numerous labs shows that CK2 is involved in controlling transcription of non-coding genes, such as RNA pol I-specific rRNA (Saez-Vasquez et al., 2001), RNA pol III- specific tRNA and SS rRNA (Hockman and Schultz, 1996; Ghavidel et al., 1999; Johnston et al., 2002), and RNA pol III-specific U6 snRNA (Hu et al., 2003; Hu et al., 2004) RNA pol l holoenzyme is a multi-subunit complex with at least 2MD in mass and is composed of 30 or more polypeptides (Seither et al., 1998; Albert et al., 1999; Hannan etal.,1999). RNA pol I transcribes the tandemly repeated genes that encode the precursor of 188, 5.68 and 28S rRNAs. Initially, transcription factor UBF (Upstream Binding Factor) binds to upstream promoter element (UPE), and SLI/T IF -IB (promoter selectivity 31 factor or transcription initiation factor IB) recognizes the core promoter element (CPE). SL-l and UBF function cooperatively in the formation of the initiation complex which then facilitates RNA pol I transcription. It has been reported that CK2 copurifies with epitope-tagged RNA pol 1 from mammalian cells (Harman et al., 1998). Also, the CK2 activity associates stably with the rRNA promoter in plants (Saez-Vasquez et al., 2001). CK2 targets the key DNA-binding transcription factor UBF in the C-terrninal hyperacidic tail, which is essential for transactivation (O’Mahony et al., 1992; Voit et al., 1992; Voit etaL,l995) CK2 is also involved in RNA pol III transcription of tRNA and SS rRNA (Hockman and Schultz, 1996; Ghavidel et al., 1999; Johnston et al., 2002). The general transcriptipn factors TF 111A, Brfl -specific TFIIIB, and TFIIIC are required for tRNA and 5S rRNA gene transcription by RNA pol 111. It has been reported that endogenous human CK2 associates stably with TFIIIB and phosphorylates the BRF component of Brfl- specific TFIIIB (Johnston et al., 2002). CK2 can also target the TBP subunit of Brfl- specific TFIIIB for phosphorylation (Ghavidel and Schultz, 1997; Ghavidel et al., 1999; Ghavidel and Schultz, 2001). Furthermore, recent studies showed that RNA pol III- specific U6 snRNA gene transcription is also regulated by CK2 (Hu et al., 2003, Hu et al., 2004). CK2 occupies U6 snRNA gene promoters, and CK2 phosphorylation of the RNA pol III holoenzyme stimulates U6 transcription (Hu et al., 2003). CK2 also targets the de1 component of Ber-specific TFIIIB complex during mitosis. Phosphorylated del, in turn, disassociates from the U6 promoter, and thus downregulating U6 transcription in a cell cycle dependent fashion (Hu et al., 2004). Taken together, these data strongly 32 suggest that CK2 plays an important role in regulating non-coding gene transcription by RNA pol I and pol 111. As mentioned previously, some non-coding snRNA genes are transcribed by RNA pol 11. Whether CK2 plays a role in regulating RNA pol II-specific snRNA gene transcription is unknown. Because CK2 appears to globally regulate non-coding gene transcription and human U1 and U6 snRNA genes share similar promoter elements and their gene products have related functions, it is reasonable to hypothesize that CK2 also regulates U1 snRNA gene transcription by RNA pol 11. Thus, the major focus of this thesis has been to investigate the role of CK2 in U1 snRNA gene transcription. 3. Main hypothesis and experimental design The main objective of this doctoral thesis research is to explore the role of CK2 for snRNA gene transcription and to investigate the mechanism of CK2 regulation. To directly test this idea, CK2 promoter occupancy was detected by a chromatin immunoprecipitation (ChIP) assay. Nascent U1 snRNA transcripts in Hela cells were monitored by reverse transcriptase polymerase chain reaction (RT-PCR) following endogenous CK2 depletion by RNAi. U1 snRNA gene transcription in the presence of CK2 and/or CK2 inhibitors was employed in vitro to understand the mechanism of CK2 regulation. The general transcription factor SNAPC was identified as a potential CK2 target by an in vitro kinase assay and mass spectrometry (MS). The ability of SNAPc to bind DNA and to recruit TBP in the presence of CK2 was tested using an electrophoretic mobility shift assay (EMSA). 33 In the current studies, we demonstrated that CK2 downregulates U1 snRNA gene transcription by RNA pol II. Decreased levels of endogenous CK2 correlates with increased U1 expression while CK2 associates with U1 gene promoters, indicating that it plays a direct role in U1 gene regulation. CK2 phosphorylates the general transcription factor SNAPc that is required for both RNA pol II and III transcription. SNAPC phosphorylation by CK2 inhibits binding to snRNA gene promoters. However, restricted promoter access by phosphorylated SNAPC can be overcome by cooperative interactions with TBP at the U6 promoter but not at a U1 promoter. We have further determined that within SNAP190, CK2 phosphorylates the N-terminal half of SNAP190 at two regions that contain multiple consensus CK2 sites (amino acid 20-63 and 518-557) by mass spectrometric analysis. To test whether these regions are important for SNAPC function, partial SNAPC complexes containing full length SNAP43, full length SNAPSO, and various truncated SNAP190 molecules [SNAP190 (1-719), SNAP190 (63-719), SNAP190 (1-505), or SNAP190 (63-505)] were co-expressed and tested for DNA binding and function in transcription. All four complexes maintain normal DNA binding activity in electrophoretic mobility shifi assays. Furthermore, these complexes all support U1 and U6 snRNA gene transcription using SNAPC depleted HeLa nuclear extracts in vitro, indicating that these regions are not essential for SNAPC activity. However, the SNAP190 (1-719)-containing SNAPC complex shows less DNA binding and transcription activity upon CK2 treatment, while the SNAP190 (1-505)-containing SNAPC complex shows no change under this condition. Thus, SNAP190 phosphorylation by CK2 plays a negative regulatory role on SNAPC function. 34 References Abbott, K.L, Renfrow, M.B., Chalmers, M.J., Nguyen, B.D., Marshall, A.G., Legault, P., Omichinski, J.G. (2005). Enhanced binding of RNAP ll CTD phosphatase F CPI to RAP74 following CK2 phosphorylation. Biochemistry 44, 2732-2745. Abramczyk, 0., lion, P., Zielinski, R., Pilecki, M., Hellman, U., Szyszka, R. (2003). The protein kinase 60S is a free catalytic CK2alpha' subunit and forms an inactive complex with superoxide dismutase SODl. Biochem Biophys Res Commun 307, 31-40. Ackerman, P., and Osheroff, N. (1989). Regulation of casein kinase II activity by epidermal growth factor in human A-431 carcinoma cells. J Biol Chem 264, 11958-11965. Ackerman, P., Glover, C. V., and Osheroff, N. (1990). Stimulation of Casein Kinase II by Epidermal Growth Factor, Relationship Between the Physiological Activity of the Kinase and the Phosphorylation State of its [3 Subunit. Proc Natl Acad Sci U S A 87, 821- 825. Ahmed, K., Gerber, D.A., and Cochet, C. (2002). Joining the cell survival squad, An emerging role for protein kinase CK2. Trends in Cell Biology 12, 226-230. Albet, A.C., Denton, M., Kermekchiev, M., Pikaard, CS. (1999). Histone acetyltransferase and protein kinase activities copurify with a putative Xenopus RNA polymerase I holoenzyme self-sufficient for promoter-dependent transcription. Mol Cell Biol 19, 796-806. Allende, J.B., Allende, CC. (1995). Protein kinases. 4. Protein kinase CK2, an enzyme with multiple substrates and a puzzling regulation. F ASEB J 9, 313-23. Appel, K., Wagner, P., Boldyreff, B., Issinger, O.G. and Montenarh, M. (1995). Mapping of the interaction sites of the growth suppressor protein p53 with the regulatory beta- subunit of protein kinase CK2. Oncogene 11, 1971 1978. Ares, M. Jr., Chung, J.S., Giglio, L., Weiner, A.M. (1987). Distinct factors with Spl and NF-A specificities bind to adjacent functional elements of the human U2 snRNA gene enhancer. Genes Dev 8, 808-817. Bai, L., Wang, Z., Yoon, J.B., Roeder, RC. (1996). Cloning and characterization of the beta subunit of human proximal sequence element-binding transcription factor and its involvement in transcription of small nuclear RNA genes by RNA polymerases II and 111. Mol Cell Biol 16, 5419-5426. Bartkiewicz, M., Gold, H. A., and Altman, S. (1989). Identification and characterization of an RNA molecule that copurifies with RNase P activity from HeLa cells. Genes Dev 3, 489-499. 35 Barz, T., Ackermann, K., Dubois, G., Eils, R., Pyerin, W. (2003). Genome-wide expression screens indicate a global role for protein kinase CK2 in chromatin remodeling. J Cell Sci 116, 1563-1577. Bibby, A.C., Litchfield, D.W. (2005). The Multiple Personalities of the Regulatory Subunit of Protein Kinase CK2, CK2 Dependent and CK2 Independent Roles Reveal a Secret Identity for CK2beta. Int J Biol Sci 1, 67-79. Bidwai, A. P., Reed, J. C., Glover, C. V.C. (1995). Cloning and Disruption of CKBI, the Gene Encoding the 38-kDa B Subunit of Saccharomyces cerevisiae Casein Kinase II (CKII). Deletion of CKII Regulatory Subunits Elicits a Salt-Sensitive Phenotype. J Biol Chem 270, 10395-10404 Bjorling-Poulsen, M., Siehler, S., Wiesmuller, L., Meek, D., Niefind, K., Issinger, O.G. (2005). The 'regulatory' beta-subunit of protein kinase CK2 negatively influences p53- mediated allosteric effects on Chk2 activation. Oncogene 2005 May 23, [Epub ahead of print] Blaydes, J.P., HUpp, TR. (1998). DNA damage triggers DRB-resistant phosphorylation of human p53 at the CK2 site. Oncogene 17, 1045 1052. Bohana-Kashtan, O., Pinna, L.A., Fishelson, Z. (2005). Extracellular phosphorylation of C9 by protein kinase CK2 regulates complement-mediated lysis. Eur J Immunol 35, 1939-1948. Bohmann, D. (1990). Transcription factor phosphorylation, a link between signal transduction and the regulation of gene expression. Cancer Cells 2, 337 344. Boldyreff, B., Issinger, O.G. (1997). A-Raf kinase is a new interacting partner of protein kinase CK2 beta subunit. F EBS Lett 403, 197-199. Boldyreff, B., Mietens, U., Issinger, O.G. (1996). Structure of protein kinase CK2, dimerization of the human beta-subunit. FEBS Lett 379, 153 156. Bonnet, H., F ilhol, O., Truchet, 1., Brethenou, P., Cochet, C., Amalric, F. and Bouche, G. (1996). Fibroblast growth factor-2 binds to the regulatory beta subunit of CK2 and directly stimulates CK2 activity toward nucleolin. J Biol Chem 271, 24781 24787. Brenneisen, P., Wlaschek, M., Schwambom, E., Schneider, L.A., Ma, W., Sies, H., Scharffetter-Kochanek, K. (2002). Activation of protein kinase CK2 is an early step in the ultraviolet B-mediated increase in interstitial collagenase (matrix metalloproteinase-l , MMP-1) and stromelysin-l (MMP-3) protein levels in human dermal fibroblasts. Biochem 365, 31 40. 36 Bronner, C., Trotzier, M.A., Filhol, O., Cochet, C., Rochette-Egly, C., Scholler-Guinard, M., Klein, J.P., Mousli, M. (2004). The antiapoptotic protein ICBP90 is a target for protein kinase 2. Ann N Y Acad Sci 1030, 355-60. Burge, C.B., Tuschl, T., Sharp, RA. (1999). Splicing of precursors to mRNAs by the spliceosomes. In, RF Gesteland, TR Cech and JF Atkins, eds, The RNA World (2nd Edition ed.), Cold Spring Harbor Laboratory Press, Cold Spring Harbor (1999), pp. 525 560. Burnett, G., Kennedy, ER (1954). The enzymatic phosphorylation of proteins. J Biol Chem 211, 969-980. Cabart, P., Murphy, S. (2001). BRFU, a TFIIB-like Factor, Is Directly Recruited to the TATA-box of Polymerase 111 Small Nuclear RNA Gene Promoters through Its Interaction with TATA-binding Protein. J Biol Chem 276, 43056-43064. Cabrejos, M. E., Allende, C. C, Maldonado, E. (2004). Effects of phosphorylation by protein kinase CK2 on the human basal components of the RNA polymerase II transcription machinery. J Cell Biochem 93, 2-10. Caceres, J.F., McKenzie, D., Thimmapaya, R., Lund, E., Dahlberg, J.E. (1992). Control of mouse Ula and Ulb snRNA gene expression by differential transcription. Nucleic Acids Res 20, 4247-4254. Canton, D.A., Zhang, C., Litchfieldi, D.W. (2001). Assembly of protein kinase CK2, investigation of complex formation between catalytic and regulatory subunits using a zinc-finger-deficient mutant of CKZB. Biochem J 358, 87-94. Channavajhala, P., Seldin, D. C. (2002). Functional interaction of protein kinase CK2 and c-Myc in lymphomagenesis. Oncogene 21, 5280 5288. Chantalat, L., Leroy, D., Filhol, O., Nueda, A., Benitez, M.J., Chambaz, E.M., Cochet, C., Dideberg, O. (1999). Crystal structure of the human protein kinase CK2 regulatory subunit reveals its zinc finger-mediated dimerization. EMBO J 18, 2930 2940. Chen, R., Yang, Z., Zhou, Q. (2004). Phosphorylated positive transcription elongation factor b (P-TEFb) is tagged for inhibition through association with 78K snRNA. J Biol Chem 279, 4153-4160. Cheng, Y., Lund, E., Kahan, B.W., Dahlberg, J.E. (1997). Control of mouse U1 snRNA gene expression during in vitro differentiation of mouse embryonic stem cells. Nucleic Acids Res 25, 2197-54204. Cheung, W.L., Turner, F.B., Krishnamoorthy, T., Wolner, B., Ahn, S.H., Foley, M., Dorsey, J.A., Peterson, C.L., Berger, S.L., Allis, CD. (2005). Phosphorylation of histone 37 H4 serine 1 during DNA damage requires casein kinase II in S. cerevisiae. Curr Biol 15, 656-60. Collinge, M.A., Walker, JC. (1994). Isolation of an Arabidopsis thaliana casein kinase 11 beta subunit by complementation in Saccharomyces cerevisiae. Plant Mol Biol 25, 649- 658. Dahmus, ME. (1981). Purification and Properties of Calf Thymus Casein Kinases I and II. J Biol Chem 256, 3332-3339. Damania, B., Mital R, Alwine JC. (1998). Simian virus 40 large T antigen interacts with human TFIIB-related factor and small nuclear RNA-activating protein complex for transcriptional activation of TATA-containing polymerase III promoters. Mol Cell Biol 18,1331-1338. Davies, S.P., Reddy, H., Caivano, M., Cohen, P. (2000). Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J 351, 95-105. Desagher, S., Osen-Sand, A., Montessuit, S., Magnenat, E., Vilbois, F ., Hochmann, A., Joumot, L., Antonsson, B., Martinou, JC. (2001). Phosphorylation of bid by casein kinases I and II regulates its cleavage by caspase 8. Mol Cell 8, 601 611. Diana, C. B., Pombo, A., Murphy, S. (2003). Interaction of proteins with promoter elements of the human U2 snRNA genes in vivo. Gene 315, 103-112. Diaz-Nido, J ., Mizuno, K., Nawa, H., Marshak, DR. (1994). Regulation of protein kinase CK2 isoforrn expression during rat brain development. Cell Mol Biol Res 40, 581-585. Egyhazi, E., Ossoinak, A., F ilhol-Cochet, O., Cochet, C., Pigon, A. (1999). The binding of the alpha subunit of protein kinase CK2 and RAP74 subunit of TFIIF to protein-coding genes in living cells is DRB sensitive. Mol Cell Biochem 191, 149-59. El-Kady, A., Klenova, E. (2005). Regulation of the transcription factor, CTCF, by phosphorylation with protein kinase CK2. F EBS Lett 579, 1424-34. Escalier, D., Silvius, D., Xu, X. (2003). Sperrnatogenesis of mice lacking CKZalpha', failure of germ cell survival and characteristic modifications of the sperrnatid nucleus. Mol Reprod Dev 66, 190-201. Fatica, A., Tollervey, D. (2002). Making ribosomes, Curr Opin Cell Biol 14, 313 318. Faust, M., Montenarh, M. (2000). Subcellular localization of protein kinase CK2, A key to its function. Cell Tissue Res 301, 329-340. 38 Faust, R.A., Niehans, G., Gapany, M., Hoistad, D., Knapp D., Cherwitz, D., Davis, A., Adams, G.L., Ahmed, K. (1999). Subcellular immunolocalization of protein kinase CK2 in normal and carcinoma cells. Int J Biochem Cell Biol 31, 941 949. Filhol, O., Baudier, J., Delphin, C., Loue-Mackenbach, P., Chambaz, E. and Cochet, C. (1992). Casein kinase II and the tumor suppressor protein p53 associate in a molecular complex that is negatively regulated upon p53 phosphorylation. J Biol Chem 267, 20577 20583. Ford, E., Strubin, M., Hernandez, N. (1998). The Oct-1 POU domain activates snRNA gene transcription by contacting a region in the SNAPC largest subunit that bears sequence similarities to the Oct-l coactivator OBF -1. Genes Dev 12, 3528-3540. Fritz, G., Kaina, B. (1999). Phosphorylation of the DNA repair protein APE/REF-l by CKII affects redox regulation of AP-l. Oncogene 18, 1033 1040. Gavin, A. C., Bosche, M., Krause, R., Grandi, P., Marzioch, M., Bauer, A., Schultz, J., Rick, 1. M., Michon, A. M. et al. (2002). Functional organization of the yeast proteome by systematic analysis of protein complexes. Nature 415,141-147. Gerber, D.A., Souquere-Besse, S., Puvion, F ., Dubois, M.F., Bensaude, O., Cochet, C. (2000). Heat-induced relocalization of protein kinase CK2. Implication of CK2 in the context of cellular stress. J Biol Chem 275, 23919 23926. Ghavidel, A., Hockman, D.J., Schultz, MC. (1999). A review of progress towards elucidating the role of protein kinase CK2 in pol III transcription, regulation of the TATA binding protein. Mol Cell Biochem 191, 143-148. Ghavidel, A., Schultz, MC. (1997). Casein kinase II regulation of yeast TFIIIB is mediated by the TATA-binding protein. Genes Dev 11, 2780-2789. Ghavidel, A., Schultz, M. C. (2001). TATA binding protein-associated CK2 transduces DNA damage signals to the RNA polymerase III transcriptional machinery. Cell 106, 575-584. Gietz, R.D., Graham, K.C., Litchfield, D.W. (1995). Interactions between the subunits of casein kinase II. J Biol Chem 270, 13017 13021. Glibetic, M., Larson, D.E., Sienna, N., Bachellerie, J.P., Sells, B.H. (1992). Regulation of U3 snRNA expression during myoblast differentiation. Exp Cell Res 202, 183-189. Gdtz, C., Scholtes, P., Prowald, A., Schuster, N., Nastainczyk,W., Montenarh, M. (1999). Protein kinase CK2 interacts with a multi-protein binding domain of p53. Mol Cell Biochem 191, 111-120. 39 Gdtz, C., Wagner, P., Issinger, O.G., Montenarh, M. (1996). p21WAF1/CIP1 interacts with protein kinase CK2. Oncogene. 13, 391-398. Graham, K.C., Litchfield, D.W. (2000). The regulatory beta subunit of protein kinase CK2 mediates formation of tetrameric CK2 complexes. J Biol Chem 275, 5003 5010. Grandi, P., Rybin, V., Bassler, J., Petfalski, E., Strauss, D. Marzioch, M., Schafer, T., Kuster, B., Tschochner, H., Tollervey, D., et al. (2002). 908 pre-ribosomes include the 358 pre-rRNA, the U3 snoRNP, and 408 subunit processing factors but predominantly lack 6OS synthesis factors, Mol Cell 10, 105 115. Gridasova, A.A., Henry, R.W. (2005). The p53 Tumor Suppressor Protein Represses Human snRNA Gene Transcription by RNA Polymerases II and III Independently of Sequence-Specific DNA Binding. Mol Cell Biol 25, 3247-3260. Guerra, B., Issinger, O.G. (1999). Protein kinase CK2 and its role in cellular proliferation, development and pathology. Electrophoresis. 20, 391-408. Guerra, B., Siemer, S., Boldyreff, B., Issinger, O.G. (1999). Protein kinase CK2, evidence for a protein kinase CKZB subunit fraction, devoid of the catalytic CK2a subunit, in mouse brain and testicles. FEBS Lett 462, 353-357. Guo, C., Davis, A.T., Yu, S., Tawfic, S., Ahmed, K. (1999). Role of protein kinase CK2 in phosphorylation nucleosomal proteins in relation to transcriptional activity. Mol Cell Biochem. 191, 135-142. Guo, C., Yu, S., Davis, A.T., Wang, H., Green, J.E. Ahmed, K. (2001). A potential role of nuclear matrix-associated protein kinase CK2 in protection against drug-induced apoptosis in cancer. cells. J Biol Chem 276, 5992-5999. Guo, C., Davis, A. T., Ahmed, K. (1998). Dynamics of protein kinase CK2 association with nucleosomes in relation to transcriptional activity. J Biol Chem 273, 13675-13680. Hagemann, C., Kalmes, A., Wixler, V., Wixler, L., Schuster, T., Rapp, U.R. (1997). The regulatory subunit of protein kinase CK2 is a specific A-Raf activator. F EBS Lett 403, 200-202. Hall, S.R., Campbell, L.E., Meek, D.W. (1996). Phosphorylation of p53 at the casein kinase 11 site selectively regulates p53-dependent transcriptional repression but not transactivation. Nucleic Acids Res 24, 1119-26 Han, M.H., Han, D.K., Aebersold, R.H., Glomset, J.A. (2001). Effects of protein kinase CK2, extracellular signal-regulated kinase 2, and protein phosphatase 2A on a phosphatidic acid-preferring phospholipase A1. J Biol Chem 276, 27698 27708. 40 Hanna, D.E., Rethinaswamy, A., Glover, C.V.C. (1995). Casein kinase II is required for cell cycle progression during G, and Gle in Saccharomyces cerevisiae. J Biol Chem 270, 25905-25914. Harman, R.D., Cavanaugh, A., Hempel, W.M., Moss, T., Rothblum, L. (1999). Identification of a mammalian RNA polymerase I holoenzyme containing components of the DNA repair/replication system. Nucleic Acids Res 27, 3720-3727. Hannan, R.D., Hempel, W.M., Cavanaugh, A., Arino, T., Dimitrov, 8.1., Moss, T., Rothblum, L. (1998). Affinity purification of mammalian RNA polymerase 1. Identification of an associated kinase. J Biol Chem 273, 1257-1267. Hathaway, G.M., Traugh, J .A. (1983). Casein kinase 11. Methods Enzymol. 99, 317-31. Hehn, E.M., Young, A.V., Pelech, S.L., Sanghera, J.S., Shah, R.M.(1997). Developmental alterations in casein kinase 2 activity during the morphogenesis of quail secondary palate. Anat Rec 247, 102-108. Henry, R.W., Ford, E., Mital, R., Mittal, V., Hernandez, N. (1998a). Crossing the line between RNA polymerases, transcription of human snRNA genes by RNA polymerases II and 111. Cold Spring Harb Symp Quant Biol 63, 1 1 1-120. Henry, R.W., Ma, 8., Kobayashi, R., Hernandez, N. (1998b). SNAP19 mediates the assembly of a functional core promoter complex (SNAPC) shared by RNA polymerases II and III. Genes Dev 12, 2664-2672. Henry, R.W., Mittal, V., Ma, 3., Sadowski, C.L., Kobayashi, R., Hernandez, N. (1996). Cloning and characterization of SNAPSO, a subunit of the snRNA-activating protein complex SNAPC. EMBO J 15, 7129-7136. Henry, R.W., Sadowski, C.L., Kobayashi, R., Hernandez, N. (1995). A TBP-TAF complex required for transcription of human snRNA genes by RNA polymerases II and 111. Nature 374, 653-657. Hernandez, N., Lucito, R. (1988). Elements required for transcription initiation of the human U2 snRNA gene coincide with elements required for snRNA 3' end formation. EMBO J 7, 3125-3134. Hernandez, N. (2001). Small nuclear RNA genes, a model system to study fundamental mechanisms of transcription. J Biol Chem 276, 26733-26736. Hilgard, P., Czaja, M.J., Gerken, G., Stockert, RJ. (2004). Proapoptotic function of protein kinase CK2alpha" is mediated by a JNK signaling cascade. Am J Physiol Gastrointest Liver Physiol 287, G] 92-201. 41 Hilgard, P., Huang, T., Wolkoff, A.W., Stockert, R,J. (2002). Translated Alu sequence determines nuclear localization of a novel catalytic subunit of casein kinase 2. Am J Physiol Cell Physiol 283, 472 483. Hinkley, C.S., Hirsch, H.A., Gu, L., LaMere, 8., Henry, R.W. (2003). The Small Nuclear RNA-activating Protein 190 Myb DNA Binding Domain Stimulates TATA Box-binding Protein-TATA Box Recognition. J Biol Chem 278, 18649-18657. Hirsch, H.A., Gu, L., Henry, R.W. (2000). The retinoblastoma tumor suppressorprotein targets distinct general transcription factors to regulate RNA polymerase III gene expression. Mol Cell Biol 20, 9182-9191. Hirsch, H.A., Jawdekar, G.W., Lee, K.A., Gu, L., Henry, R.W. (2004). Distinct mechanisms for repression of RNA polymerase III transcription by the retinoblastoma tumor suppressor protein. Mol Cell Biol 24, 5989-5999. Ho, Y., Gruhler, A., Heilbut, A., Bader, G. D., Moore, L., Adams, S. L., Millar, A., Taylor, P., Bennett, K. et a1. (2002). Systematic identification of protein complexes in Saccharomyces cerevisiae by mass spectrometry. Nature 415,180-183. Hockman, D.J., Schultz, M.C. (1996). Casein kinase II is required for efficient transcription by RNA polymerase 111. Mol Cell Biol 16, 892-898. Hoffman, R.W., Gazitt, T., Foecking, M.F., Ortmann, R.A., Misfeldt, M., Jorgenson, R., Young, S.L., Greidinger, EL. (2004). U1 RNA induces innate immunity signaling. Arthritis Rheum 50, 2891-2896. Hovde, S., Hinkley, C.S., Strong, K., Brooks, A., Gu, L., Henry, R.W., Geiger, J. (2002). Activator recruitment by the general transcription machinery, X-ray structural analysis of the Oct-l POU domain/human U1 octamer/SNAP190 peptide ternary complex. Genes Dev 16, 2772-2777. Hu, P., Samudre, K., Wu, S., Sun, Y., Hemandez, N. (2004). CK2 phosphorylation of del executes cell cycle-specific RNA polymerase III transcription repression. Mol Cell 16, 81-92. Hu, P., Wu, 8., Hernandez, N. (2003). A minimal RNA polymerase III transcription system from human cells reveals positive and negative regulatory roles for CK2. Mol Cell 12, 699-709. Hu, P., Wu, 8., Hernandez, N. (2004). A role for beta-actin in RNA polymerase III transcription. Genes Dev 18, 3010-3015. Issinger, O.G. (1993). Casein kinases, pleiotropic mediators of cellular regulation. Pharrnacol Ther 59, 1-30. 42 Jakobi, R., Voss,H., Pyerin,W.(1989). Human phosvitin/casein kinase type 11. Molecular cloning and sequencing of full-length cDNA encoding subunit beta. Eur J Biochem 183, 227-233. Johnston, I.M., Allison, S.J., Morton, J.P., Schramm, L., Scott, P.H., White, RJ. (2002). CK2 forms a stable complex with TFIIIB and activates RNA polymerase III transcription in human cells. Mol Cell Biol 22, 3757-3768. Kappes, F ., Damoc, C., Knippers, R., Przybylski, M., Pinna, L.A., Gruss, C. (2004). Phosphorylation by protein kinase CK2 changes the DNA binding properties of the human chromatin protein DEK. Mol Cell Biol 24, 6011-6020. Karin, M. (1994). Signal transduction from the cell surface to the nucleus through the phosphorylation of transcription factors. Curr Opin Cell Biol 6, 415 424. Kassavetis, G.A., Joazeiro, C.A., Pisano, M., Geiduschek, E.P., Colbert, T., Hahn, S., Blanco, J .A. (1992). The role of the TATA-binding protein in the assembly and function of the multisubunit yeast RNA polymerase III transcription factor, TFIIIB. Cell 71, 1055- 1064. Keller, D. M., Zeng, X., Wang, Y., Zhang, Q. H., Kapoor, M.,. Shu, H., Goodman, R., Lozano, G., Zhao, Y., Lu, H. (2001). A DNA damage-induced p53 serine 392 kinase complex contains CK2, hSpt16, and SSRPl. Mol Cell 7, 283-292. Klarlund, J. K., Czech, M. P. (1988). Insulin-like grth factor I and insulin rapidly increase casein kinase II activity in BALB/c 3T3 fibroblasts. J Biol Chem 263, 15872- 15875 Kom, 1., Gutkind, S., Srinivasan, N., Blundell, T.L., Allende, C.C., Allende, J.E. (1999). Interactions of protein kinase CK2 subunits. Mol Cell Biochem 191, 75-83. Krippner-Heidenreich, A., Talanian, R.V., Sekul, R., Kraft, R., Thole, H., Ottleben, H., Lilscher, B. (2001). Targeting of the transcription factor Max during apoptosis, phosphorylation-regulated cleavage by caspase-5 at an unusual glutamic acid residue in position P1. Biochem J 358, 705-715. Krogan, N. J., Kim, M., Ahn, S. H., Zhong, G., Kobor, M. S., Cagney, G., Emili, A., Shilatifard, A., Buratowski, S., Greenblatt, J. F. (2002). RNA polymerase II elongation factors of Saccharomyces cerevisiae, a targeted proteomics approach. Mol Cell Biol 22, 6979-6992. Krohn, N. M., Yanagisawa, S., Grasser, K. D. (2002). Specificity of the stimulatory interaction between chromosomal HMGB proteins and the transcription factor Dof2 and its negative regulation by protein kinase CK2-mediated phosphorylation. J Biol Chem 277, 32438-32444. 43 Kudlicki, W., Grankowski, N., Gasior, E. (1978). Isolation and properties of two protein kinases from yeast which phosphorylate casein and some ribosomal proteins. Eur J Biochem 84, 493 498. Kuhlman, T.C., Cho, H., Reinberg, D., Hernandez, N. (1999). The general transcription factors 11A, 113, 11F, and IIE are required for RNA polymerase II transcription from the human U1 small nuclear RNA promoter. Mol Cell Biol 19, 2130-2141. Kunkel, G. R., Pederson, T. (1989). Transcription of a human U6 small nuclear RNA gene in vivo withstands deletion of intragenic sequences but not of an upstream TATATA box. Nucleic Acids Res 17, 7371-7379. Kwek, K.Y., Murphy, 8., F urger, A., Thomas, B., O'Gorman, W., Kimura, H., Proudfoot, N.J., Akoulitchev, A. (2002). U1 snRNA associates with TFIIH and regulates transcriptional initiation. Nat Struct Biol 9, 800-5. Landesman-Bollag, E., Romieu-Mourez, R., Song, D.H., Sonenshein, G.E., Cardiff, R.D., Seldin, DC. (2001). Protein kinase CK2 in mammary gland tumorigenesis. Oncogene 20, 3247 3257. Lasa, M., Marin, O., Pinna, LA. (1997). Rat liver Golgi apparatus contains a protein kinase similar to the casein kinase of lactating mammary gland. Eur J Biochem 243, 719- 725 Lebrin, F., Chambaz, E.M., Bianchini, L. (2001). A role for protein kinase CK2 in cell proliferation, evidence using a kinase-inactive mutant of CK2 catalytic subunit alpha. Oncogene. 20, 2010-2202. Lehner, B., Semple, J.I., Brown, S.E., Counsel], D., Campbell, R.D., Sanderson, CM. (2004). Analysis of a high-throughput yeast two-hybrid system and its use to predict the function of intracellular proteins encoded within the human MHC class III region. Genomics 83, 153-167. Leroy, D., Heriche, J. K., Filhol, O., Chambaz, E. M., Cochet, C. (1997). Binding of Polyamines to an Autonomous Domain of the Regulatory Subunit of Protein Kinase CK2 Induces a Conformational Change in the Holoenzyme. J Biol Chem 272, 20820 20827. Lewis, B.A., Sims, R.J. 3rd., Lane, W.S., Reinberg, D. (2005). Functional characterization of core promoter elements, DPE-specific transcription requires the protein kinase CK2 and the PC4 coactivator. Mol Cell 18, 471-481 Li, Y., Keller, D.M., Scott, J.D., Lu, H. (2005). CK2 phosphorylates SSRPl and inhibits its DNA-binding activity. J Biol Chem 280, 11869-11875. Li, D., Meier, T., Dobrowolska, G., Krebs, EC. (1997). Specific interaction between casein kinase 2 and the nucleolar protein Nopp140. J Biol Chem 272, 3773 3779. 44 Lieberman, S.L., Ruderrnan, J.V. (2004). CK2 beta, which inhibits Mos function, binds to a discrete domain in the N-terminus of Mos. Dev Biol 268, 271-279. Lin, R., Beauparlant, P., Makris, C., Meloche, S., Hiscott, J. (1996). Phosphorylation of IkappaBalpha in the C-terminal PEST domain by casein kinase II affects intrinsic protein stability. Mol Cell Biol 16, 1401-1409. Litchfield D.W. (2003). Protein kinase CK2, structure, regulation and role in cellular decisions of life and death. Biochem J 369, 1 15. Litchfield, D.W., Liischer, B., Lozeman, F.J., Eisenman, R.N., Krebs, E.G.(l992). Phosphorylation of casein kinase II by p34cdc2 in vitro and at mitosis. J Biol Chem 267, 13943-13951. Litchfield, D. W., Llischer, B. (1993). Casein kinase II in signal transduction and cell cycle regulation. Mol Cell Biochem 127-128, 187-199. Litchfield, D.W., Bosc, D.G., Canton, D.A., Saulnier, R.B., Vilk, G., Zhang, C. (2001). Functional specialization of CK2 isoforms and characterization of isoforrn-specific binding partners. Mol Cell Biochem 227, 21 29. Lobo, S.M., Hernandez, N. (1989). A 7 bp mutation converts a human RNA polymerase II snRNA promoter into an RNA polymerase III promoter. Cell 58, 55-67. Loizou, J.I., El-Khamisy, S.F., Zlatanou, A., Moore, D.J., Chan, D.W., Qin, J., Samo, S., Meggio, F ., Pinna, L.A., Caldecott, K.W. (2004). The protein kinase CK2 facilitates repair of chromosomal DNA single-strand breaks. Cell 117, 17 28. Lozeman, F.J., Litchfield, D.W., Piening, C., Takio, K., Walsh, K.A., Krebs, E.G.(l990). Isolation and characterization of human cDNA clones encoding the alpha and the alpha' subunits of casein kinase II. Biochemistry 29, 8436-8447. Lund, E., Bostock, C.J., Dahlberg, LE. (1987). The transcription of Xenopus laevis embryonic U1 snRNA genes changes when oocytes mature into eggs. Genes Dev 1, 47- 56. Ltischer, B., Christenson, E., Litchfield, D.W., Krebs, E.G., Eisenman, RN. (1990). Myb DNA binding inhibited by phosphorylation at a site deleted during oncogenic activation. Nature 344, 517 522. Ma, 3., Hernandez, N. (2001). A Map of Protein-Protein Contacts within the Small Nuclear RNA-activating Protein Complex SNAPC. J Biol Chem 276, 5027-5035. Ma, B., Hernandez, N. (2002). Redundant cooperative interactions for assembly of a human U6 transcription initiation complex. Mol Cell Biol 22, 8067-8078. 45 Marin, O., Meggio, F., Samo, S., Cesaro, L., Pagano, M.A., Pinna, L,A. (1999). Tyrosine Versus Serine/I‘hreonine Phosphorylation by Protein Kinase Casein Kinase-2, a study with peptide substrates derived from immunophilin fpr3. J Biol Chem 274, 29260-29265 Marin, O., Meggio, F., Marchiori, F ., Borin, G., Pinna, L.A. (1986). Site specificity of casein kinase-2 (TS) from rat liver cytosol. A study with model peptide substrates. Eur J Biochem 160, 239-244. Marshak, D. R., Russo, G. L. (1994). Regulation of protein kinase CKII during the cell division cycle. Cell Mol Biol Res 40, 513-517 Mattaj, I.W., Dathan, N.A., Parry, H.D., Carbon, P., Krol, A. (1988). Changing the RNA polymerase specificity of U snRNA gene promoters. Cell 55, 435-442. McElhinny, J.A., Trushin, S.A., Bren, G.D., Chester, N., Paya, CV. (1996). Casein kinase II phosphorylates I kappa B alpha at S-283, S-289, S-293, and T-291 and is required for its degradation. Mol Cell Biol 16, 899-906. Meggio, F., Pinna, L.A. (1984). Subunit structure and autophosphorylation mechanism of casein kinase-TS (type-2) from rat liver cytosol. Eur J Biochem 145, 593-599. Meggio, F ., Boldyreff, B., Marin, O., Marchiori, F., Perich, J.W., Issinger, O.G., Pinna, L.A. (1992). The effect of polylysine on casein-kinase-2 activity is influenced by both the structure of the protein/peptide substrates and the subunit composition of the enzyme. Eur J Biochem 205, 939-45. Meggio, F., Marin, O., Pinna, L.A. (1994). Substrate specificity of protein kinase CK2. Cell. Mol Biol Res 40, 401-409. Meggio, F., Negro, A., Samo, S., Ruzzene, M., Bertoli, A. (2000). Bovine prion protein as a modulator of protein kinase CK2. Biochem J 352, 191-196. Meggio, F ., Pinna, L. A. (2003). One-thousand-and-one substrates of protein kinase CK2? The FASEB Journal 17, 349-368. Meisner, H., Heller-Harrison, R., Buxton, J., Czech, MP. (1989). Molecular cloning of the human casein kinase II alpha subunit Biochemistry 28, 4072-4076. Meissner, W., Ahlers, A., Seifart, K.H. (1995). The activity of transcription factor PBP, which binds to the proximal sequence element of mammalian U6 genes, is regulated during differentiation of F9 cells. Mol Cell Biol 15, 5888-5897. Messenger, M.M., Saulnier, R.B., Gilchrist, A.D., Diamond, P., Gorbsky, G.J., Litchfield, D.W. (2002). Interactions between Protein Kinase CK2 and Pinl. evidence for phosphorylation-dependent interactions. J Biol Chem 277, 23054-23064. 46 Michels, A.A., Nguyen, V.T., Fraldi, A., Labas, V., Edwards, M., Bonnet, F ., Lania, L., Bensaude, O. (2003). MAQI and 78K RNA interact with CDK9/cyclin T complexes in a transcription-dependent manner. Mol Cell Biol 23, 4859-4869. Mittal, V., Cleary, M.A., Herr, W., Hernandez, N. (1996). The Oct-1 POU-specific domain can stimulate small nuclear RNA gene transcription by stabilizing the basal transcription complex SNAPC. Mol Cell Biol 16, 1955-1965. Mittal, V., Ma, 8., Hernandez, N. (1999). SNAPC, a core promoter factor with a built-in DNA-binding damper that is deactivated by the Oct-1 POU domain. Genes Dev 13, 1807-1821. Molkentin, J.D., Li, L., Olson, EN. (1996). Phosphorylation of the MADS-Box transcription factor MEFZC enhances its DNA binding activity. J Biol Chem 271, 17199- 17204. Munstermann, U., Fritz, G., Seitz, G., Yiping, L., Schneider, H.R., Issinger, O.G. (1990). CKII is elevated in solid tumors and rapidly proliferating non-neoplasic tissue. Eur J Biochem 189, 251 257. Murphy, 8., Yoon, J.B., Gerster, T., Roeder, RC. (1992). Oct-l and Oct-2 potentiate functional interactions of a transcription factor with the proximal sequence element of small nuclear RNA genes. Mol Cell. Biol 12, 3247 3261. Myslinski, E., Krol, A., Carbon, P. (1998). ZNF76 and ZNF143 are two human homologs of the transcriptional activator Staf. J Biol Chem 273, 21998-2006. Nambirajan, S., Radha, V., Kamatkar, S., Swarup, G. (2000). PTP-SZ, a nuclear tyrosine phosphatase, is phosphorylated and excluded from condensed chromosomes during mitosis. J Biosci 25, 33-40. Nash, M.A., Kozak, S.E., Angerer, L.M., Angerer, R.C., Schatten, H., Schatten, G., Marzluff, W.F. (1987). Sea urchin maternal and embryonic U1 RNAs are spatially segregated in early embryos. J Cell Biol 104, 1133-1142. Nguyen, V.T., Kiss, T., Michels, A.A., Bensaude, O. (2001). 78K small nuclear RNA binds to and inhibits the activity of CDK9/cyclin T complexes. Nature 414, 322-325. Niefind, K., Guerra, B., Ermakowa, 1., Issinger, O.G. (2001). Crystal structure of human protein kinase CK2, Insights into basic properties of the CK2 holoenzyme. EMBO J 20, 5320-5331. Niefind, K., Putter, M., Guerra, B., Issinger, O.G., Schomburg, D. (1999). GTP plus water mimic ATP in the active site of protein kinase CK2. Nat Struct Biol 6, 1100-1103. 47 O'Mahony, D.J., Smith, S.D., Xie, W., Rothblum, L]. (1992). Analysis of the phosphorylation, DNA-binding and dimerization properties of the RNA polymerase I transcription factors UBF] and UBF2. Nucleic Acids Res 20, 1301-1308. Ouchida, R., Kusuhara, M., Shimizu, N., Hisada, T., Makino, Y., Morimoto, C., Handa, H., Ohsuzu, F ., Tanaka, H. (2003). Suppression of NF-kappaB-dependent gene expression by a hexamethylene bisacetamide-inducible protein HEXIMl in human vascular smooth muscle cells. Genes Cells 8, 95-107. Peng, J., Zhu. Y., Milton. J.T., Price, DH. (1998). Identification of multiple cyclin subunits of human P-TEFb. Genes Dev 12, 755-762. Pepperkok, R., Lorenz, P., Pyerin, W. (1994). Casein kinase II is required for transition of GO/Gl, early GI, and 01/8 phases of the cell cycle. J Biol Chem 269, 6986-6991. Pflum, M. K., Tong, J. K., Lane, W. S., Schreiber, S. L. (2001). Histone Deacetylase 1 Phosphorylation Promotes Enzymatic Activity and Complex Formation J Biol Chem 276, 47733-47741. Pilecki, M., Grankowski, N., Jacobs, J., Gasior, E. (1992). Specific protein kinase from Saccharomyces cerevisiae cells phosphorylating 6OS ribosomal proteins. Eur J Biochem 206, 259-267. Pinna, L.A. (2002). Protein kinase CK2, a challenge to canons. Journal of Cell Science 115, 3873-3878 Pinna, L.A., Meggio, F. (1997). Protein kinase CK2 ("casein kinase-2") and its implication in cell division and proliferation. Prog Cell Cycle Res 3, 77-97. Porter, C.M., Havens, M.A., Clipstone, NA. (2000). Identification of amino acid residues and protein kinases involved in the regulation of NFATc subcellular localization. J Biol Chem 275, 3543-3551. Raman, C., Kuo, A., Deshane, J ., Litchfield, D.W., Kimberly, RP. (1998). Regulation of casein kinase 2 by direct interaction with cell surface receptor CD5. J Biol Chem 273, 19183 19189. Reed, J.C., Bidwai, A.P., Glover, C.V.C. (1994). Cloning and disruption of CKBZ, the gene encoding the 32-kDa regulatory beta'-subunit of Saccharomyces cerevisiae casein kinase II. J Biol Chem 269, 18192-18200. Rincon, J.C., Engler. S.K., Hargrove, B.W., Kunkel, GR. (1998). Molecular cloning of a cDNA encoding human SPH-binding factor, a conserved protein that binds to the enhancer-like region of the U6 small nuclear RNA gene promoter. Nucleic Acids Res 26, 4846-4852. 48 Robert, C., McGraw, S., Massicotte, L., Pravetoni, M., Gandolfi, F., Sirard, MA. (2002). Quantification of housekeeping transcript levels during the development of bovine preimplantation embryos. Biol. Reprod. 67, 1465-1472. Rodriguez, P., Pelletier, J ., Price, G.B., Zannis-Hadjopoulos, M. (2000). NAP-2, histone chaperone function and phosphorylation state through the cell cycle. J Mol Biol 298, 225- 238. Roebuck, K.A., Szeto, D.P., Green, K.P., Fan, Q.N., Stumph, W.E. (1990). Octamer and SPH motifs in the U1 enhancer cooperate to activate U1 RNA gene expression. Mol Cell Biol 10, 341-352. Romero-Oliva, F ., Allende, J,E. (2001). Protein pZIWAn/Cm is phosphorylated by protein kinase CK2 in vitro and interacts with the amino terminal end of the CK2 beta subunit. Journal of Cellular Biochemistry 81, 445-452. Ruzzene, M., Penzo, D., Pinna, L.A. (2002). Protein kinase CK2 inhibitor 4,5,6,7- tetrabromobenzotriazole (TBB) induces apoptosis and caspase-dependent degradation of haematopoietic lineage cell-specific protein 1 (H81) in Jurkat cells. Biochem J 364, 41- 47. Sacerdoti-Sierra, N., Jaffe, CL. (1997). Release of ecto-protein kinases by the protozoan parasite Leishmania major. J Biol Chem 272, 30760-5. Sadowski, C.L., Henry, R.W., Kobayashi, R., Hernandez, N. (1996). The SNAP45 subunit of the small nuclear RNA (snRNA) activating protein complex is required for RNA polymerase II and III snRNA gene transcription and interacts with the TATA box binding protein. Proc Natl Acad Sci U S A 93, 4289-4293. Sadowski, C.L., Henry, R.W., Lobo, S.M., Hernandez, N. (1993). Targeting TBP to a non-TATA box cis-regulatory element, a TBP-containing complex activates transcription from snRNA promoters through the PSE. Genes Dev 7, 1535-1548. Saez-Vasquez, J., Meissner, M., Pikaard, CS. (2001). RNA polymerase I holoenzyme- promoter complexes include an associated CK2-like protein kinase. Plant Mol Biol 47, 449-459. Samo, S., Reddy, H., Meggio, F., Ruzzene, M., Davies, S.P., Donella-Deana, A., Shugar, D., Pinna, L.A. (2001). Selectivity of 4,5,6,7-tetrabromobenzotriazole, an ATP site- directed inhibitor of protein kinase CK2 ('casein kinase-2'). FEBS Lett 496, 44-48. Samo, S., Ghisellini, P., Pinna, L. A. (2002). Unique activation mechanism of protein kinase CK2, the N-terminal segment is essential for constitutive activity of the catalytic subunit but not of the holoenzyme. J Biol Chem 277, 22509-22514. 49 Schaub, M., Myslinski, E., Schuster, C., Krol, A., Carbon, P. (1997). Staf, a promiscuous activator for enhanced transcription by RNA polymerases II and III. EMBO J 16, 173- 181. Schoonbroodt, S., Ferreira V., Best-Belpomme M., Boelaert J.R., Legrand-Poels S., Komer M., Piette J. (2000). Crucial role of the amino-terminal tyrosine residue 42 and the carboxyl-terminal PEST domain of IKB in NF -1(B activation by an oxidative stress. J. Immunol 164, 4292 4300. Schramm, L., Pendergrast, P.S., Sun, Y., Hernandez, N. (2000). Different human TFIIIB activities direct RNA polymerase III transcription from TATA-containing and TATA- less promoters. Genes Dev 14, 2650-2663. Schultz, M.C. (2003). DNA damage regulation of the RNA components of the translational apparatus, new biology and mechanisms. IUBMB Life 55, 243-7. Schuster, N., Gdtz, C., Faust, M., Schneider, E., Prowald, A. (2001). Wild-type p53 inhibits protein kinase CK2 activity. J Cell Biochem 81, 172-183 Schuster, C., Myslinski, E., Krol, A., Carbon, P. (1995). Staf, a novel zinc finger protein that activates the RNA polymerase III promoter of the selenocysteine tRNA gene. EMBO J 14, 3777-3787. Seither, P., Iben. S., Grummt, I. (1998). Mammalian RNA polymerase 1 exists as a holoenzyme with associated basal transcription factors. J Mol Biol 275, 43-53. Seldin, D., Leder, P. (1995). Casein kinase II? transgene-induced murine lymphoma, relation to theileriosis in cattle. Science 267,894-897. Shi, X., Potvin, 8., Huang, T., Hilgard, P., Spray, D.C., Suadicani, S.O., Wolkofi', A.W., Stanley, P., Stockert, RJ. (2001). A novel casein kinase 2 alpha-subunit regulates membrane protein traffic in the human hepatoma cell line HuH-7. J Biol Chem 276, 2075 2082. Skubitz, K.M., Ehresmann, D.D., Ducker, T.P. (1991). Characterization of human neutrophil ecto-protein kinase activity released by kinase substrates. J Immunol 147, 638- 650. Sommercom, J., Mulligan, J. A., Lozeman, F. J., Krebs, E. G. (1987). Activation of Casein Kinase II in Response to Insulin and to Epidermal Growth Factor. Proc Natl Acad Sci USA. 84, 8834-8838. Soncin, F., Zhang, X., Chu, B., Wang, X., Asea, A., Ann Stevenson .M, Sacks, D.B., Calderwood, SK. (2003). Transcriptional activity and DNA binding of heat shock factor- 1 involve phosphorylation on threonine 142 by CK2. Biochem Biophys Res Commun 303, 700-706. 50 Song, Q., Combest, W.L., Gilbert, LL (1994). Spermine and polylysine enhanced phosphorylation of calmodulin and tubulin in an insect endocrine gland. Mol Cell Endocrinol 99, 1-10. Sonka, J ., Kubler, D., Kinzel, V. (1989). phosphorylation by cell surface protein kinase of bovine and human fibrinogen and fibrin. Biochim Biophys Acta 997, 268-277. Stepanova, V., Jerke, U., Sagach, V., Lindschau, C., Dietz, R., Haller, H., Dumler, I. (2002). Urokinase-dependent human vascular smooth muscle cell adhesion requires selective vitronectin phosphorylation by ecto-protein kinase CK2. J Biol Chem 277, 10265-10272 Sugano, S., Andronis, C., Green, R.M., Wang, Z.Y., Tobin, EM. (1998). Protein kinase CK2 interacts with and phosphorylates the Arabidopsis circadian clock-associated 1 protein. Proc Natl Acad Sci U S A 95, 11020-11025. Sun, J. M., Chen, H. Y., Moniwa, M., Litchfield, D. W., Seto, E., Davie, J. R. (2002). The transcriptional repressor Sp3 is associated with CK2-phosphorylated histone deacetylase 2. J Biol Chem 277, 35783-35786. Szyszka, R., Boguszewska, A., Shugar, D., Grankowski, N. (1996). Halogenated benzimidazole inhibitors of phosphorylation, in vitro and in vivo, of the surface acidic proteins of the yeast ribosomal 6OS subunit by endogenous protein kinases CK-II and PK6OS. Acta Biochim P01 43, 389-396. Tawfic, S., Yu, S., Wang, H., Faust, R., Davis, A., Ahmed, K. (2001). Protein kinase CK2 in neoplasia. Histol Histopathol 16,573-582 Teichmann, M., Wang, Z., Roeder, R.G. (2000). A stable complex of a novel transcription factor IIB- related factor, human TFIIIBSO, and associated proteins mediate selective transcription by RNA polymerase III of genes with upstream promoter elements. Proc Natl Acad Sci U S A 97, 14200-1425. Toczyski, D.P., Galgoczy, D.J., Hartwell, L.H. (1997). CDCS and CKII control adaptation to the yeast DNA damage checkpoint. Cell 90, 1097 1106. Trembley, J.H., Tatsumi, S., Sakashita, E., Loyer, P., Slaughter, C.A., Suzuki, H., Endo, H., Kidd, V.J., Mayeda, A. (2005). Activation of pre-mRNA splicing by human RNPSl is regulated by CK2 phosphorylation. Mol Cell Biol 25, 1446-14457. Tsai, S. C., Seto, E. (2002). Regulation of histone deacetylase 2 by protein kinase CK2. J Biol Chem 277,31826-31833. 51 Tuazon, P.T., Traugh, J .A. (1991). Casein kinase 1 and II—multipotential serine protein kinases, structure, function, and regulation. Adv. Second Messenger Phosphoprotein Res 23, 123 164. Ubeda, M., Habener, J.F. (2003). CHOP transcription factor phosphorylation by casein kinase 2 inhibits transcriptional activation. J Biol Chem 278, 40514-40520. Unger, G.M., Davis, A.T., Slaton, J.W., Ahmed, K. (2004). Protein kinase CK2 as regulator of cell survival, implications for cancer therapy. Curr Cancer Drug Targets 4, 77-84. Vilk, G., Saulnier, R.B., St Pierre, R., Litchfield, D.W. (1999). Inducible expression of protein kinase CK2 in mammalian cells. Evidence for functional specialization of CK2 isoforms. J Biol Chem 274, 14406 14414 Voit, R., Kuhn, A., Sander, E.E., Grummt, I. (1995). Activation of mammalian ribosomal gene transcription requires phosphorylation of the nucleolar transcription factor UBF. Nucleic Acids Res 23, 2593-2599. Voit, R., Schnapp, A., Kuhn, A., Rosenbauer, H., Hirschmann, P., Stunnenberg, H.G., Grummt, I. (1992). The nucleolar transcription factor mUBF is phosphorylated by casein kinase II in the C-terminal hyperacidic tail which is essential for transactivation. EMBO J 11, 2211-2218. Waldschmidt, R., Wanandi, 1., Seifart, K.H. (1991). Identification of transcription factors required for the expression of mammalian U6 genes in vitro. EMBO J 10, 2595-2603. Wei, P., Garber, M.B., Fang, S.M., Fischer, W.H., Jones, K.A. (1998). A novel CDK9- associated C-type cyclin interacts directly with HIV-1 Tat and mediates its high-affinity, loop-specific binding to TAR RNA. Cell 92, 451-462. Wilhelm, V., Rojas, P., Gatica, M., Allende, C. C., Allende, J. E. (1995). Expression of the subunits of protein kinase CK2 during oogenesis in Xenopus laevis. Eur J Biochem 232, 671-676. Willis, I.M. (2002). A universal nomenclature for subunits of the RNA polymerase III transcription initiation factor TFIIIB. Genes Dev 16, 1337-1338. Winter, 3., Kautzner, 1., Issinger, O.G., Arnold, H.H. (1997). Two putative protein kinase CK2 phosphorylation sites are important for Myf-S activity. Biol Chem. 378, 1445-1456. Wirkner, U., Voss, H., Lichter, P., Ansorge, W., Pyerin, W. (1994). The human gene (CSNK2A1) coding for the casein kinase II subunit alpha is located on chromosome 20 and contains tandemly arranged Alu repeats. Genomics 19, 257 265. 52 Wisniewski, J. R., Szewczuk, Z., Petry, 1., Schwanbeck, R., Renner, U. (1999). Constitutive phosphorylation of the acidic tails of the high mobility group 1 proteins by casein kinase II alters their conformation, stability, and DNA binding specificity. J Biol Chem 274,20116 20122. Wong, M.W., Henry, R.W., Ma, 8., Kobayashi, R., Klages, N., Matthias, P., Strubin, M., Hernandez, N. (1998). The large subunit of basal transcription factor SNAPC is a Myb domain protein that interacts with Oct-1. Mol Cell Biol 18, 368-377. Xiao, C.Y., Jans, P., Jans, DA. (1998). Negative charge at the protein kinase CK2 site enhances recognition of the SV40 large T-antigen NLS by importin, effect of conformation. FEBS Lett 440, 297-301. Xu, X., Toselli, P.A., Russell, L.D., Seldin, DC. (1999). Globozoosperrnia in mice lacking the casein kinase II alpha' catalytic subunit. Nat Genet 23, 118-121. Yamaguchi, Y., Wada, T., Suzuki, F., Takagi, T., Hasegawa, J., Handa, H. (1998). Casein kinase II interacts with the bZIP domains of several transcription factors. Nucleic Acids Res 26, 3854-3861. Yamane, K., Kinsella, T.J. (2005a). Casein kinase 2 regulates both apoptosis and the cell cycle following DNA damage induced by 6-thioguanine. Clin Cancer Res 11, 2355-2363. Yamane, K., Kinsella, T.J (2005b). CK2 inhibits apoptosis and changes its cellular localization following ionizing radiation. Cancer Res 65, 4362-4367. Yang, Z., Zhu, Q., Luo, K., Zhou, Q. (2001). The 7SK small nuclear RNA inhibits the CDK9/cyclin T1 kinase to control transcription. Nature 414, 317-322. Yik, J .H., Chen, R., Nishimura, R., Jennings, J.L., Link, A.J., Zhou, Q. (2003). Inhibition of P-TEF b (CDK9/Cyclin T) kinase and RNA polymerase II transcription by the coordinated actions of HEXIMI and 7SK snRNA. Mol Cell 12, 971-982. Yik, J.H., Chen, R., Pezda, A.C., Zhou, Q. (2005). Compensatory contributions of HEXIM] and HEXIM2 in maintaining the balance of active and inactive positive transcription elongation factor b complexes for control of transcription. J Biol Chem 280, 16368-16376. Yim, H., Lee, Y. H., Lee, C. H., Lee, S. K. (1999). Emodin, an anthraquinone derivative isolated from the rhizomes of Rheum palmatum, selectively inhibits the activity of casein kinase II as a competitive inhibitor. Planta Med 65, 9 13. Yoon, J.B., Murphy, S., Bai, L., Wang, Z., Roeder, R.G. (1995). Proximal sequence element-binding transcription factor (PTF) is a multisubunit complex required for transcription of both RNA polymerase II- and RNA polymerase III-dependent small nuclear RNA genes. Mol Cell. Biol 15, 2019 2027. 53 Yoon, J.B., Roeder, R.G. (1996). Cloning of two proximal sequence element-binding transcription factor subunits (gamma and delta) that are required for transcription of small nuclear RNA genes by RNA polymerases II and III and interact with the TATA-binding protein. Mol Cell Biol 16, 1-9. Zamrod, Z., Stumph, W.E. (1990). U4B snRNA gene enhancer activity requires functional octamer and SPH motifs. Nucleic Acids Res 18, 7323-7330. Zandomeni, R., Zandomeni, M.C., Shugar, D., Weinmann, R. (1986). Casein kinase type II is involved in the inhibition by 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole of specific RNA polymerase II transcription. J Biol Chem 261, 3414-3419. Zhao, T., Heyduk, T., Eissenberg, J. C. (2001). Phosphorylation site mutations in heterochromatin protein 1 (HPl) reduce or eliminate silencing activity. J Biol Chem 276,9512-9518. 54 CHAPTER TWO COOPERATION BETWEEN SNAPC AND TBP ANTAGONIZES PROTEIN KINASE CK2 INHIBITION OF DNA BINDING BY SNAPC Abstract Protein kinase CK2 regulates RNA polymerase III transcription of human U6 snRNA genes, both negatively and positively, depending upon whether the general transcription machinery or RNA polymerase III is preferentially phosphorylated. Human U1 snRNA genes share similar promoter architectures as that of U6 genes, but are transcribed by RNA polymerase II. Herein, we report that CK2 inhibits U1 snRNA gene transcription by RNA polymerase II. Decreased levels of endogenous CK2 correlates with increased U1 expression while CK2 associates with U1 gene promoters, indicating that it plays a direct role in U1 gene regulation. CK2 phosphorylates the general transcription factor SNAPC that is required for both RNA polymerase II and III transcription, and SNAPC phosphorylation inhibits its binding to snRNA gene promoters. However, restricted promoter access by phosphorylated SNAPc can be overcome by cooperative interactions with TBP at a U6 promoter but not at a U1 promoter. Thus, CK2 may have the capacity to differentially regulate U1 and U6 transcription even though SNAPC is universally utilized for human snRNA gene transcription. This work was published as the following manuscript: Gu L, Esselman WI, Henry RW. (2005) Cooperation between SNAPC and TBP antagonizes protein kinase CK2 inhibition of DNA binding by SNAPC. J Biol Chem. 2005 Jun 14; [Epub ahead of print] 55 Introduction Protein kinase CK2 is an important regulator of cellular growth (Guerra and Issinger, 1999; Meggio and Pinna, 2003; Pinna, 2002; Pinna and Meggio, 1997) and abnormal CK2 activity may contribute to tumor progression (Tawfic et al., 2001). CK2 is a tetrameric enzyme composed of two catalytic subunits or and or’ and two copies of the regulatory [3 subunit (Niefind et al., 2001). One role for CK2 is to function as a regulatory protein that controls gene transcription. For example, general RNA synthesis in yeast is impaired when a temperature sensitive mutant of the CK2a’ subunit is shifted to a restrictive temperature (Hanna etal., 1995). This decline in total RNA synthesis also suggests that expression of highly transcribed genes encoding ribosomal (r), transfer (t), and small nuclear (sn) RNAs is sensitive to levels of functional CK2. In yeast, CK2 is important for active RNA polymerase III transcription (Hockman and Schultz, 1996), and yet paradoxically, CK2 has been proposed to be the terminal effector in a DNA damage response pathway that represses RNA polymerase III transcription (Ghavidel and Schultz, 2001). In humans, CK2 exhibits differential effects on gene transcription during the cell cycle. During mitosis, CK2 inhibits RNA polymerase III transcription, whereas at other stages CK2 can stimulate transcription (Hu et al., 2004). The nature of the regulation is dictated by CK2 target selection. One key target for CK2 is the general transcription factor TFIIIB (Ghavidel and Schultz, 1997; Hu et al., 2003; Johnston et al., 2002). There are at least two versions of human TFIIIB that function for transcription of distinct classes of genes (Schramm and Hernandez, 2002). The Brfl -TFIIIB complex functions for SS rRNA and tRNA transcription and is composed of the TATA-box binding protein (TBP) and the TBP associated factors, deI 56 and Brfl. The Brfl-TFIIIB complex functions for U6 snRNA transcription and is composed of TBP plus de1 but Brf2 instead of Brfl. Brfl -TFIIIB phosphorylation during M-phase results in the selective release of deI from tRNA promoters (F airley et al., 2003). Hernandez and colleagues further demonstrated that del is the critical CK2 target within Brfl-TFIIIB for mitotic repression of U6 transcription (Hu et al., 2004). As de1 is a shared component of both TFIIIB complexes, CK2 may target this factor to repress global RNA polymerase III transcription. However, CK2 inhibitors also interfere with Brfl -TFIIIB binding to the TFIIIC complex (Johnston et al., 2002), which itself recognizes intragenic promoter elements of SS rRNA and tRNA genes, suggesting that CK2 also has a stimulatory role in RNA polymerase III transcription through enhanced preinitiation complex assembly. Consistent with this positive role, CK2 can also activate RNA polymerase III transcription in human cells (Johnston et al., 2002), and in this process may additionally phosphorylate RNA polymerase III itself (Hu et al., 2003). Together, these data point to an important but complex role for CK2 control of RNA polymerase III transcription. Human U6 snRNA genes are interesting because they are transcribed by RNA polymerase III and yet their promoters are similar to other snRNA genes, such as U1 and U2, which are transcribed by RNA polymerase 11 (Henry et al., 1998a; Hernandez, 2001; Lobo and Hernandez, 1994). Consequently, the mechanisms regulating human snRNA gene transcription by RNA polymerases II and 111 may also be shared. Nonetheless, the RNA polymerase II-transcribed genes do not use TFIIIB and thus rely on other factors for regulatory intervention. Regardless of po1ymerase specificity, all human snRNA genes contain a distal sequence element (DSE) encompassing an octamer element that is 57 recognized by Oct-1. Additional sites for the Spl (Ares et al., 1987) and STAF (Schaub et al., 1997) transcriptional activator proteins are adjacently located to the DSE at some snRNA genes (Hernandez, 1992). Oct-1 activates snRNA transcription by direct protein contacts (Ford et al., 1998; Hovde et al., 2002; Mittal et al., 1999) with the basal transcription factor called the snRNA activating protein complex (SNAPC) (Sadowski et al., 1993), which is also referred to as the proximal sequence element transcription factor (PTF) (Murphy et al., 1992). SNAPC binds to the proximal sequence element (PSE) common to the core promoters of human snRNA genes and functions for both RNA polymerase 11 and III transcription (Henry et al., 1998a; Henry et al., 1996; Henry et al., 1998b; Henry et al., 1995; Sadowski et al., 1996; Sadowski et al., 1993; Wong et al., 1998). SNAPC contains at least five proteins called SNAP190 (PTFOL), SNAPSO (PTFB), SNAP45 (PTFB), SNAP43 (PTFy), and SNAP19 (Bai et al., 1996; Henry et al., 1996; Henry et al., 1998b; Henry et al., 1995; Sadowski et al., 1996; Wong et al., 1998; Yoon and Roeder, 1996). The largest subunit SNAP190 plays a centrally important role in human snRNA gene transcription first by serving as the scaffold for SNAPc assembly though interactions with most other members of SNAPC (Ma and Hernandez, 2001; Ma and Hernandez, 2002). Once the complex is assembled, SNAP190 further recognizes the PSE through its Myb DNA binding domain (Wong et al., 1998) and is also the direct target for Oct-1 (Ford et al., 1998; Mittal et al., 1996). In an unexpected twist, SNAP190 can make DNA contacts within the U1 DSE and stimulate the binding of Oct-1 to this enhancer, suggesting that in some contexts coordinated binding of the activator and general transcription machinery is important for transcriptional activation (Hovde et al., 2002) 58 Human U6 snRNA genes, but not U1 genes, also contain a TATA box that is located adjacently to the PSE, and this promoter arrangement dictates that transcription occurs by RNA polymerase III (Lobo and Hernandez, 1989). The TATA box is recognized by the TBP component of the Brf2-TFIIIB complex (Cabart and Murphy, 2001; Cabart and Murphy, 2002; Hinkley et al., 2003; Ma and Hernandez, 2002; Zhao et al., 2003). SNAPc, through its SNAP190 subunit, stimulates TBP binding to the U6 TATA box as an early critical step in RNA polymerase III transcription (Hinkley et al., 2003; Ma and Hernandez, 2002). TBP is also required for human snRNA gene transcription by RNA polymerase II (Sadowski et al., 1993), but how TBP is recruited to these TATA-less promoters is unclear. Nonetheless, it is likely that SNAPC contributes to TBP activity at these genes. SNAPC and TBP co-purify extensively during the biochemical fractionation of SNAPC (Henry et al., 1995), and those fractions enriched for SNAPC and TBP can reconstitute U1 snRNA transcription in vitro from extracts that have been depleted of endogenous TBP (Sadowski et al., 1993). Thus, SNAPC plays a pivotal role in snRNA gene transcription by providing core promoter recognition, serving as a target for transcription activation by Oct-1, and coordinating TBP activity and pre-initiation complex assembly for both RNA polymerases II and 111. Additional RNA polymerase 11 general transcription factors are also required for U1 transcription including TFIIA, TFIIB, TFIIE, and TFIIF (Kuhlman et al., 1999). As in RNA polymerase III transcription, CK2 also has a complex role in regulating RNA polymerase II transcription. CK2 phosphorylation of TFIIA and TFIIE stimulates preinitiation complex assembly at the adenovirus major late promoter while TFIIF phosphorylation can stimulate RNA polymerase II elongation. In 59 contrast, CK2 phosphorylation of RNA polymerase II inhibits transcription, potentially by impairing elongation (Cabrejos et al., 2004). The striking parallel between RNA polymerase II and III transcription of human snRNA genes prompted an investigation into the role of phosphorylation in U1 transcription. In this study, we report that CK2 inhibits overall U1 snRNA gene transcription by RNA polymerase II and can phosphorylate SNAPC to inhibit its DNA binding. Interestingly, cooperative interactions of SNAPC with TBP at U6 but not at U1 promoter DNA can overcome the repressive effects of CK2. Together, these data suggest that CK2 may differentially affect preinitiation complex assembly for RNA polymerase II and III transcription of human snRNA genes depending upon the promoter architectures. Materials and Methods Chromatin immunoprecipitation assays ChIP assays from HeLa cells were performed using the anti-CKZa (Ab245), anti- CK213(Ab278) antibodies (Yu etal., 1991) as well as anti-SNAP43 (CS48) (Henry et al., 1995) and anti-TBP antibodies described previously (Hirsch et al., 2004). Enrichment of genomic sequences in the immunoprecipitation reactions was measured by PCR as previously described (Hirsch et al., 2004). RNAi CK20t and CK2a' cDNA were generated with a T7 promoter at both ends by RT- PCR using total RNA from HeLa cells as a template. The primers for CK2a are CK2a 60 forward 5 -GCGTAATACGACTCACTATAGGAAATAATGAAAAAGTTGTTG-3 , and CK20t reverse 5 -GCGTAATACGACTCACTATAGGCTCTTGCAGTAAGCCGT GAC-3 . The primers for CK2a' are CKZOt' forward 5 -GCGTAATAC GACTCACTAT AGGCAACAATGAGAGAGTGGTTG-3 , and CKZa' reverse 5 -GCGTAATACGACT CACTATAGGTCTGTTGATGGTCGTATCGC-3 . LacZ cDNA with a T7 promoter at both ends was generated by PCR using pPelican-lacZ as a template. The primers used are lacZ forward 5 - TTAATACGACTCACTATAGGGAGACGATAACCACCACGCT CATCG-3 , and lacZ reverse 5 - TTAATACGACTCACTATAGGGAGAGCGTTACC CAACTTAATCGCC-3 . Resultant cDNAs were subjected to in vitro transcription with T7 polymerase to produce double stranded (ds) RNA. After DNase I treatment, dsRNA was incubated with recombinant Dicer and resultant Dicer generated small interfering RNA (d-siRNA) were purified according to the manufacturer’s instructions (lnvitrogen). Approximately 250 ng of d-siRNA for lacZ, CK2a, or CK20L plus 250 ng CKZa’ d- siRNA were transfected into HeLa cells using Lipofectamine 2000 (lnvitrogen). Cells were harvested 30 hr later and total RNA was extracted using TriZol (Gibco). Reverse transcription (RT)-PCR was carried out using Titan One Tube RT-PCR System (Roche). The primers used to amplify U1 primary transcript are U1 prim forward 5 - ACTTGCTGCTTCACCACGAA-3 , and U1 prim reverse 5 - ACAGCCTCATACGCCTCACT-3 . The primers used to amplify the total U1 snRNA population are U1 forward 5 -ATACTTACCTGGCAGGGGAG-3 , and U1 reverse 5 - CAGGGGAAAGCGCGAACGCA-3 . RT-PCR products were separated by 3% Tris borate EDTA agarose electrophoresis, stained with ethidium bromide, and visualized with Kodak imaging software. 61 In vitro transcription assays In vitro transcription of human U1 and U6 snRNA genes were performed as described previously (Lobo and Hernandez, 1989), with the following modifications. The HeLa cell nuclear extracts were pre-incubated with Dignam buffer D either with or without recombinant CK2 and kinase inhibitors for 60 min at 30°C prior to initiating transcription by addition of transcription buffers, nucleoside triphosphates, and DNA templates. The amounts of recombinant CK2, and kinase inhibitors used are indicated in the figure legend. Transcripts were separated by denaturing PAGE and visualized by Phosphorlmager analysis (Molecular Dynamics). Expression and purification of recombinant proteins GST-SNAP19O (1-719) was expressed in Escherichia coli BL21 (DE3) using the vector pSBet-GST-SNAP190 (1-719) and was purified for in vitro kinase assays by affinity chromatography using glutathione-sepharose beads (Amersham Biosciences). Recombinant mini-SNAPC, containing SNAP190 (1-719), SNAP43, and SNAPSO, was co-expressed in E. coli using the vector combination pSBet-GST-SNAP190 (1-719) and pE’I21-His-SNAP43/HA-SNAP50. Recombinant mini-SNAPC was affinity purified using glutathione agarose beads followed by digestion with thrombin to release the complex from the GST-tag, and dialysis against Dignam buffer D containing 80 mM KCl. 62 Immunoprecipitation and in vitro kinase assays For the experiment presented in Figure 11-3 A, 60 and 180 ILL of HeLa cell nuclear extract (~10 mg/mL) was incubated with 20 uL of rabbit anti-SNAP43 (C848) (Henry et al., 1995), anti-SNAP190 (C8398, C8402) (Henry et al., 1995), anti-CKZOL (AB245) (Yu er al., 1991) or pre-immune antibodies covalently coupled to protein-G agarose beads. Recovered proteins were analyzed by Western blot using a mouse mAb against CK2a (Transduction Laboratories). For Figure II-3 B, 40 1.1L of HeLa cell nuclear extract was used for each immunoprecipitation. After extensive washing with HEMGT-ISO buffer (20 mM Hepes, pH 7.9, 0.1 mM EDTA, 5 mM MgC12, 10% glycerol, 0.5% Tween-20, 150 mM KCI), the beads were suspended in 40 11L of HEMGT-ISO buffer containing 2 uL of y[32P]-ATP (6000 Ci/mmol, 150 mCi/mL), and the samples were incubated at 30°C for 15 min. The beads were then washed extensively in HEMGT-ISO buffer and proteins were separated by 12.5% SDS-PAGE. Radiolabeled proteins were visualized by autoradiography. For Figure 11-3 C, 100 uL of HeLa cell nuclear extracts were used for immunoprecipitation. After kinase reactions, proteins were separated by SDS-PAGE and transferred to nitrocellulose membrane. Radiolabeled proteins were detected first by autoradiography. Subsequently, Western blot analyses were performed using anti-SNAP190 (C8402) antibodies. For Figure II-SB and 5C, approximately 5 pg of GST-SNAP190 (1-719) was bound to glutathione agarose beads (10 11L). Immobilized GST-SNAP190 (1-719) was incubated with 10 uL HeLa cell nuclear extract for 30 min at 30°C. The beads were washed extensively with HEMGT-ISO. In vitro kinase assays were then performed 63 directly on the beads. Kinase reactions were also performed using untreated GST- SNAP190 (1-719) plus 10 units of recombinant CK2 (New England Biolabs). Where indicated, kinase reactions were performed in the presence of 20 nM y[32P]-ATP or y[32P]-GTP with or without D-ribofuranosyl benzimidazole (DRB; Sigma) or 3,3 ,4 ,5,7- pentahyroxyflavone (quercetin; Sigma). T typtic phosphopeptide mapping To obtain material for thin layer chromatography (TLC) analysis, approximately 1 pg GST-SNAP190 (1-719) was labeled with y[32P]-ATP by using HeLa cell nuclear extracts or recombinant CK2. Phosphorylated G8T-SNAP190 (1-719) was gel purified prior to digestion with sequencing grade modified trypsin (Promega). The tryptic fragments from each of these reactions were spotted individually or were combined at a 1:1 ratio and spotted onto a cellulose TLC plate. Peptides were separated in the first dimension by electrophoresis in pH 1.9 buffer (formic acid (88% w/v)/glacia1 acetic acid/dH20, 25:78:897, v/v/v) and in the second dimension by chromatography in chromatography buffer (n-butanol/pyridine/ glacial acetic acid/dHZO, 15:10:13:12, v/v/v/v) prior to detection by Phosphorlmager analysis (van der Geer and Hunter, 1994). Phosphoamino Acid Analysis Endogenous SNAP190 was immunoprecipitated, phosphorylated in vitro in the presence of 2 uL of y[32P]-ATP (6000 Ci/mmol, 150 mCi/mL), separated by 7.5% SDS- PAGE, and transferred to nitrocellulose membrane. The ~l90 kDa radioactive protein corresponding to SNAP190 was excised and hydrolyzed in 5.7N HCl for 1 hr at 100°C. 64 Recovered amino acids were vacuum dried and dissolved in 10 ILL pH 1.9 buffer containing unlabeled phosphoserine, phosphothreonine and phosphotyrosine mixture. The mixture was separated by one-dimensional electrophoresis (500V) on cellulose thin layer chromatography (TLC) plates (Eastman Kodak Co.) for 1 hr at 0 °C in pH 2.5 buffer [67% pH 3.5 buffer (glacial acetic acid/pyridine/water, 50:5:945, v/v/v, containing 0.5 mM EDTA) and 33% pH 1.9 buffer (glacial acetic acid/88% formic acid/water, 78:25:897, v/v/v)]. Unlabeled amino acid standards were visualized by spraying the cellulose plates with ninhydrin. The 32P-labeled amino acid residues were visualized by autoradiography with a phosphorimager. Electrophoretic mobility shift assays Approximately 100 ng of purified mini-SNAPC and/or 30 ng TBP were preincubated alone or with 15 or 150 units CK2 and/or 7 mM ATP for 30 min at 30°C. EMSAs were then performed using DNA probes containing a wild-type mouse U6 PSE with a wild-type or mutant human U6 TATA box as described previously (Hinkley et al., 2003; Mittal and Hernandez, 1997). Results CK2 inhibits U1 snRNA gene transcription. The conservation of similar promoter architectures among the human snRNA gene family suggests that these genes could be coordinately regulated. That CK2 regulates human U6 snRNA gene transcription by RNA polymerase III (Hu et al., 2003) 65 prompted us to examine whether CK2 similarly regulates human snRNA gene transcription by RNA polymerase II. First, chromatin immunoprecipitation (ChIP) experiments were performed to determine whether endogenous CK2 could associate with the promoter regions of both U6 and U1 snRNA genes. As shown in Figure II-l A, both U6 and U1 promoter regions were enriched in immunoprecipitation reactions using anti- CKZa antibodies (lane 7), while U1 but not U6 promoter regions were enriched in the anti-CKZB immunoprecipitation reactions (lane 8). Similar results were obtained in experiments performed with different antibodies directed against CK2a and CK2|3 (data not shown). Possibly the epitopes recognized by the CKZB antibodies may be occluded by other transcription factors at the U6 promoter. However, this result stands in contrast with that noted previously (Hu et al., 2003), wherein a more robust CKZB association with this U6 promoter was noted and only weak Ul promoter association by any CK2 subunit was detected. The reason for this discrepancy is unclear, but differences in chromatin immunoprecipitation or cell growth conditions could potentially affect promoter recovery by CK2 antibodies. The levels of U1 and U6 promoter recovery in this reaction were less than that those observed in either anti-SNAP43 (lane 6) or anti- TBP (lane 9) reactions, but markedly greater than that seen in reactions using IgG (lane 5). No significant enrichment of the GAPDH exon 2 or U1 upstream regions was observed in any reactions. Therefore, endogenous CK2 associates with the promoter regions of both U1 and U6 genes, and suggests the possibility that CK2 could additionally affect human snRNA gene expression by RNA polymerase II. To test whether endogenous CK2 influences human U1 gene expression in living cells, CK2 levels were reduced by RNA interference (RNAi), and the effect on U1 66 Figure II-l. CK2 inhibits U1 snRNA in vivo gene expression. (A) Endogenous CK2 associates with snRNA gene promoters. Chromatin immunoprecipitation experiments were performed using HeLa cell chromatin and the indicated antibodies. Enrichment of the U6 and U1 promoter regions was detected by PCR and was compared to recovery of the U1 upstream regions (U1 up) and GAPDH exon 2 (GAPDH), as negative controls. (B) U1 primary transcripts accumulate after CK2 reduction. CK2 levels were reduced by transient transfection of dicer generated small interfering RNA (Myers et al., 2003) corresponding to CK20t (lane 7), or CK2a plus CKZOL’ (lane 8). Cells were also treated with LacZ d-siRNA as a reference (lane 6). Levels of the U1 primary transcript and total U1 population were monitored by RT-PCR (top). Endogenous CK2 and actin levels were measured by Western analysis (bottom). For reference, lanes 1-5 contain two-fold decreasing increments of material harvested from untreated cells to serve as a standard curve for each assay. 67 68 A Q a a .. . Input 2 Q Q m (g, a) o o 1- ~ - a :5 a :5 - c- - a —- .- U6 - - “ - — -- - U1 - «- -- U1 up - .- - GAPDH 1 2 3 4 5 6 7 8 9 B d-siRNA w 1 Input 5 fir-m..- mfg U1primary RT-PCR u- - '9 an- U1 total '- r“ "" “r“ CK2a Western ~-— '“" "" - Actin 1 2 3 4 5 6 7 8 snRNA production was monitored by RT-PCR (Figure II-l B). As a negative control, RNAi was also performed using lacZ-specific RNA. As it was demonstrated that phosphorylation of the carboxy terminal domain (CTD) of RNA polymerase II contributes to 3 processing of human U2 snRNA (Jacobs et al., 2004; Medlin et al., 2003), it was possible that CK2 could also play a role in U1 3 processing. Therefore, in this experiment, different primer combinations were used to detect either the primary U1 snRNA transcript that is normally processed rapidly or the total U1 snRNA steady state population. Lanes 1-5 (top panel) shows that amplification of the U1 primary transcript and total U1 snRNA levels were directly correlated with the amount of RNA included in the reaction. Interestingly, U1 primary transcript levels were increased in cells treated with CK2a RNAi (lane 7) and this effect was enhanced by RNAi directed against both CK2a and CKZOL’ (lane 8) as compared to cells treated with LacZ-specific RNAi (lane 6). Steady state U1 snRNA levels remained effectively unchanged by any of these treatments, consistent with their abundant and extremely stable nature. In this experiment, endogenous CK2 levels were reduced approximately 2-4 fold relative to the levels observed in the LacZ-RNAi treated cells, whereas actin levels were unaltered by any of the treatments (bottom panel). The increase in U1 primary transcripts with reduced CK2 levels suggests that endogenous CK2 normally stimulates U1 3 processing and/or inhibits U1 transcription. To determine whether CK2 plays a direct role in U1 snRNA gene transcription, recombinant CK2 and commonly used inhibitors of CK2 were tested for their effect using in vitro U1 transcription assays. One characteristic of CK2 is that it can be inhibited by both DRB and quercetin, and therefore, these inhibitors were selected. As a positive 69 control for CK2 activity, in vitro U6 transcription was also examined (Figure 11-2 A). First, U6 transcription was inhibited with increasing amounts of the kinase inhibitor DRB (lanes 2 and 3) and DRB action was reversed by addition of CK2 (lane 4). Together, these data suggest that CK2 has an overall positive role in U6 transcription, consistent with observations previously described (Hu et al., 2003). No measurable effect on U1 transcription was observed by addition of recombinant CK2 (compare lanes 5 and 6), perhaps because the HeLa cell extracts used for these experiments contain high levels of CK2 and CK2 activity is not rate limiting for transcription. U1 transcription was inhibited by DRB (lane 7),but inhibition was not reversed by addition of CK2 (lane 8), suggesting, in addition to CK2, DRB inhibits a kinase activity that is important for U1 snRNA gene activity. Indeed, the DRB sensitive elongation factor p-TEFb, which phosphorylates the CTD of RNA polymerase 11 (Price, 2000), also contributes positively to efficient U1 transcription in vitro (data not shown). Next, the effect of quercetin in U1 transcription was tested. As shown in Figure 11-2 B, and in contrast with DRB, quercetin addition stimulated U1 transcription (lanes 2 and 3). The increased background in reactions containing quercetin indicates this inhibitor may have non-specific positive effects on transcription possibly from cryptic promoters on the reporter plasmid. While quercetin can inhibit a variety of kinases, the increase in Ul-specific transcription was reversed by addition of increasing amounts of recombinant CK2 (lanes 4 and 5), suggesting that CK2 has a direct and overall negative role in controlling U1 transcription. Previously, U2 snRNA gene transcription by RNA polymerase II in nuclear run-on assays was not sensitive to DRB (Jacobs et al., 2004; Medlin et al., 2003), suggesting that CK2 is not involved in the transcription of these 70 Figure II-2. CK2 represses in vitro U1 transcription. (A) CK2 reverses inhibition of U6, but not U1, transcription by the kinase inhibitor DRB. In vitro U6 transcription assays were performed in the absence (lane 1) or presence of DRB (lane 2, 1 uM DRB; lanes 3 and 4, 7 uM DRB). The reaction shown in lane 4 also contains 10 units of recombinant CK2. In vitro U1 transcription assays were performed using a U1 G-less cassette in the absence (lanes 5 and 7) or presence of 10 units of recombinant CK2 (lanes 6 and 8), either in the absence (lanes 5 and 6) or presence of 7 uM DRB (lanes 7 and 8). (B) In vitro U1 transcription is sensitive to the CK2 inhibitor quercetin. Additional U1 transcription assays were performed in the absence (lane 1) or presence of 1 uM (lanes 2) and 7 uM quercetin (lanes 3-5), respectively, while an additional 10 and 100 units of CK2 were added to reactions shown in lanes 4 and 5, respectively. 71 + DRB ‘lDRBI I ——— +cK2 -_:':—.+ CK2 a...»- U6 “on“? or U1 -_—--—+ 10 IC 1234 5678 Quercetin Pa}. -'. . stile-.1 1'. "-IN' '2: ”’1’ A." , , .1 A. ' I '. 1'.- L1 72 U1 genes. The possibility remains that U1 and U2 gene transcription are differentially sensitive to regulation by CK2. Nonetheless, as U1 and U2 genes utilize similar promoter elements and general transcription factors for efficient transcription, a role for CK2 in U2 transcription cannot yet be dismissed. Endogenous CK2 targets SNAP190 for phosphorylation at multiple sites. SNAPc recognizes the core promoters of human snRNA genes and plays an important early role in coordinating transcription of snRNA genes by both RNA polymerases II and III. The findings that CK2 can affect both human U1 and U6 transcription (Hu et al., 2003) implicates SNAPC as a potential target for CK2. First, we examined whether CK2 co-purifies with SNAPc. As shown in Figure II-3 A, endogenous CK2 from HeLa cell nuclear extract was recovered with SNAPC during immunoprecipitation using anti-SNAP190 (lanes 4 and 5) or anti-SNAP43 (lane 3) antibodies but not while using IgG antibodies (lane 2). These levels of recovered CK2 are significantly less than that observed in reactions using antibodies against CK2a (lane 7), suggesting that only a minor proportion of CK2 is associated with SNAPc or that the interaction between SNAPC and CK2 is not stable. In separate experiments, recombinant CK2 alone did not cross-react with the anti-SNAP43 antibodies (data not shown), suggesting that recovery of CK2 in these assays requires SNAPC. These results indicate that endogenous CK2 associates with SNAPC. Next, to determine whether any subunits of SNAPC can be phosphorylated by SNAPc-associated kinase(s), including CK2, the anti-SNAP43 immuno-precipitated 73 Figure 11-3. Endogenous CK2 associates with SNAPC. (A) HeLa cell nuclear extracts were immunoprecipitated with pre-immune (lane 3), anti-SNAP190 (lane 4, CS398; lane 5, C8402), anti-SNAP43 (lane 6, C848), or anti-CK2a (lane 7, Ab245) antibodies, as indicated. Recovered proteins were analyzed by Western blot analysis using antibodies directed against the or subunit of CK2. Lanes 1 and 2 contain 3 and 1 uL of nuclear extract, respectively. (B) Multiple subunits of SNAPC are phosphorylated in vitro by a SNAPc-associated kinase activity. Immunoprecipitation reactions were performed from HeLa cell nuclear extracts using anti-SNAP43 (lane 1) or pre-immune (lane 2) antibodies that were immobilized on protein G agarose beads. After extensive washing, an in vitro kinase assay was performed on the beads in the presence of 2 uL of y[32P]-ATP (6000 Ci/mmol, 150 mCi/mL). Proteins were then separated by 15% SDS-PAGE and radiolabeled proteins were detected by autoradiography. The positions of SNAP190 and SNAP43 are labeled whereas a protein that migrates similarly to SNAP19 is indicated by an asterisk. (C) SNAP190 co-migrates with a 190-kDa phosphoprotein. HeLa cell nuclear extracts were immunoprecipitated with pre-immune (lane 1), anti-SNAP43 (lane 2), or anti-SNAP190 antibodies (lanes 3 and 4). Recovered samples were subjected to an in vitro kinase assay in the presence (lanes 1-3) or absence (lane 4) of y[32P]-ATP. Proteins were separated by 7.5% SDS-PAGE and transferred to nitrocellulose membrane. Radiolabeled proteins were detected by autoradiography for 1 hour (lanes 1-4). This membrane was then used for Western blot analysis using antibodies directly against SNAP190. Lanes 5 and 6 are the same as lanes 3 and 4 but exposed on film for 5 seconds. The position of SNAP190 is indicated. *The experiment in B was performed by RW Henry. 74 . ea... mvm15)”, >46)”. 3'0)”. W9)”, Y(10)H, 3'03)”, K16)”, W17)”. and W18)” ions were highlighted in the enlarged spectrum in the mass range of m/z 200-700 (bottom panel). "8 - 18" designates a dehydroalanine, which results from the B-elimination of phosphoserine (labeled as p8 in the peptide sequence). (B) Each of the 20 serines in the serine rich region could be phosphorylated. A total of 62 MS/MS spectra data from numerous phosphopeptides encompassing the serine rich region aa 514-545 were analyzed. The occurrence of phosphorylation at each amino acid residue is shown in y- axis; the corresponding amino acid sequence is indicated in x-axis. (C) Sequence of the SNAP190 peptide containing amino acids 514-545. CK2 consensus sites within these peptides are bracketed underneath. Serine residues that match CK2 consensus motif within this peptide are highlighted with asterisks. Other sites that are bracketed above indicate the possible CK2 sites generated by the nascent phosphoserines. 114 y2 “4444-444-wipers-1444444 D19 W3 1110”? 117116115114 M3 M4 b16 y19y18y17y16 b7 y24 y23 y21 m m a an ............... .- flu 1 ....... is... ............. l u. . IIIIII I H I r IIIIII it: ....... - m am.- I I .I I I > .................. I 1 8 .16: 1.8 50> ...... W A” P} IIIIIIIIIIIIIIII III I “M I AM 1 IIIIIIM I W m .3... E:..:...............-................. .... 1 s AVSS...‘ K IIIIIIIIIII J1I llllll ++AM a) IIIIIIII +4.8... ......... vm ............ 1 En ........ m 3 fl; m +1 rm: ++¢N~O>w ..... m M +1639. ........................ “ 13:9. 68...... m 6.? ---.+.+...m............. .... II II I I 18.: ..................... .xk.x-- -w . -IIIIIII. m .zo:os:-:-:-->s 1.3.2 ........... met-unnuuuu m s I IIIIIII Ia... .............. m is... ......... p m... ..... m is .......... 4.9.8 ............... M. aim-1.4 ....... ,..-...u.... ............. .2; tag... A30» ................... + 4.6-.) ---------- m M I T IIIIIII 1.9; .................. W IE. 1.2. ................ .m- IIIIII ta; ................ - IIIIIIIIIIIIII S 1...... as .......... m a: llllll + IIIIIIIIIIIIIIIIIIIIIIIII as. ........... m 3.22:. 9.5.9". 2.2!: 2.1.... 115 GammmaHBZmuquWawmmman I IE HSVRWSSTSS8088808800888888888850 SNAP190 aa 514—545 E. 514 *** 545 HSVRWSSTSSSGSSSGSSGGSSSSSSSSSEED * CK2 consensus: SXXD/E 116 serine rich region and the numbers are summarized in Figure III-3 B, showing that each of the 20 serines in this serine rich region was phosphorylated. Although other kinases derived from HeLa nuclear extracts could contribute to phosphorylation in this region, CK2 is likely the major kinase responsible for phosphorylation in this region (Figure II-5). SNAP190 (514-545) has three CK2 consensus substrate recognition sequences at the C- terrninal side of multiple blocks of contiguous serine residues, that is, Glum, Glus“, and Asp545 (illustrated in Figure III-3 C). As a result, Serm, Sers‘", and Ser542 are the first sites that can be phosphorylated. Phosphorylation introduces a negative charge to that residue, which potentially creates a new CK2 site at a nearby serine residue located at the N - 3 position. Therefore, the initial phosphorylation at Serm‘542 leads to propagation of upstream serine phosphorylation. This observation is ftuther supported by recent study that CK2 can phosphorylate seven contiguous serine residues (Serm’84 ) in the serine tract 78sssssssmn87 in dehydrin (DHNl) protein (Jiang and Wang, 2004). A schematic representation of SNAP190 and the position of the CK2 sites relative to other functional regions are shown in Figure III-4 A (top panel). Interestingly, the CK2 phosphorylated region aa (514-545) is adjacent to the Myb DNA binding domain, and both CK2 phosphorylated regions are adjacent to the TBP-recruiting regions (TRRl and TRR2) in SNAP190 that are involved in cooperative promoter recognition by SNAPC and TBP, suggesting a potential role for CK2 in controlling DNA binding and/or TBP recruitment. Our previous data demonstrated that PSE-specific DNA binding by m8 was inhibited by CK2, and that cooperation between SNAPC and TBP can antagonize CK2 inhibition of DNA binding by SNAPC (Figure II-6). The mechanism of the inhibitory effect of CK2 phosphorylation on SNAPC DNA binding is under investigation. 117 Figure III-4. Deletion of CK2 phosphorylation regions within SNAP190 did not affect the transcription properties of SNAPC. (A) A schematic representation of full- length SNAP190 and the position of the CK2 sites relative to other functional regions are shown (top panel). Myb domain: Myb DNA binding domain; SRR: the serine rich region; RRR: the arginine rich region; TRRl: TBP-interacting region 1; TRR2: TBP-interacting region 2; OIR: Oct-1 interacting region, 43/19: SNAP43 and SNAP19 interacting region; and 45: SNAP45 interacting region (modified from Ma and Hernandez, 2001). The wavy vertical line around position 719 indicates the C-terminus of the truncated SNAP190 protein used for these studies. Sequence of the SNAP190 amino acids 1-719 is also given and thirteen CK2 sites are highlighted (bottom panel). (B) SNAP190 phosphorylation was diminished in the deletion mutants. Schematic representation of various GST fusion proteins containing truncated SNAP190 is shown on the left side. The GST fusion proteins were overexpressed in E. coli and purified for in vitro kinase assays by affinity chromatography using glutathione-sepharose beads. Phosphorylation signals were detected by autoradiography (lane 1), and the amount of GST-SNAP190 proteins used in each reaction was detected by staining with Coomassie Blue (lane 2). (C) SNAP190 deletion mutants are competent for both U1 and U6 transcription. Partial SNAP complexes mS, mSAN, mSAC, and m8A(N+C), were assembled by co-expressed in E. coli and purified through affinity chromatography. The titrations of the mini-SNAP complexes were over a threefold range. The correctly initiated transcripts are labeled U6 and U1 5'. 118 .Hzgmmmg maggmmm Baum—ONAOH ugomghz ggmgubs. Saddam Hmm Ray—Mdnvdmm Edmundnudm mm>mom4m4m >O>mmBMMOm fidhfimdmxm UGOmdmmOMm How «g5» Eda—Ha u again mgmfimo mu ammo"; may—Human m H2. gnaw; $30335 3:55.88 Huang? Sm 5853mm 9:25.22 moomuoaoom «nausea—M2 Roam—Bo: 4533mm”. Sm Humane—Em Bumzosba Humenmmomm szmHumxnm 8523.5. mmqozHoon in umemmooxma «gnaw-m: mob—052......“ Sumoaqmoa momgmxmaq SEE; SA 35553 flames? Eugene—ax Engages $3qu >3 HS mag game-um... a? Eda-Sang S m 8935 Imaqmafi o mam $5. 39323 535.39. Damage. H mm 5... Pa 2.... 82 a: :28 _ as as... 9.0 5.0 119 1 719 F1" GST-190(1-719) IGSL] l [3" T” 63 719 i' I GST-190(63-719) Esrffi I l. ,:l 1 505 I T GST-190(1-505) l I“, T 63 505 I I GST-190(63-505) r—jesr I. I l fl 1 2 g a SNAP190 dep NE 8 I a %_ mSAN mSAC mSAN-I-C j J O i l. "r - -.. N , ’]—us ..“”-~--“-- I. * 1 120 Heal—U1 5 234567891011121314 SNAP190 deletion mutants diminish phosphorylation but remain functional for transcription. Computer-assisted screening (Blom et al., 1999) of the amino acid sequence of SNAP190 (1-719) reveals a total of thirteen putative CK2 phosphorylation sites containing serines (highlighted in Figure III-4 A, bottom panel). There are six sites clustered in the N-terminal region (Serzs, Ser”, Set”, Ser“, Sets9 and Set“) and three 540 541 542 , Ser sites in the serine rich region (Ser , and Ser ). Because mass spectrometric analysis of SNAP190 (1-719) revealed that at least 4 serines are phosphorylated in the N- tenninal region and up to 20 in the serine rich region. We therefore assumed that a truncated SNAP190 lacking the N-terminal region and/or the serine rich region would be a poor substrate for CK2 phosphorylation. The GST fusion deletion mutants containing SNAP190 (63-719), SNAP190 (1-505), and SNAP190 (63-505) were expressed in E. coli, purified through glutathione sepharose beads (Figure III-4 B lane 2, right panel), and tested for CK2 phosphorylation in an in vitro kinase assay by recombinant CK2. As shown in Figure III-4 B lane 1, as compared to GST-SNAP190 (1-719), GST-SNAP190 (63-719) phosphorylation was slightly decreased, while GST-SNAP190 (1-505) phosphorylation was dramatically reduced, indicating that the serine rich region we mapped harbors the majority of CK2 phosphorylation sites within SNAP190 (1-719). The truncated polypeptide lacking both the N-terminal region and the serine rich region GST- SNAP19O (63-505) was phosphorylated less well than GST-SNAP190 (1-719) and GST- SNAP19O (1-505). Collectively, these findings demonstrate that CK2 phosphorylates SNAP190 (1-719) predominantly at two regions aa (20—63) and aa (514-545). 121 Next, we asked whether these SNAP190 deletion mutants are capable of reconstituting snRN A gene transcription in an in vitro transcription system. Partial SNAP complexes containing full-length SNAP43, full-length SNAPSO, and various truncated SNAP190 molecules mS, mSAN, mSAC, and mSA(N+C) were assembled in E. coli and tested for function in transcription. In our experiments, a titration for each complex was performed and showed that all four complexes can support both U1 and U6 snRNA gene transcription equally well using SNAPc depleted HeLa nuclear extracts in vitro (Figure III-4 C). Thus, these recombinant mini-SNAP complexes are capable of mediating transcription by both RNA polymerases II and III. Thus, the regions of a (1-62) and/or a (506-719) are not essential for SNAPC activity and may play a regulatory role on SNAPC function. The region of SNAP190 (506-719) is required for CK2 inhibition. To further investigate a role of CK2 phosphorylation in snRNA transcription, we tested the SNAPC function in the presence of recombinant CK2 using the partial SNAP complexes m8 and mSAC. As shown in Figure III-5 A, both mini-SNAP complexes mS and mSAC were able to reconstitute U6 transcription in the SNAPc-depleted HeLa nuclear extracts (comparing lanes 3 and 6 to lane 2). However, when mS was preincubated with CK2 at 30°C for 30 minutes in the presence of ATP, U6 transcription was decreased remarkably (lane 3), while addition of same amount of CK2 and ATP without preincubation did not affect U6 transcription (lane 5), indicating that allowing mS phosphorylation by CK2 causes U6 transcription downregulation. Moreover, when 122 Figure III-5. A functional comparison between mini-SNAP complexes mS and mSAC. (A) U6 transcription of m8 and mSAC showed differential sensitivity upon CK2 treatment. Approximately 100 ng of purified mS and mSAC were used in each reaction. SNAPC were preincubated with either Dignam buffer D (lanes 3, 5, 6, and 8) or Dignam buffer D containing 20 units of recombinant CK2 and 1 mM ATP for 30 min. The reactions in lane 5 and 8 were then complemented with equal concentration of CK2 and ATP prior to initiation of transcription. The correct transcripts are labeled U6. (B) PSE- specific DNA binding by mS and mSAC was assayed by EMSA. Approximately 20 ng of purified mini-SNAP complex mS (lanes 2 ~ 7) and mSAC (lanes 8 ~ 13) were preincubated alone (lanes 2 and 8) or with 5 units (lanes 3, 6, 9, and 12) or 50 units (lanes 4, 7,10,13, and 14) of CK2 and/or 1 mM ATP (lanes 5 ~ 7 and 11 ~ 14) for 30 min at 30°C. EMSA was then performed using a DNA probe containing a wild-type mouse U6 PSE with mutant human U6 TATA box as described previously (Mittal and Hernandez, 1997), and incubated for an additional 30 min at 30°C (left panel). Alternatively, EMSA was performed by preincubating mS (Lanes 15 ~ 17) and mSAC (Lanes 18 ~ 20) with the DNA probe for 30 min at 30°C (lanes 8 ~ 10). Then, 5 units (lanes 16 and 19) or 50 units (lanes 17, 20 and 21) of CK2 and/or 1 mM ATP (Lanes 8 ~ 10) were added and reactions were incubated for an additional 30 min at 30°C (right panel). The reaction shown in lane 1 contains probe alone, and the reaction in lanes 14 and 21 contained only 50 units CK2 and 1 mM ATP. The positions of the protein/DNA complexes are indicated. 123 mS transcription start ‘0 ¥_30 min 1g). min ICK2+ATP (30 min preincubation) ICK2+ATP (0 min preincubation) m E aSNAP43 depleted NE r %_ mS mSAC G — l fir I (.3 _ _ + + — + + CK2+ATP 30 0 30 0 min preincubation 124 0 30min 60min 3 r i a IIMini-SNAPc+CK2/ATP +Probe J b FMini-SNAPc+Probe +CK2/ATP l a mS mSAC b m8 mSAC - F u l- r II I— mS or mSAC _—4-4—4-A+ -‘ —A+CK2 --—-+++———++++ +++ ++++ ATP m PSE .. .. - e. .0...- u ‘ O.- + mglAC/PSE 172 3 4 5 6 7 8 91011121314 15161718192021 125 mSAC and CK2 were preincubated and tested for U6 transcription, no difference was observed compared to the same sample without preincubation (comparing lane 7 to lane 8), suggesting that the region of SNAP190 (506-719) is critical for CK2 inhibition. As mentioned previously, the CK2 phosphorylated region aa (514-545) is adjacent to the Myb DNA binding domain (Figure III-4 A), and CK2 phosphorylation of SNAP190 at the region aa (514-545) might block the ability of the Myb domain to recognize the PSE sequence. To test this, EMSA was performed with approximately equal amounts of mS or mSAC that was preincubated with either CK2 or ATP or both for 30 minutes, followed by addition of a DNA probe. As shown in Figure III-5 B (panel a), robust DNA binding was observed by Iris (lane 2) and mSAC (lane 8), even in the presence of ATP (lanes 5 and 11) or increased amounts of CK2 (lanes 3—4 and 9-10). However, preincubation of mS with ATP along with CK2 diminished mS/PSE complex formation (lanes 6 and 7), but did not affect mSAC /PSE complex formation (lanes 12 and 13). These data strongly suggest that CK2 phopshorylation of SNAP190 (506-719) plays a negative role in SNAPC DNA binding. SNAPC DNA binding can be prevented by CK2 phosphorylation of SNAPC (Figure III-5 B, panel a), thus, whether the SNAPc/DNA complex can be disrupted by CK2 phosphorylation of SNAPC after DNA binding had occurred was tested. To address this, EMSAs was performed by allowing SNAPc/DNA complexes to form followed by CK2 phosphorylation. As shown in Figure III-5 B (panel b), when mS bound to PSE, subsequent addition of CK2 and ATP leads to less complex formation (comparing lanes 16 and 17 to lane 15). A similar response for mSAC mutant was not observed. Thus, 126 CK2 phosphorylation in the serine rich region of SNAP190 can inhibit SNAPc PSE binding both before and after DNA binding has occurred. Taken together, we conclude that the region of SNAP190 (506-719) is critical for CK2 inhibition. CK2 targets SNAP190 for phosphorylation in the serine rich region, which in turn inhibits SNAPc DNA binding activity and transcription properties. Discussion Previously I have demonstrated that CK2 inhibits overall U1 snRNA gene transcription in human HeLa cells. CK2 targets the snRNA activating protein complex (SNAPC) for phosphorylation. Here, I provide additional evidence that CK2 may downregulate snRN A gene transcription by inhibiting SNAPC DNA binding and transcription activity. The CK2 phosphorylation sites within SNAP190 were identified by mass spectrometry, and the region in SNAP190 that is required for CK2 inhibition was narrowed down to the region between aa 506-719 encompassing a serine rich region with multiple CK2 consensus sites. It has been determined that SNAPC PSE-specific DNA binding activity was inhibited by CK2 (Figure II-6). The mechanism of the inhibitory effect of CK2 phosphorylation on SNAPC DNA binding is not known; however, it is unlikely due to phosphorylation at a critical residue within the SNAP190 Myb domain, because this same complex is capable of binding DNA when TBP is present (Figure II-6). For the same reason, that phosphorylation outside the Myb DNA binding domain would affect intermolecular interactions between SNAP190 and SNAP43 is unlikely. Instead, we 127 favor the idea that CK2 phosphorylates SNAP190 outside the Myb DNA binding domain, leading to intramolecular interactions in SNAP190 that block SNAP190 Myb domain PSE recognition. Two CK2 target regions within SNAP190 (1-719) have been identifed by mass spectrometric analysis. One region (aa 20-63) is located in the N-terminal region upstream of the SNAP43 interacting region (Ma and Hernandez, 2001), consistent with our prediction that the SNAP43 interacting region within SNAP190 is not a CK2 target. Another region (aa 514-545) is located in the serine rich region downstream of the Myb-DNA binding domain. Deletion mutant of SNAP190 (1-505) lacking the serine rich region was generated and tested for transcriptional properties in the presence of CK2. Both DNA binding and transcription activities of mSAC were not affected by CK2 (Figure III-5), while mS was sensitive to CK2 treatment, suggesting that CK2 phosphorylation of SNAP190 in the serine rich region downregulates SNAPc DNA binding and transcription activity. It is interesting that the serine rich region with CK2 consensus sites in SNAP190 is conserved between mouse (accession number: AK077522.1) and human, thus, if CK2 phosphorylation of this serine rich region has a physiological function, it could be restricted to those organisms. It has been proposed previously that the fiill-length SNAP190 contains a built-in damper located in the region from a 506-1469, which downregulates SNAP190 binding to DNA (Mittal et al., 1999). It will be interesting to determine the exact location of this damper. In our experiments, when comparing the DNA binding activities of SNAP complexes m8 and mSAC using equal concentrations, we observed that mSAC has higher DNA binding affinity than mS (Figure III-5 B), raising the possibility that the region of SNAP190 aa (506-719) acts as a damper of DNA binding. Within SNAP 190, there is an 128 arginine rich region located between the Myb domain and the serine rich region (illustrated in Figure III-4 A) and we speculate that a conformational change in SNAP190 mediated by the interaction between the negatively charged residues (Glum’ 544’ ”d 546, 545) and the positively charged arginines is unfavorable for SNAPc/PSE complex and Asp formation. CK2 phosphorylation introduces more negative charges in the serine rich region, which will strengthen the interaction between the serine rich region and the arginine rich region, and make it more unfavorable for the PSE recognition. The observation that the weak DNA binding activity of mS was further diminished by CK2 treatment (Figure III-5 B) further supports this charge effect mechanism for inhibition of DNA binding. Another interesting finding in this study is that SNAPC DNA binding inhibited by CK2 occurred both before and after SNAPc/PSE complex had formed (Figure III-5). This observation raises the possibility that the CK2 phosphorylation sites within SNAPc might be exposed so that CK2 can reach them even when SNAP190 is complexed with the PSE. CK2 phosphorylation not only prevents the free SNAPc from binding DNA, but it can also increase the off-rate of SNAPC from the promoter. This double checkpoint mechanism ensures a maximum CK2 inhibition on snRNA gene transcription. 129 References Armstrong, S.A., Barry, D.A., Leggett, R.W., Mueller, CR. (1997). Casein kinase II- mediated phosphorylation of the C terminus Spl decreases its DNA binding activity. J Biol Chem 272, 13489-13495. Bai, L., Wang, Z., Yoon, J .B., Roeder, R.G. (1996). Cloning and characterization of the beta subunit of human proximal sequence element-binding transcription factor and its involvement in transcription of small nuclear RNA genes by RNA polymerases II and III. Mol Cell Biol 16, 5419-5426. Bieker, J .J ., Ouyang, L., Chen, X. (1998). Transcriptional factors for specific globin genes. Ann NY Acad Sci 850, 64-69. Byford, M.F. (1991). Rapid and selective modification of phosphoserine residues catalysed by Ba2+ ions for their detection during peptide microsequencing. Biochem J 280, 261-265. Cabrejos, M.B., Allende, C.C., Maldonado, E. (2004). Effects of phosphorylation by protein kinase CK2 on the human basal components of the RNA polymerase II transcription machinery. J Cell Biochem 93, 2-10. Dahmus, ME. (1981). Purification and Properties of Calf Thymus Casein Kinases I and II. J Biol Chem 256, 3332-3339. Dignam, J .D. (1990). Preparation of extracts from higher eukaryotes. Methods Enzymol 182, 194-203. Ford, E., Strubin, M., Hernandez, N. (1998). The Oct-1 POU domain activates snRNA gene transcription by contacting a region in the SNAPC largest subunit that bears sequence similarities to the Oct-1 coactivator OBF -1. Genes Dev 12, 3528-3 540. Ghavidel, A., Hockman, D.J., Schultz, M.C. (1999). A review of progress towards elucidating the role of protein kinase CK2 in polymerase III transcription, regulation of the TATA binding protein. Mol Cell Biochem 191, 143-148. Ghavidel, A., Schultz, M.C. (1997). Casein kinase II regulation of yeast TFIIIB is mediated by the TATA-binding protein. Genes Dev 11, 2780-2789. Ghavidel, A., Schultz, M. C. (2001). TATA binding protein-associated CK2 transduces DNA damage signals to the RNA polymerase III transcriptional machinery. Cell 106,575-584. 130 Guerra, B., Issinger, O.G. (1999). Protein kinase CK2 and its role in cellular proliferation, development and pathology. Electrophoresis. 20, 391-408. Hannan, R.D., Hempel, W.M., Cavanaugh, A., Arino, T., Dimitrov, S.I., Moss, T., Rothblum, L. (1998). Affinity purification of mammalian RNA polymerase 1. Identification of an associated kinase. J Biol Chem 273, 1257-1267. Henry, R.W., Ford, E., Mital, R., Mittal, V., Hernandez, N. (1998a).Crossing the line between RNA polymerases, transcription of human snRN A genes by RNA polymerases II and 111. Cold Spring Harb Symp Quant Biol 63, 111-120. Henry, R.W., Ma, B., Kobayashi, R., Hernandez, N. (1998b). SNAP19 mediates the assembly of a functional core promoter complex (SNAPC) shared by RNA polymerases II and III. Genes Dev 12, 2664-2672. Henry, R.W., Mittal, V., Ma, B., Sadowski, C.L., Kobayashi, R., Hernandez, N. (1996). Cloning and characterization of SNAPSO, a subunit of the snRNA-activating protein complex SNAPC. EMBO J 15, 7129-7136. Henry, R.W., Sadowski, C.L., Kobayashi, R., Hernandez, N. (1995). A TBP-TAP complex required for transcription of human snRNA genes by RNA polymerases II and 111. Nature 374, 653-657. Hinkley, C.S., Hirsch, H.A., Gu, L., LaMere, B., Henry, R.W. (2003). The Small Nuclear RNA-activating Protein 190 Myb DNA Binding Domain Stimulates TATA Box-binding Protein-TATA Box Recognition. J Biol Chem 278, 18649-18657. Hovde, S., Hinkley, C.S., Strong, K., Brooks, A., Gu, L., Henry, R.W., Geiger, J. (2002). Activator recruitment by the general transcription machinery, X-ray structural analysis of the Oct-1 POU domain/human U1 octamer/SNAP190 peptide ternary complex. Genes Dev 16, 2772-2777. Hu, P., Samudre, K., Wu, S., Sun, Y., Hernandez, N. (2004). CK2 phosphorylation of del executes cell cycle-specific RNA polymerase III transcription repression. Mol Cell 16, 81-92. Hu, P., Wu, S., Hernandez, N. (2003). A minimal RNA polymerase III transcription system from human cells reveals positive and negative regulatory roles for CK2. Mol Cell 12, 699-709. Jiang, X., Wang, Y. (2004). Beta-elimination coupled with tandem mass spectrometry for the identification of in vivo and in vitro phosphorylation sites in maize dehydrin DHNl protein. Biochemistry 43, 15567-15576. 131 Johnston, I.M., Allison, S.J., Morton, J.P., Schramm, L., Scott, P.H., White, RI. (2002). CK2 forms a stable complex with TFIIIB and activates RNA polymerase III transcription in human cells. Mol Cell Biol 22, 3757-3768. Liao, P.C., Leykam, J ., Andrews, P.C., Gage, D.A., Allison, J. (1994). An approach to locate phosphorylation sites in a phosphoprotein: mass mapping by combining specific enzymatic degradation with matrix-assisted laser desorption/ionization mass spectrometry. Anal Biochem 219, 9-20. Lobo, S.M., Hernandez, N. (1989). A 7 bp mutation converts a human RNA polymerase II snRNA promoter into an RNA polymerase III promoter. Cell 58, 55-67. Ma, B., Hernandez, N. (2001). A Map of Protein-Protein Contacts within the Small Nuclear RNA-activating Protein Complex SNAPc. J Biol Chem 276, 5027-5035. Ma, B., Hernandez, N. (2002). Redundant cooperative interactions for assembly of a human U6 transcription initiation complex. Mol Cell Biol 22, 8067-8078. Meggio, F., Pinna, L. A. (2003). One-thousand-and-one substrates of protein kinase CK2? The FASEB Journal 17, 349-368. Meisner, H., Heller-Harrison, R., Buxton, J ., Czech, MP. (1989). Molecular cloning of the human casein kinase II alpha subunit Biochemistry 28, 4072-4076. Mittal, V., Cleary, M.A., Herr, W., Hernandez, N. (1996). The Oct-1 POU-specific domain can stimulate small nuclear RNA gene transcription by stabilizing the basal transcription complex SNAPC. Mol Cell Biol 16, 1955-1965. Mittal, V., Ma, B., Hernandez, N. (1999). SNAPC, a core promoter factor with a built-in DNA-binding damper that is deactivated by the Oct-1 POU domain. Genes Dev 13, 1807-1821. Murphy, S., Yoon, J .B., Gerster, T., Roeder, R.G. (1992). Oct-l and Oct-2 potentiate functional interactions of a transcription factor with the proximal sequence element of small nuclear RNA genes. Mol Cell. Biol 12, 3247—3261. Pinna, L.A. (2002). Protein kinase CK2, a challenge to canons. Journal of Cell Science 115, 3873-3878 Pinna, L.A., Meggio, F. (1997). Protein kinase CK2 ("casein kinase-2") and its implication in cell division and proliferation. Prog Cell Cycle Res 3, 77-97. Resing, K.A., Johnson, R.S., Walsh, K.A. (1995). Mass spectrometric analysis of 21 phosphorylation sites in the internal repeat of rat profilaggrin, precursor of an intermediate filament associated protein. Biochemistry 34, 9477-87. 132 Sadowski, C.L., Henry, R.W., Kobayashi, R., Hernandez, N. (1996). The SNAP45 subunit of the small nuclear RNA (snRN A) activating protein complex is required for RNA polymerase II and III snRNA gene transcription and interacts with the TATA box binding protein. Proc Natl Acad Sci U S A 93, 4289-4293. Sadowski, C.L., Henry, R.W., Lobo, S.M., Hernandez, N. (1993). Targeting TBP to a non-TATA box cis-regulatory element, a TBP-containing complex activates transcription from snRNA promoters through the PSE. Genes Dev 7, 1535-1548. Tsutsui, H., Geltinger, C., Murata, T., Itakura, K., Wada, T., Handa, H., Yokoyama, K.K. (1999). The DNA-binding and transcriptional activities of MAZ, a myc-associated zinc finger protein, are regulated by casein kinase II. Biochem Biophys Res Commun 262, 198-205. Waldschmidt, R., Wanandi, I., Seifart, K.H. (1991). Identification of transcription factors required for the expression of mammalian U6 genes in vitro. EMBO J 10, 2595-2603. Wong, M.W., Henry, R.W., Ma, B., Kobayashi, R., Klages, N., Matthias, P., Strubin, M., Hernandez, N. (1998). The large subunit of basal transcription factor SNAPc is a Myb domain protein that interacts with Oct-1. Mol Cell Biol 18, 368-377. Yoon, J .B., Roeder, R.G. (1996). Cloning of two proximal sequence element-binding transcription factor subunits (gamma and delta) that are required for transcription of small nuclear RNA genes by RNA polymerases II and III and interact with the TATA-binding protein. Mol Cell Biol 16, 1-9. Zhao, X., Pendergrast, P.S., Hernandez, N. (2001). A positioned nucleosome on the human U6 promoter allows recruitment of SNAPC by the Oct-1 POU domain. Mol Cell 7, 539-549. 133 CHAPTER FOUR SUMMARY AND FUTURE PLAN CK2 is a highly conserved, ubiquitous kinase that appears to influence non- protein coding gene transcription, such as RNA pol I-specific rRNA and RNA pol III- specific tRNA, 5S rRNA and U6 snRNA gene transcription. Whether CK2 regulates RNA pol II-specific snRNA gene transcription has been uncertain. Here we report that CK2 regulates U1 snRNA gene transcription by RNA pol II. Inhibition of CK2 by either depleting it using RNAi in cells (Figure II-l B) or blocking CK2 activity with its specific inhibitor in human cell extracts (Figure II-2 B) stimulates U1 snRNA synthesis. The promoter occupancies of both CKZa- and CKZB subunits on the U1 gene in human cells (Figure II-l A) provide further evidence that CK2 is in close proximity with U1 transcriptional machinery. Thus, these observations demonstrate that CK2 is involved in regulating RNA pol II-specific snRN A transcription. Previously, it has been demonstrated that CK2 targets TFIIIB and RNA pol III for RNA pol III-specific U6 snRNA transcription regulation (Hu et al., 2003; Hu etal., 2004). However, TFIIIB or RNA pol III are not required for U1 transcription by RNA pol II. One candidate target for CK2 regulation is the basal transcription factor SNAPC, which is used for all snRNA gene transcription by both RNA pol II and pol III. SNAPc plays a pivotal role in snRN A gene transcription by providing core promoter recognition, serving as a target for transcription activation by Oct-1, and coordinating TBP activity thereby promoting preinitiation complex assembly for both RNA pols II and III. Indeed, 134 endogenous CK2 associates with SNAPC and phosphorylates the SNAP190 subunit (Figure II-3). SNAP190 likely plays a central role in coordinating SNAPC phosphorylation by recruiting CK2 to the complex. This idea is supported by the observation that GST-SNAP190 (1-719) can recruit CK2 from HeLa cell nuclear extracts (Figure II-S A). Although CK2 is involved in regulating snRNA gene transcription by both RNA pol II and III, the mechanism of its function is unclear. Hu et al. (2003; 2004) discovered that CK2 stimulates U6 snRNA gene transcription by targeting RNA pol 111. On the other hand, CK2 inhibits U6 transcription by phosphorylating del. Notably, CK2 treatment of recombinant SNAPc did not affect U6 transcription in their minimal RNA pol 111 system (Hu et al., 2003). Our experiments demonstrate that mini-SNAPC phosphorylation by CK2 inhibits its DNA binding activity and TBP is capable of counteracting this inhibition. As shown in Figure II-6, CK2 inhibition of SNAPc DNA binding is not altered by the addition of TBP for the U1 like probe (Figure II-6, right panel). This observation supports the proposal that downregulation of SNAPc DNA binding activity by CK2 may contribute to reduced U1 snRN A gene transcription. However, CK2 inhibition can be reversed by the addition of TBP for the U6 like probe (Figure II-6, middle panel), which can explain why U6 snRNA gene transcription was not affected by CK2 phosphorylation of SNAPc (Hu etal., 2003). These findings indicate that cooperation between SNAPc and TBP antagonizes CK2 inhibition of DNA binding by SNAPC. It has been demonstrated that CK2 targets SNAPC for phosphorylation. Whether CK2 phosphorylation of SNAPC directly contributes to CK2 downregulation of U1 snRNA gene transcription was tested. As a first, I have established an in vitro 135 transcription system wherein the endogenous SNAPc was immuno-depleted and reconstituted with recombinant E. coli assembled mini-SNAPC to support transcription (Figure III-1 and Figure III-4 B). In this system, mini-SNAP complexes alone are capable of reconstituting snRNA gene transcription by both RNA pol II and pol III in the SNAPC- depleted HeLa nuclear extracts without addition of recombinant TBP and ATP mix (0.3 M ATP, 10 Kg of phosphocreatine kinase per ml, and 10 mM creatine kinase) (Ma and Hernandez, 2002). Next, this system was used to directly test the effect of SNAPc phosphorylation by CK2 for transcription. I observed that preincubation of SNAPC with CK2 and ATP exhibited less U1 (Figure III-1) and U6 snRNA transcripts (Figure III-5 A), suggesting that CK2 phosphorylation inhibits SNAPC transcription activity. To further investigate the mechanism of SNAPc phosphorylation by CK2, phosphopeptide mapping of SNAP190 (1-719) was performed by mass spectrometry. The MS data revealed that SNAP190 was phosphorylated at two regions that contain multiple CK2 consensus sites [amino acid (a) 20-63 and 514-545]. As the serine rich region from amino acids 514-545 represents the majority of CK2 phosphorylation within SNAP190 (1-719) (Figure III-3 B and Figure III-4 B), the importance of this region for CK2 regulation of SNAPC function was tested. Mini-SNAP complexes containing SNAP43, SNAPSO, and SNAP190 (1-719) (mS) or SNAP190 lacking the serine rich region SNAP190 (1-505) (mSAC) were compared for DNA binding and transcription activity in the presence of CK2. As described in Figure III-5, mS showed lower DNA binding affinity than mSAC and it was sensitive to CK2 treatment. Thus, SNAP190 (506- 719) plays an important role in downregulating SNAPC DNA binding activity and CK2 phosphorylation enhances this negative regulation. 136 It has been determined that SNAPc PSE-specific DNA binding activity was inhibited by CK2 (Figure II-6) and the region of SNAP190 (506—719) is required for CK2 inhibition (Figure III-5). The mechanism of the inhibitory effect of CK2 phosphorylation on SNAPC DNA binding is not known. We favor the idea that CK2 phosphorylates SNAP190 outside the Myb DNA binding domain, leading to intramolecular interactions in SNAP190 that masks PSE recognition by the SNAP190 Myb domain. An alignment of the SNAP190 Myb domain with the human c-Myb protein showed that SNAP190 Rd repeat (a 451-503) and c-Myb R3 repeats have 30% identity (Wong et al., 1998). NMR studies of c-Myb indicate that the R3 repeat is composed of three helices (Jamin et al., 1993). Computational modeling of SNAP190 (1-719) revealed that within the SNAP190 Myb domain, Rd repeat may also contain three helices with the last helix extended to residue 510, meaning that the arginine rich region (a 506-511) is part of the helix- turn- helix (HTH) motifs important for DNA binding. Therefore, I hypothesized a charge- directed mechanism in which the heavily negative charges in the serine rich region introduced by CK2 phosphorylation can attract the positively charged arginines and may influence the HTH motif in the DNA binding domain. To test this idea, I have made the non-phosphorylatable mutant complex by disrupting the CK2 consensus sites in SNAP190 (EED543'545—>AAA) and the phospho-mimetic mutant with S—+D substitution (SSSS549'542—»DDDD). My preliminary data shows that DNA binding activity of the non- phosphorylatable mutant mS (EED543'545—>AAA) is less sensitive to CK2 treatment, while the phospho-mimetic mutant mS (SSSS549'542—>DDDD) itself exhibites weak DNA binding compared to wild type mS, suggesting a charge effect mechanism. If so, then a mutant with R—+Q substitution (RRRRR506'5“—+QQQQQ) in SNAP190 will deactivate 137 the CK2 inhibition by phophorylating the serine rich region. This mutant has been constructed. The ability of DNA binding and transcription of this mutant can be tested in an EMSA and in vitro reconstitution transctiption system. To explore whether the N-terminal region (a 20-63) of SNAP190 also contributes to CK2 inhibition, the non-phosphorylatable mutant with SNAP190 (861.2835, 39’ 41’ 59—*AAAAA) was generated and tested for DNA binding activity. My preliminary data shows that DNA binding activity of the non-phosphorylatable mS (Serzs' 35' 39’ 41’ 59—->AAAAA) is less sensitive to CK2 treatment compared to wild type mS. However, the double mutant ms (Ser28’35’39’4"59-—»AAAAA /EED543'545—iAAA) DNA binding did not alter upon CK2 treatment. These results suggest that CK2 phosphorylation at both regions (aa 20-63 and aa 514—545) of SNAP190 negatively regulates SNAPC function. Further experiments to test mutant SNAPC DNA binding activity and their abilities to direct snRN A transcription are needed to understand the mechanism. It has been proposed that in the full length SNAPc, SNAP190 had a built-in damper located within the C-terminal two-thirds of SNAP190 aa (506-1469) to downregulate its binding to DNA, which was deactivated by a direct protein-protein contact between SNAP190 (aa 869-912) and the Oct-1 POU domain (Mittal et al., 1999). In the context of mini-SNAP complexes, upon CK2 treatment, DNA binding activity of mS containing SNAP190 aa (1-719) was greatly reduced, while mSAC lacking the serine rich region did not change (Figure III-5), however, mS(EED543'545—+AAA) with the non- phosphorylatable serine rich region was also reduced, but to a lesser degree comparing to mS. These observations imply that the region of SNAP190 aa 506-719 probably acts as a damper of DNA binding. As the cooperative binding between TBP and SNAPC can 138 overcome the CK2 inhibition (Figure II-6), it is not clear whether TBP functions through direct contact with the CK2 phosphorylated region of SNAP190 (a 20-63), or TBP has higher affinity to interact with SNAP190 RcRd than the CK2 phosphorylated region of SNAP190 (aa 514-545) in order to release CK2 inhibition. To test this, TBP recruitment to the promoter DNA by the various deletions or point-mutations within mini-SNAP complexes can be measured by EMSA. The region within SNAP190 that is targeted by CK2 to inhibit DNA recognition can be determined by comparing the deletions or mutations in SNAP190 in the context of mini-SNAPC. Thus, we might be able to distinguish which region contributes to DNA recognition inhibition and the mechanism for TBP antagonizing fimction. Previously, we had demonstrated that SNAP190 is phosphorylated in vivo (Figure II-4). Endogenous SNAP190 has been reported to be phosphorylated in HeLa cells (Beausoleil et al., 2004). It is unclear, however, whether CK2 sites in SNAP190 we mapped in vitro are also phosphorylated in the cells. Raising antisera specifically recognize phosphoserines in SNAP190 (anti-phospho-SNAP19O antibody) will help us to answer the questions about whether the CK2 sites in SNAP190 are also phosphorylated in the cells. It will also be interesting to know whether SNAP190 phosphorylation by CK2 is regulated in a cell cycle dependent manner and/or whether the level of CK2 phosphorylation correlates with snRNA gene transcription. In conclusion, CK2 negatively regulates human U1 snRNA gene transcription by inhibiting the basal transcription factor SNAPC DNA binding and transcription activity. The cooperation between SNAPC and TBP can antagonize CK2 inhibition of DNA 139 binding by SNAPC, implying that CK2 may differentially regulate snRNA gene transcription by different RNA polymerases. 140 References Beausoleil, S. A., Jedrychowski, M., Schwartz, D., Elias, J.E.,Villén, J., Li, J., Cohn, M. A., Cantley, L.C., Steven P. Gygi, SP. (2004). Large-scale characterization of HeLa cell nuclear phosphoproteins. PNAS 101, 12130-12135. Hu, P., Samudre, K., Wu, S., Sun, Y., Hernandez, N. (2004). CK2 phosphorylation of del executes cell cycle-specific RNA polymerase III transcription repression. Mol Cell 16, 81-92. Hu, P., Wu, S., Hernandez, N. (2003). A minimal RNA polymerase III transcription system from human cells reveals positive and negative regulatory roles for CK2. Mol Cell 12, 699-709. Jamin, N., Gabrielsen, O.S., Gilles, N., Lirsac, P.N., Toma, F. (1993). Secondary structure of the DNA-binding domain of the c-Myb oncoprotein in solution. A multidimensional double and triple heteronuclear NMR study. Eur J Biochem 216, 147— 154. Ma, B., Hernandez, N. (2002). Redundant cooperative interactions for assembly of a human U6 transcription initiation complex. Mol Cell Biol 22, 8067-8078. Mittal, V., Ma, B., Hernandez, N. (1999). SNAP(c): a core promoter factor with a built-in DNA-binding damper that is deactivated by the Oct-1 POU domain. Genes Dev 13, 1807- 1821. Wong MW, Henry RW, Ma B, Kobayashi R, Klages N, Matthias P, Strubin M, and Hernandez N. (1998). The large subunit of basal transcription factor SNAPC is a Myb domain protein that interacts with Oct-l. Mol. Cell. Biol. 18:368-377. 141