.. .2. .05.... .. ...A 115...... x s .9 .3. al.— §._‘!Vé -“-J( 53' ll I9514 I Jams! .‘11! i1 15.... I 3391:: 1"). ..¢ ; 73““..3173 .1: 2 ..fix.:. :. £252.12 .1 1:4 . .. first .Shuxfi. nu... :3 3.4.31.1? v ,a 4 l... I . x . u . , «SnfidfimfiflWfimu a .x 5.334.. «9.3.0.7... , Kr. . .. fig)...“ 0 v . Va}: 3 .. .311... "I... I3: VAR»... .«uwmn .. 83v: ‘ . .11.?! 3 .. 411...: . 11......95 3...; .33 69.519. . . 2‘21 .3 .5 _ )f E... v gdwb‘. {- ' .VI 5:. 9.1: ~\‘b Z :v3vx: . .ul‘ » u.- .I l “53’“ 10%;? This is to certify that the dissertation entitled TRANSCRIPTIONAL REGULATION OF THE HUMAN SMALL NUCLEAR RNA GENE FAMILY presented by Gauri W. Jawdekar has been accepted towards fulfillment of the requirements for the PhD. degree in Microbiology and Molecular Genetics 5/ a”; ’ Major erssor’s Signature 41(94qu X3; 200$ Date MSU is an Afiinnativo ActiorVEquaI Opportunity Institution LIBRARY Michig-.. State University PLACE IN RETURN BOX to remove this checkout from your record. To AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 2/05 p:/CIRC/DateDue.indd-p.1 TRANSCRIPTIONAL REGULATION OF THE HUMAN SMALL NUCLEAR RNA GENE FAMILY By Gauri W. Jawdekar A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Microbiology and Molecular Genetics 2006 ABSTRACT TRANSCRIPTIONAL REGULATION OF THE HUMAN SMALL NUCLEAR RNA GENE FAMILY By Gauri W. Jawdekar Human RNA polymerase III synthesizes small, non-translated RNAs including SS rRNA, tRNA, and U6 snRNA that control numerous critical steps during the flow of genetic information in biological systems. U6 snRNA genes and other related family members are unusual because they are transcribed by either RNA polymerase II or III depending on the arrangement of core promoter elements. Thus human snRNA genes in general provide a good model to study polymerase preference and activity during normal and deregulated growth. One characteristic feature of all snRNA genes is the presence of an essential proximal sequence element (PSE) in the core promoter region that is recognized by the multi-subunit general transcription factor called SNAPC. DNA bindingby SNAPC is a crucial early event during preinitiation complex assembly, and is a target for regulatory intervention. The mechanism of DNA binding by SNAPc was investigated in this study. The SNAPSO subunit of SNAPC plays an important role in preinitiation complex assembly for RNA polymerase II and III transcription in a process involving an unorthodox but highly conserved zinc finger domain. This zinc finger domain functions directly in DNA binding and is essential for cooperative promoter recognition by SNAPc. The Retinoblastoma (RB) tumor suppressor protein represses U6 snRNA transcription by RNA polymerase III and interestingly can interact with the SNAPSO subunit of SNAPC suggesting that RB could impair DNA binding by SNAPC to repress transcription. However, studies from our lab suggest that RB does not affect DNA binding by SNAPC during repression. Therefore, to further understand the mechanism of RB repression of U6 snRNA transcription, I examined the role of RB co-factors such as histone deacetylases (HDACs) and components of the ATP-dependent SWI/SNF chromatin remodeling complex in RB repression. In this study I show that endogenous RB co-occupies the U6 snRNA promoter with HDACs and stimulates association of HDAC2 and Brgl with the U6 promoter during repression. Interestingly, HDAC enzymatic activity is required for RB repression of chromatin template but not naked DNA template, suggesting that RB repression and HDAC activity are biochemically separable. In a search for additional factors that associate with SNAPC and potentially regulate human snRNA transcription, a protein called HEXIMI was identified. In this study, I show that HEXIMI co-purifies with SNAPC and stimulates DNA binding by SNAPC and TBP. HEXIMI positively regulates 78K transcription by potentially enhancing preinitiation complex assembly by these factors. Interestingly, HEXIMI and 78K RNA are part of a complex that inhibits the activity of P-TEFb, which is required for transcription of HIV-1 and cellular genes by RNA polymerase 11. Our observation that HEXIMI stimulates transcription of its co-repressor partner suggests a mechanism by which P-TEFb activity and hence global RNA polymerase II transcription may be regulated. ACKNOWLEDGEMENTS I would earnestly like to thank my mentor R. William Henry for his guidance and excellent mentorship. His encouragement and patience were vital for me to continue with research and not quit graduate school. I wish to thank my thesis guidance committee: Dr. Susan Conrad, Dr. Lee Kroos, Dr. Richard Schwartz and Dr. Steve Triezenberg. I would like to thank Dr. James Geiger for a fruitful collaboration. I also want to thank Dr. David Arnosti and Dr. James Geiger for writing letters of recommendation for my post-doctoral job. Members of the Henry laboratory, past and present: Dr. Craig Hinkley, Dr. Heather Hirsch, Dr. Zakir Ulla, Dr. Anastasia Gridasova, Dr. LiPing Gu, Tharakeswari Selvakumar and Xianzhou Song have been great colleagues to work with and I am glad to have gained their friendship. I would like to thank Dr. Shibani Mukherjee for engaging in stimulating scientific discussions and help with new techniques. I am also thankful to Dr. Debbie Yoder-Himes and Rim Shanna for their friendship. Finally I would like to thank my parents, brother, sister-in law and my nephew for their love and support. They have always encouraged me to pursue my dreams, even if it meant living far away from them for extended periods of time. iv TABLE OF CONTENTS LIST OF FIGURES .............................................................................. vii LIST OF TABLES ................................................................................. ix KEY TO ABBREVIATIONS .................................................................... x CHAPTER 1: Introduction ......................................................................................... 1 RNA polymerase III transcription ................................................................. l 1. Diverse cellular functions of genes transcribed by RNAP III ..................... 1 1.]. RNAP III and information flow in biological systems .................. 4 1.2. RNAP III transcripts and cellular homeostasis ........................... 6 2. Regulation of RNAP III transcription ................................................. 7 2.1. Transcriptional regulation during normal grth ........................ 7 2.2. Transcriptional regulation and disease ..................................... 9 3. Mechanisms of RNAP III regulation- Promoter structure ......................... 11 4. Mechanisms of RNAP III regulation- General transcription factors ............. 15 5. Mechanisms of RNAP III regulation- Regulatory factors ......................... 19 5.1. Factors that positively regulate transcription ............................. 19 5.2. Factors that positively & negatively regulate transcription ............ 23 5.3. Factors that negatively influence transcription ........................... 24 CHAPTER 2: RB recruits HDAC activity for repression of human U6 snRNA transcription by RNA Polymerase III .............................................................................. 46 Abstract ...................................................................................... 46 Introduction ................................................................................. 47 Materials and Methods ..................................................................... 51 Results ....................................................................................... 54 Discussion ................................................................................... 68 References ................................................................................... 76 CHAPTER 3: The unorthodox SNAP50 zinc finger domain contributes to co-operative promoter recognition by human SNAPc .................................................................. 79 Abstract ...................................................................................... 79 Introduction ................................................................................. 80 Materials and Methods ..................................................................... 82 Results ....................................................................................... 86 Discussion ................................................................................. l 12 References ................................................................................. 1 19 CHAPTER 4: The HEXIMI repressor of P-TEFb stimulates transcription of its 78K snRNA co- repressor partner by RNA polymerase III .................................................. 124 Abstract ..................................................................................... 124 Introduction ................................................................................ 125 Materials and Methods ................................................................... 128 Results ...................................................................................... 131 Discussion .................................................................................. 153 References ................................................................................. 158 CHAPTER 5: Summary .......................................................................................... 163 APPENDIX A Multiple subunits of SNAPC co-expressed in E. coli are active for transcription by human RNA polymerase II and III .......................................................... 169 Results and Discussion ................................................................... 169 References ................................................................................. 176 APPENDIX B Differential U6 snRNA promoter association by RB family members ................ 177 Results and Discussion .................................................................. 177 References ................................................................................. 1 83 APPENDIX C RB recruitment to human U6 snRNA promoter through interactions with SNAPc and TFIIIB ........................................................................................ 184 Results and Discussion .................................................................. 184 References ................................................................................. 193 vi Figure 1-1. Figure 1-2. Figure 2-1. Figure 2-2. Figure 2-3. Figure 2-4. Figure 2-5. Figure 2-6. Figure 3-1. Figure 3-2. Figure 3-3. Figure 3-4. Figure 3-5. Figure 3-6. Figure 3-7. Figure 4-1. LIST OF FIGURES Schematic representation of the promoter elements of genes transcribed by RNA polymerase III and II transcribed .................................... 13 The general transcription machinery required for RNA polymerase III transcription ........................................................................ l7 Endogenous chromatin modifying proteins associate with a human U6 snRN A promoter ................................................... 56 RB concurrently occupies an endogenous U6 snRN A promoter with HDAC] , HDAC2 and RNA polymerase III ................................... 60 RB stimulates enrichment of HDACs and SWI/SNF at a U6 snRNA promoter during repression ...................................................... 63 RB repression of U6 transcription and HDAC activity are biochemically separable activities ................................................................ 67 RB requires HDAC activity to repress U6 transcription from a chromatin DNA template but not from a naked DNA template ......................... 70 Model for RB repression ......................................................... 74 The C-terminal region of SNAP50 is required for DNA binding by SNAPc .......................................................................... 88 Single amino acid changes in the SNAP50 subunit abolish DNA binding by SNAPC ................................................................ 96 The SNAP50 zinc finger domain is required from human snRNA gene transcription by RNA polymerase II and III ................................. 101 SNAPC recruits Ber-TFIIIB to U6 promoter DNA in vitro ............... 104 TFIIIB suppresses DNA binding defects in SNAPC ........................ 108 SNAP50 C-terminus binds zinc ................................................ 111 Model for the SNAP50 zinc finger domain .................................. 116 Endogenous HEIMI associates with SNAPC ................................ 134 vii Figure 4-2. Figure 4-3. Figure 4-4. Figure 4-5. Figure A-l. Figure A-2. Figure B Figure C-l. Figure C-2. HEXIMI cooperates with SNAPc and TBP for DNA binding ........... 139 HEXIMI occupies endogenous snRN A promoters ........................ 144 P-TEF b is differentially used for snRNA gene transcription by RNA polymerase II and III in vitro ................................................... 148 HEXIM] positively regulates 7SK snRNA transcription in viva ......... 151 mSNAPcy4 is competent for DNA binding and TBP promoter recruitment ........................................................................ 173 mSNAPcy4 supports human snRNA gene transcription in vitro by both RNA polymerase II and III ........................................... 175 Differential U6 snRN A promoter association by human RB family members .......................................................................... 179 Characterization of the regions in SNAP50 and de1 that are required for interactions with RB ........................................................ 187 The A/B pocket domain and the C-terminal region of RB is required for interactions with RNA polymerase III-general transcription machinery ........................................................................ 191 Images in this dissertation are presented in color. viii LIST OF TABLES Table 1-1 RNA polymerase III and II transcribed non—translated genes with the known cellular function ....... . ..................................................... 3 Table 3-1. Table showing zinc and nickel analysis of SNAPC ........................... 92 ix ATP de1 Brfl Brf2 CDK CHI-I ChIP CK2 Co-IP CSB CTD DNMTI DSE EDGl EMSA ERE FAA GAPDH GST KEY TO ABBREVIATIONS Adenosine triphosphate B double prime TFIIIB related protein 1 TFIIIB related protein2 Cyclin dependent kinase Cartilage-hair hypoplasia Chromatin immunoprecipitation Casein Kinase 2 I Co-immunoprecipitation Cokayne syndrome group B protein Carboxy terminal domain DNA methyl transferase 1 Distal sequence element Estrogen down-regulated gene 1 Electrophoretic mobility shift assay Estrogen receptor response element F lame atomic absorption spectroscopy Glyceraldehyde-3-phosphate dehydrogenase Glutathione S transferase Heamagglutinin Histone aoetylase HDAC HEXIMI HIV ICP-MS ICR LTR MBD mSNAPc mSNAPcy4 NLS PCR PNC PSE P-TEF b rRNA RT-PCR SANT Histone deacetylase Hexamethylene bis-acetamide inducible 1 Human immunodeficiency virus Heterochromatin protein 1 Inductively coupled plasma resonance mass spectroscopy Internal control region Long terminal repeat Methylated DNA binding protein Mitochondrial RNA processing mini-SNAPc mini-SNAPc gamma 4 Mutant Nuclear localization signal Polymerase chain reaction Perinucleolar compartment Proximal sequence element Positive transcription elongation factor b Retinoblastoma tumor suppressor RNA interference RNA polymerase Ribosomal RNA Reverse transcriptase polymerase chain reaction SWI-SNF, ADA, N-CoR and TFIIIB xi siRN A SNAPC snRNA snRNP SRP STAF SV40 TBP TFIIA TFIIB TFIID TFIIF TFIIH TFIIIA TFIIIB TFIIIC tRNA TSA UV VSMC vRNA WCE Wt Short interfering RNA Small nuclear RNA activating protein complex Small nuclear RNA Small nuclear ribonucleoprotein Signal recognition particle Selenocysteine tRN A gene transcription activating factor Simian virus 40 TATA- binding protein Transcription factor II A Transcription factor II B Transcription factor II D Transcription factor II F Transcription factor II H Transcription factor 111 A Transcription factor 111 B Transcription factor III C Transfer RNA Trichostatin A Ultra violet Vascular smooth muscle cells Vault RNA Whole cell extract Wild type xii CHAPTER 1 INTRODUCTION RNA polymerase III transcription In most eukaryotes three major related nuclear RNA polymerases are responsible for catalyzing the synthesis of RNA from DNA. RNA polymerase I is dedicated to transcribing only one set of genes, the tandemly repeated ribosomal RNA (rRNA) genes. RNA polymerase II transcribes protein coding messenger-RNA (mRNA) genes, micro RNA (miRNA), and some non-translated small nuclear RNA (snRNA) genes. In contrast, RNA polymerase III transcribes the well-studied SS ribosomal RNA (rRNA) and transfer RNA (tRN A) genes, and a diverse collection of small nuclear RNA (snRNA) genes. One common feature shared by these RNA polymerase III transcribed genes is that they encode non-translated RNAs that are almost always shorter than ~400 base pairs (111). I. Diverse cellular functions of genes transcribed by RNA polymerase III The products of RNA polymerase III transcription such as U6 snRNA, SS rRNA and tRNA have been best studied for their function in mRNA splicing and protein synthesis, respectively. However, recent evidence suggests that RNA polymerase III transcripts function for most, if not all, crucial steps during the flow of information in biological systems and for maintaining cellular homeostasis. Table 1 describes the various RNAs molecules transcribed by RNA polymerase III and their known functions. Table 1-1. Diverse cellular functions of some non-translated RNAs transcribed by RNA polymerase III and [1. RNA polymerase III transcribes non-translated RNAs that are characteristically less than 400 base pairs long. These gene products play an important role in diverse cellular processes indicated in the table. 8.8.8. 5 8:928. waves... 9:05.... 25883828: «3.8 E 85:: 23:. 5.3.. : 8 c8? 8 new 2. a 88288 226 =8 so: ea 98 =. 88> m £205 cm? =. 58:: (Zr... mm 9. 92% h0= R 5.3.3353 0 Banana m 96 g a-SNAP431P D[ I E <<<<<< <<<<<<<< s...c".‘."~.§.§ §§$§%§§§§§‘§§§§§ ‘35§oo:ozz: 10500 11:: ” www—h—¢ w ‘J-HASNAPSO l ~“*”~~*~“1~~*”“ 1% —|NPUT 1234 5 6 7 8 910111213141516171819202122 O mSNAPcH C-xs-C-----H-xa-C--x--H-x1o-H--H-x2"c-x12-H-xe- 'XZ'C'X19‘C’X2‘C'X2‘C“"'X4""'H W1 m-A C312-A 11313-A C317-Ai'1319-A WA HmI-AWA ”(HT-A C354-A C357-A C377-A m-A C383~A R385-A MA Free probe 12 3 4 5 6 7 8 91011121314151617181920212223242526272829308132333435 97 the extent of SNAP50 association, as a measure of complex integrity, was determined by Western blot analysis against the HA-tag contained at the N-terminus of SNAP50. In this approach, any SNAP50 detected in this analysis had to be in a complex with both SNAP43 and SNAP190 (1-505). As shown in Figure 3-2B, the amounts of SNAP50 recovered after the two-step affinity purification were similar for the wild type and mutant complexes, suggesting that SNAP50 assembly into SNAPC is not markedly dependent upon any individual cysteine or histidine in this domain. Next, partially purified wild type and mutant complexes were then tested for PSE binding function in EMSA to determine whether the cysteine and histidine residues within the SNAP finger domain are important for DNA binding by SNAPC. The results revealed three categories of DNA binding effects, as shown in Figure 3-2C. The first category of mutants exhibited either wild-type DNA binding (H330A, H347A, C377A, R385) or modest defects (C302A, C312A, H331A, C334A) as compared to the complex containing wild-type SNAP50. The second category of mutant complexes exhibited severe defects in DNA binding (H313A, C317A, H319A, H388A), while complexes in the third category were completely impaired for DNA binding ability (C354A, C357A, C3 80A, C3 83A). Thus, the cysteine and histidine residues within the latter two categories are both evolutionarily conserved and critical for DNA binding by SNAPC. The SNAP50 zinc finger domain is required for RNA polymerase II and III transcription. We next tested whether mutations that affected PSE-recognition by SNAPC were also critical for human U1 snRNA gene transcription by RNA polymerase II and U6 98 snRNA gene transcription by RNA polymerase III. For these experiments, we predicted that those complexes that exhibited no DNA binding ability would also be defective in transcription, confirming that DNA binding by SNAPC is an essential aspect of snRNA gene transcription. Secondly, any previously unappreciated function for SNAP50 in communication with other components of the general transcription machinery would be revealed as a defect in transcription for those complexes with wild type DNA binding activity. As the general transcription machinery for RNA polymerase II and III transcription are different, it was possible that certain mutations would result in transcriptional defects in only one polymerase system. However, as shown in Figure 3- 3A, all complexes that were capable of DNA binding also functioned well for RNA polymerase II and III transcription with the extent of activity essentially parallel for both systems. Thus, those residues that are not evolutionarily conserved are also not critical for human snRNA gene transcription by either polymerase. In contrast, those mutant complexes that were crippled for DNA binding activity (C354A, C357A, C380A, C3 83A) did not support either RNA polymerase II or III transcription. We consider it likely that these critical cysteine residues within region 2 of the SNAP finger play a structural role for SNAP50 function and are likely important for Zn binding. Some unexpected differences between RNA polymerase II and III transcription were apparent for those mutant complexes that exhibited reduced, but not ablated, DNA binding. Some complexes (H313A, C317A, H319A) were moderately active for RNA polymerase III transcription but did not support RNA polymerase II transcription. Most noticeably, the complex containing SNAP50 H388A was fully functional for RNA polymerase III transcription. Thus, the DNA binding defects caused by altering these conserved residues, 99 Figure 3-3. The SNAP50 zinc finger domain is required for human snRNA gene transcription by RNA polymerases II and III. (A) HeLa cell nuclear extract was depleted with anti-SNAP43 antibodies to immunodeplete endogenous SNAPC. In vitro U1 and U6 transcription was then tested in the absence (lanes 1) or presence of purified SNAPcy4 (5 ng) containing wild type SNAP50 (lane 3) or mutant SNAP50 with the indicated alanine substitutions (lanes 4-19). Addition of GST alone did not reconstitute either U1 or U6 transcription as shown in lane 2. (B) Summary of mutations in the SNAP50 zinc finger domain. Alanine substitution of cysteines and histidines within the SNAP50 zinc finger domain revealed three classes of phenotypes, as indicated. 100 mSNAPcM ssssgTBP>Brf2>deL 103 - mSNAPc-[r4 + mSNAPcfi - - - + - + + + - - - + de1(1-470) - . + - + - + + - - + + 8er -+- -+++- -+++TBP mSNAPc/TBP/Brt2/de1 TBP/Brf2/de1 mSNAPc/TBP/Brf2 mSNAPc/TBP /_ TBP/Brf2 mSNAPc Free Probe -------++++++++mSNAPcy4 - - - + - + + - - - + - + + + de1(1.47o) --+-++---+-+-++Brf2 -+--+++-+--++-+TBP mSNAPchBP/Ber/del ' _ mSNAPc/TBP/Brf2 mSNAPc/TBP mSNAPc Free Probe 12345 6789101112131415 104 to visualize DNA binding for reactions containing SNAPC (right panel), and for exposures that exhibited prominent DNA complexes containing SNAPc, the DNA binding by Brf2-TFIIIB alone was essentially undetectable (not shown, and Figure 3-4B). Thus, at least under these conditions, SNAPC plays a dominant role for TBP recruitment to U6 promoter DNA. Interestingly, serial addition of Brf2 then de1 resulted in complexes that migrated incrementally more slowly, consistent with the idea that increasingly larger complexes are being assembled on the DNA. The amount of the SNAPcy4-DNA complex was not further affected by Brf2 and de1. In contrast, the amount of the SNAPcy4/TBP-DNA complex was diminished by Brf2 addition, and the SNAPcy4/TBP/Brf2-DNA complex was diminished by de1 addition. These last observations suggest a substrate-product relationship during complex assembly, and are consistent with the idea that Brf2 and de1 both exhibit a binding preference for the higher order complexes containing TBP than that complex containing only SNAPC. To further refine the pathway for assembly of SNAPC and Brf2-TFIIIB on U6 promoter DNA, combinations of each factor were tested for DNA binding ability in EMSA. As shown in Figure 3-4B, in the absence of SNAPC, none of the Brf2-TFIIIB components bound to DNA when tested singly or in combination under conditions that support robust DNA binding by SNAPcy4. Interestingly, SNAPcy4 was capable of recruiting TBP, but not Brf2 or de1, when tested in pair wise combination, whereas Brf2 could be incorporated into the complex only in reactions containing SNAPcy4 and TBP. This observation further supports the premise that Brf2 preferentially recognizes the SNAPc/TBP promoter-bound complex. Similarly, deI was not recruited to the ' SNAPc/TBP complex, but again it required the presence of Brf2 for DNA association. 105 Based on these results, we propose a sequential assembly pathway with the initial promoter recognition performed by SNAPC and TBP followed by Brf2 and del. The eventual recruitment of del is predicted to enable RNA polymerase III recruitment under transcription conditions. To test the hypothesis that Brf2-TFIIIB can suppress DNA binding defects in SNAPC, recombinant SNAPC containing wild type or mutant SNAP50 were tested for DNA binding in the absence or presence of each component of Brf2-TFIIIB (Figure 3-5). For this assay, examples from each category of mutant SNAP50 were tested including H388A and H313A that were fully or partially active in U6 transcription, respectively, as well as C383A that was devoid of measurable activity for either DNA binding or transcription. In these assays, the same U6 promoter sequence was used for DNA binding as that contained on the reporter plasmids that were previously used for in vitro U6 transcription. As expected, wild-type recombinant SNAPc bound DNA well and supported robust recruitment of Brf2-TFIIIB. In contrast, mutant SNAPc containing SNAP50 (H388A), SNAP50 (C383A), or SNAP50 (H313A) were each inactive when tested alone for DNA binding. Thus, both wild-type and mutant SNAPc bound with reduced affinity to the U6 promoter sequences tested in this assay relative to those experiments done with the artificial high affinity PSE promoter probes as shown in Figures 3-2 and 3-4. Importantly, DNA binding by the SNAP50 (H388A)-containing complex was restored by TBP, although not to levels seen with the wild type SNAP50 complex, and higher order complex formation with Brf2 and de1 occurred at nearly wild type levels. Thus, Brf2-TFIIIB was capable of restoring DNA binding activity to mutant SNAPC. This result stands in contrast to the SNAP50 (C3 83A)-containing 106 Figure 3-5. TFIIIB suppresses DNA binding defects in SNAPC. DNA binding reactions were carried out as previously described for the artificial AC probe instead using the natural U6 promoter containing DSE, PSE, and TATA-box elements. Reactions were performed with 8 ng of SNAPcy4 containing either wild type SNAP50 (lanes 2-8) or mutant SNAP50 with the substitutions H388A (lanes 9-12), C383A (lanes 13-16), and H313A (lanes 17-20). Reactions additionally containing TBP, TBP and Brf2, or TBP, Brf2 and de1 (1-470) were performed as indicated. DNA binding by mutant SNAPcy4 harboring the H388A mutation was restored by TBP alone whereas TBP plus Brf2 were required to restore DNA binding by SNAPcy4 harboring the SNAP50 (H313A) mutation. No DNA binding under any condition was observed for SNAPcy4 containing the SNAP50 (C3 83A) substitution. 107 Wt H388-A CS83-A H313-A ‘______—___fi mSNAPcy4 - +++++++++++++++++++ de1(1-470)- ---+— ++---+—--+---+ Brl‘2 ---+++-+--++--++--++ TBP --+++----+++-+++-+++ mSNAPc/TBP/BrlZ/deix mS NAPclTBP/Brfz _ mSNAPchB mSNAP Free Probe 1 2 3 4 5 6 7 8 91011121314151617181920 108 complex that was unable to bind DNA under any conditions. Interestingly, TBP did not restore DNA binding by the SNAP50 (H313A)-containing complex but Brf2 plus TBP did, suggesting that the H313A substitution presents a more dramatic defect to DNA binding by SNAPC. Nonetheless, deI could be recruited at reduced levels by this SNAP(:(H3 13A)/T BP/Brf2 complex, consistent with the markedly reduced transcription supported by this complex for in vitro U6 transcription. The C-terminal SNAP50 zinc finger binds zinc. As the previous analyses revealed that certain amino acids within the SNAP finger domain are differentially required for DNA binding and snRNA gene transcription, we postulated that those amino acids that are absolutely critical for both functions are also important for zinc binding. To test this hypothesis, Zn binding studies of wild type and mutant SNAP50 were undertaken. While our initial analysis of zinc content was performed for the four-member complex (SNAPcy4), suitable amounts of mutant complexes for zinc analysis were not obtained in this context. Nor were we able to obtain sufficient amounts of full-length SNAP50 (1- 411) or truncated SNAP50 (301-411) for these studies when expressed individually in E. coli. However, suitable amounts of truncated SNAP50 (315-411) were obtained when expressed as fusion protein with a His- SUMO N-terminal tag, and zinc binding studies of this protein were therefore pursued. As shown in Figure 3-6, analysis of SNAP50 (315-411) by flame atomic absorption showed that this region of SNAP50 bound substantial amounts of zinc (~0.7 mole zinc/mole protein). This level of zinc binding by the isolated SNAP50 zinc finger domain ' is comparable to that seen for full-length SNAP50 (1 -41 1) in the context of the four- 109 Figure 3-6. The SNAP50 zinc finger domain binds zinc. Wild type and mutant SNAP50 (315-411) containing the indicated mutations were tested for Zn content by flame atomic absorption, and the calculated molar ratios of zinc to protein are shown. Error bars indicate the relative standard deviation. 110 Ratio Zn/Protein 1.00 0.80 0.70 0.60 ' 0.50- 0.40 0.30 0.20»- 0.10...- 0.00 53")“ 956‘ 0'56 O'fqu’ 0‘56? efvodgroeh Protein ID. 0 o 111 protein complex SNAPcy4 (~0.9 mole zinc/mole protein), indicating that the SNAP finger domain is likely responsible for zinc binding by SNAPC. Zinc binding by SNAP50 (315- 411) was markedly reduced by individual alanine substitution at positions C354, C357, C380, and C383, whereas alanine substitutions of C377 and H388 did not substantially affect zinc binding. Overall, these data indicate that those cysteines within the C354x2C357x22C3 80x2C3 83 motif are important for zinc binding by SNAPC, whereas the adjacent C377 and H388 residues are not. Moreover, SNAP50 (315-411) harboring either the C354A:C380A or C354A:C383A double alanine substitutions was further incapacitated as compared to SNAP50 C354A or C383A, but zinc levels were comparable to the reduced levels observed for SNAP50 harboring the single C380A substitution. Thus, C380 plays a more critical role in zinc binding than C354 and C383. Discussion DNA binding by SNAPc is a cooperative event wherein SNAP190, SNAP50, and SNAP43 are all required for promoter recognition (ref. (13), and Figure 3-1). Our data demonstrate that an unorthodox zinc finger domain in SNAP50 plays a critical role in this process. A comparison of this region with SNAP50 homologues from other species revealed that the arrangement of many cysteine and histidine residues within the SNAP50 C-terrninal region is remarkably well conserved (Figure 3-2A), and a mutational analysis of all histidine and cysteine amino acids throughout this region showed that these highly conserved residues are critical for DNA binding by SNAPC. The high degree of sequence conservation within the SNAP finger domain ' throughout evolution suggests that SNAP50 function is conserved in other species. 112 Indeed, Drosophila PBPSO (Proximal element binding protein 50 kDa), a homologue of human SNAP50, makes direct DNA contacts within the U6 and U1 promoters although the promoter sequences recognized by human SNAPC and Drosophila PBP are different (34). Besides SNAP50, Drosophila also maintains homologues of SNAP43 and SNAP190, but not of SNAP45 or SNAP19. The identification of SNAPSO- and SNAP43- related proteins in Trypanosoma and Leishmania suggests an ancient fimction for SNAPC in non-translated RNA production, in this case RNA polymerase II transcription of spliced leader (SL) RNA. Trypanosoma SNAPc also contains a Myb domain-containing protein (3, 29) reminiscent of human SNAP190, which contains an unusual Myb DNA binding domain (35). The conservation of these three subunits throughout evolution remarkably parallels the experimental definition of a minimal human SNAPC composed of SNAP190 (1-505), SNAP50, and SNAP43 that retains full activity in RNA polymerase II and III transcription (7, 24). Interestingly, the overall spacing of potential zinc coordination sites within SNAP50 does not resemble other known zinc finger motifs, although the arrangement for a subset of these cysteine residues resembles that of hormone receptor DNA binding domains (20), consistent with a role in DNA binding for this region in SNAP50. Nonetheless, the unusual arrangement of zinc coordination sites combined with secondary structure predictions suggest that human SNAP50 is the founding member for a novel class of zinc finger domains that we refer to as the SNAP finger domain. To date, an experimental structure of SNAP50 has not been determined. Therefore, computational methods were employed to predict model structures to assist our understanding of the mechanism for DNA binding by SNAP50. Comparative 113 modeling, which is often successful in other cases, was not possible because no sequence homologs of SNAP50 with known structures are available. However, as the SNAP finger domain is sufficiently short (~100 residues) ab initio modeling based only on the amino acid sequence and the predicted secondary structure was performed. Although ab initio structure prediction methods in general cannot accurately predict protein structures at the level of experimental structures, it is often possible to obtain approximate models of relatively small domains (<100 residues) with an overall root mean square deviation of S- 10 A from the correct, native structure. The first round of ab initio prediction with the Robetta server resulted in ten models for the SNAP finger domain. The resulting models were substantially different; however, the conserved cysteines 354, 357, 380, and 383 were in sufficiently close proximity to serve as zinc coordination sites in 5 out of the 10 models. Such a result is non-trivial given that the pair C354/C357 is separated by 22 residues from C380/C383, which lends support to the hypothesis that these four cysteines coordinate zinc in a novel zinc finger fold topology. In order to examine a wider range of possible structures and arrive at a model for the entire SNAP finger domain, additional ab initio sampling was carried out under the constraint that the two pairs of residues, C354/C380 and C357/C3 83, are each in close proximity. The best-scoring model consists mainly of 8- sheets and a small a-helical segment according to the predicted secondary structure (Figure 3-7A), and the predicted structure is shown in Figure 3-7B. Submission of the model shown to the DALI server (14, 15) resulted in two known structures with remotely similar topologyzdomain II from calpain, a cysteine protease (PDB ID: IKXR) and a beta-propeller domain of sialidase (PDB ID: lEUT). However, the structural similarity is 114 Figure 3-7. Model for the SN AP50 zinc finger domain. (A) Predicted secondary structure of SNAP50 (301-411) from SABLE (39). (B) Stereo image of the predicted model structure for the C-terminal domain of SNAP50 (residues 305-411). Conserved cysteines C354, C357, C3 80, C383 (yellow) are shown in a zinc coordination geometry (zinc is shown in orange). The conserved residues H313, C317, H319, and H388 are shown in green, and other conserved residues are shown in blue. (C) Electrostatic potential projected onto the molecular surface of the SNAP50 model in the same orientation as in figure 78 (left) and rotated to show the back (right). Modeling of SNAP50 zinc finger domain was done by Dr. Michael Feig. 115 a a. a a a a.a a ooooooouummmmooooooooooommmmmammmoooouoooooooummuoooommmmunmmoooouummuuuuooooouooooumuoummmmmmooooumummoooommmoo zuaean>aaa¢quuequzeamew-qxuuu>nummomanm4umnzze>xa¢aasuu>uuumaxnxuuuHaamwaamoqonnum>umnnaH>H>unnaeouoaqaaueqxuoao cow can can can can can can can can can can 116 117 sufficiently low to suggest that the predicted SNAP finger fold has a novel architecture. The current model for the SNAP finger domain highlights the zinc coordination by C354, C3 57, C380, and C383, and is independently supported by experimental data indicating that these four cysteine residues are involved in zinc binding. The proposed model is vaguely reminiscent of the GATA-l zinc finger motif wherein an or-helix plays a critical role in DNA recognition (33), but is substantially different because of a much longer inserted sequence between the two pairs of cysteines. In the SNAP50 model, other highly conserved residues, G315, P341, and P376, are located at critical turn regions for stabilization of the proposed structure. Interestingly, this SNAP50 model also brings the conserved residues H313, C317, C319 in proximity with H388, which could provide an alternate metal binding site. We note, though, that the binding of an additional metal atom to SNAPC is not supported by the experimental data, and the exact function of these residues remains unclear. These residues may stabilize structures involved in DNA binding by SNAP50, or instead participate directly in DNA contacts. Of note, these residues are located adjacently to the a-helix, which in other zinc finger proteins is frequently used to make specific base contacts within the major groove during DNA binding (20). However, only the C- terminus of the corresponding a-helix within the SNAP finger domain model is fully exposed, which would likely limit major groove contacts. Thus, stable DNA binding through this region may require additional contacts by flanking residues. Interestingly, the electrostatic surface potential for the SNAP finger domain (Figure 3-7C) shows that the presented model clearly distinguishes between positively and negatively charged faces. A large well-defined positively charged surface patch surrounding the a-helix 118 suggests the potential for DNA binding, although a specific mode of interaction between SNAP50 and DNA cannot yet be predicted because of the uncertainty associated with this working model. Nonetheless, the current study provides insight into the DNA binding properties of SNAPC, and identifies the evolutionarily conserved zinc finger domain of SNAP50 as critical for promoter recognition and human snRNA gene transcription. Acknowledgements We thank Craig Hinkley, Heather Hirsch, Liping Gu, Dorothy Tappenden, and Justin Bammer for technical assistance, and Tharakeswari Selvakumar for critical reading of the manuscript. We thank David Szymanski for assistance with the ICP-MS. This research was supported by the NIH Grants R01-GM59805 and R01-GM063894 to R.W.H. and J .H.G., respectively. 119 References 10. Brooks, B., R. Bruccoleri, B. Olafson, D. States, S. Swaminathan, and M. Karplus. 1983. CHARMM: A program for macromolecular energy, minimization, and dynamics calculations. Journal of Computational Chemistry 4:187-217. ' Cabart, P., and S. Murphy. 2002. Assembly of human small nuclear RNA gene- specific transcription factor IlIB complex de novo on and off promoter. J Biol Chem 277:26831-8. Das, A., Q. Zhang, J. B. Palenchar, B. Chatterjee, G. A. Cross, and V. Bellofatto. 2005. Trypanosoma] TBP functions with the multisubunit transcription factor tSNAP to direct spliced-leader RNA gene expression. Mol Cell Biol 25:7314-22. Feig, M., J. Karanicolas, and C. L. Brooks, 3rd. 2004. MMTSB Tool Set: enhanced sampling and multiscale modeling methods for applications in structural biology. J Mol Graph Model 22:377-95. Ford, E., M. Strubin, and N. Hernandez. 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-l. Genes Dev 12:3528-40. Gu, L., W. J. Esselman, and R. W. Henry. 2005. Cooperation between small nuclear RNA-activating protein complex (SNAPC) and TATA-box-binding protein antagonizes protein kinase CK2 inhibition of DNA binding by SNAPC. J Biol Chem 280:27697-704. Hanzlowsky, A., B. Jelencic, G. Jawdekar, C. S. Hinkley, J. H. Geiger, and R. W. Henry. 2006. Co-expression of multiple subunits enables recombinant SNAP(C) assembly and function for transcription by human RNA polymerases II and 111. Protein Expr Purif. Henry, R. W., E. Ford, R. Mital, V. Mittal, and N. Hernandez. 1998. Crossing the line between RNA polymerases: transcription of human snRNA genes by RNA polymerases II and 111. Cold Spring Harb Symp Quant Biol 63:111-20. Henry, R. W., B. Ma, C. L. Sadowski, R. Kobayashi, and N. Hernandez. 1996. Cloning and characterization of SNAP50, a subunit of the snRNA- activating protein complex SNAPc. Embo J 15:7129-36. Henry, R. W., V. Mittal, B. Ma, R. Kobayashi, and N. Hernandez. 1998. SNAP19 mediates the assembly of a functional core promoter complex (SNAPc) shared by RNA polymerases II and III. Genes Dev 12:2664-72. 120 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. Henry, R. W., C. L. Sadowski, R. Kobayashi, and N. Hernandez. 1995. A TBP-TAF complex required for transcription of human snRNA genes by RNA polymerase II and 111. Nature 374:653-6. Hernandez, N. 2001. Small nuclear RNA genes: a model system to study fundamental mechanisms of transcription. J Biol Chem 276:26733-6. Hinkley, C. S., H. A. Hirsch, L. Gu, B. LaMere, and R. W. Henry. 2003. The small nuclear RNA-activating protein 190 Myb DNA binding domain stimulates TATA box-binding protein-TATA box recognition. J Biol Chem 278:18649-57. Holm, L., and C. Sander. 1994. Searching protein structure databases has come of age. Proteins 19:165-73. Holm, L., and C. Sander. 1998. Touring protein fold space with Dali/FSSP. Nucleic Acids Res 26:316-9. Hu, P., K. Samudre, S. Wu, Y. Sun, and N. Hernandez. 2004. CK2 phosphorylation of del executes cell cycle-specific RNA polymerase III transcription repression. Mol Cell 16:81-92. Humphrey, W., A. Dalke, and K. Schulten. 1996. VMD: visual molecular dynamics. J Mol Graph 14:33-8, 27-8. Im, W., D. Beglov, and B. Roux. 1998. Continuum solvation model: Computation of electrostatic forces from numerical solutions to the Poisson- Boltzmann equation. Computer Physics Communications 111:59-75. Kim, D. E., D. Chivian, and D. Baker. 2004. Protein structure prediction and analysis using the Robetta server. Nucleic Acids Res 32:W526-31. Klug, A.,-and J. W. Schwabe. 1995. Protein motifs 5. Zinc fingers. Faseb J 9:597-604. Krishna, S. S., I. Majumdar, and N. V. Grishin. 2003. Structural classification of zinc fingers: survey and summary. Nucleic Acids Res 31:532-50. Ma, B., and N. Hernandez. 2001. A map of protein-protein contacts within the small nuclear RNA-activating protein complex SNAPc. J Biol Chem 276:5027- 35. Ma, B., and N. Hernandez. 2002. Redundant cooperative interactions for assembly of a human U6 transcription initiation complex. Mol Cell Biol 22:8067- 78. Mittal, V., B. Ma, and N. Hernandez. 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 13:1807-21. 121 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. Murphy, S., J. B. Yoon, T. Gerster, and R. G. Roeder. 1992. Oct-1 and Oct-2 potentiate functional interactions of a transcription factor with the proximal sequence element of small nuclear RNA genes. Mol Cell Biol 12:3247-61. Sadowski, C. L., R. W. Henry, R. Kobayashi, and N. Hernandez. 1996. The SNAP45 subunit of the small nuclear RNA (snRNA) activating protein complex is required for RNA polymerase II and III snRN A gene transcription and interacts with the TATA box binding protein. Proc Natl Acad Sci U S A 93:4289-93. Sadowski, C. L., R. W. Henry, S. M. Lobo, and N. Hernandez. 1993. Targeting TBP to a non-TATA box cis-regulatory element: a TBP-containing complex activates transcription from snRN A promoters through the PSE. Genes Dev 7:1535-48. Saxena, A., B. Ma, L. Schramm, and N. Hernandez. 2005. Structure-function analysis of the human TFIIB-related factor II protein reveals an essential role for the C-terminal domain in RNA polymerase III transcription. Mol Cell Biol 25:9406-18. Schimanski, B., T. N. Nguyen, and A. Gunzl. 2005. Characterization of a multisubunit transcription factor complex essential for spliced-leader RNA gene transcription in Trypanosoma brucei. Mol Cell Biol 25:7303-13. Schramm, L., P. S. Pendergrast, Y. Sun, and N. Hernandez. 2000. Different human TFIIIB activities direct RNA polymerase III transcription from TATA- containing and TATA-less promoters. Genes Dev 14:2650-63. Skolnick, J., A. Kolinski, and A. R. Ortiz. 1997. MONSSTER: a method for folding globular proteins with a small number of distance restraints. J Mol Biol 265:217-41. Stumpff-Kane, A. W., and M. Feig. 2006. A correlation-based method for the enhancement of scoring functions on funnel-shaped energy landscapes. Proteins 63:155-64. Trainor, C. D., R. Ghirlando, and M. A. Simpson. 2000. GATA zinc finger interactions modulate DNA binding and transactivation. J Biol Chem 275:28157- 66. Wang, Y., and W. E. Stumph. 1998. Identification and topological arrangement of Drosophila proximal sequence element (PSE)-binding protein subunits that contact the PSEs of U1 and U6 small nuclear RNA genes. Mol Cell Biol 18:1570- 9. Wong, M. W., R. W. Henry, B. Ma, R. Kobayashi, N. Klages, P. Matthias, M. Strubin, and N. Hernandez. 1998. The large subunit of basal transcription factor SNAPc is a Myb domain protein that interacts with Oct-1. Mol Cell Biol 18:368- 77. 122 36. 37. Yoon, J. B., S. Murphy, L. Bai, Z. Wang, and R. G. Roeder. 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 15:2019-27. Zhang, C., S. Liu, Q. Zhu, and Y. Zhou. 2005. A knowledge-based energy function for protein-ligand, protein-protein, and protein-DNA complexes. J Med Chem 48:2325-35. 123 CHAPTER 4 THE HEXIMl REPRESSOR OF P-TEFB STIMULATES TRANSCRIPTION OF ITS 7SK snRNA COREPRESSOR PARTNER BY RNA POLYMERASE III Abstract The HEXIM1/7SK snRNA ribonucleoprotein complex inhibits RNA polymerase II transcription of HIV-1 and cellular genes through interactions with the elongation factor P-TEFb. The 7SK promoter is a type 3 RNA polymerase III promoter, with a core consisting of a TATA box recognized by TBP and a proximal sequence element (PSE) recognized by SNAPC. Here we show that HEXIMI co-purifies and is associated with SNAPC. HEXIMl occupies an endogenous 7SK snRNA gene promoter and stimulates 7SK snRNA gene transcription in vivo. Consistent with a role as an activator of RNA polymerase III transcription, HEXIMl stimulates the binding of SNAPc and TBP to DNA containing their cognate promoter elements. The cyclinTl and cdk9 components of P-TEFb are dispensable for 7SK transcription by RNA polymerase 111, but are required for U1 snRNA gene transcription by RNA polymerase 11. These findings reveal a novel role for HEXIMI as an activator of expression of its 7SK snRNA co-repressor partner and implicate this regulatory circuit as a potential attenuator of global RNA polymerase II transcription. 124 Introduction Small nuclear (sn) RNA genes are among the most actively transcribed genes in human cells with initiation events occurring approximately every 2-4 seconds during periods of active grth (6). While initially believed to be constitutively transcribed and relatively immune to regulatory influence, the transcription of human snRNA genes is regulated both during the cell cycle (19) and in response to DNA damage (10). Many factors that have been extensively investigated because of their regulatory roles during human neoplasm initiation and progression also regulate human snRNA gene transcription. These include the p53 and Retinoblastoma tumor suppressor proteins, as well as the protein kinase CK2 and c-Myc oncoproteins (3, 4, 9, 10, 17, 20). That these critical tumor-related factors control human snRN A gene transcription portends a global role for snRNAs in controlling cellular growth. The human snRN A gene products play central roles in global gene expression, most commonly through their function in RNA processing (26). Examples include the U- rich snRNAs U1, U2, U4, U5, and U6 that are required for messenger RNA splicing. In addition to their classical roles in RNA metabolism, snRNA complexes can also stimulate RNA polymerase II transcription possibly by coupling transcription initiation or elongation to downstream splicing events (8). More recently, U1 snRNA was shown to associate with TFIIH and to stimulate RNA polymerase II transcription (24), perhaps by stimulating TFIIH activity during transcriptional initiation. The cyclin H/cdk7 components of TFIIH phosphorylate the carboxy terminal domain (CTD) of the RNA polymerase 11 largest subunit to stimulate early initiation events (1, 7), but whether U1 snRN A modulates that process is not known. Additional CTD phosphorylation by the 125 cyclin T1/cdk9 kinase component of P-TEFb stimulates RNA polymerase II elongation (29). The P-TEFb transcriptional elongation factor, originally characterized as a RNA polymerase II elongation factor in Drosophila, was subsequently linked to HIV-1 transcription and replication initiated from the HIV long terminal repeat (LTR) (28, 46). Interestingly, P-TEF b kinase activity and consequently RNA polymerase II elongation from the HIV-1 LTR are inhibited by a ribonucleoprotein complex composed of HEXIMI and 7SK snRNA (34, 42). Thus, U1 and 7SK snRNAs associate with the cyclin/cdk complexes, TFIIH and P-TEFb, respectively, but with opposite effects on gene transcription. These observations suggest that the relative levels of U1 and 7SK snRNA in the cell may control cellular transcription potential and furthermore, the regulated transcription of snRN A genes may be important for maintaining this balance. Human snRN A genes can be grouped according to their promoter structures that dictate polymerase specificity for transcription. Some genes such as U1 and U2 are transcribed by RNA polymerase II, and others such as U6 and 7SK are transcribed by RNA polymerase III (11, 15). The use of different polymerases and transcription machinery may allow the cell to discretely control the transcription of distinct classes of snRNA genes. Regardless of polymerase choice, human snRNA genes contain a distal sequence element (DSE) that functions as an enhancer of transcription and contains binding sites for the Oct-1 (32) and STAF (33, 36) activator proteins. Each gene also contains a proximal sequence element (PSE) that is located within the core promoter region that is recognized by the general transcription factor called the snRNA activating protein complex (SNAPC) (35), or the PSE-binding transcription factor (PTF) (32). A TATA box element located at a fixed distance from the PSE determines the initial 126 pathway of preinitiation complex assembly for transcription by RNA polymerase III (25). In this context, the TATA-box is recognized by the TATA-box binding protein (TBP) as part of the snRNA-specific version of the TFIIIB complex (38, 41). The assembly of SNAPC along with TFIIIB pemrits RNA polymerase III recruitment, whereas the absence of the TATA box by default directs PSE- and SNAPc-dependent preinitiation complex assembly with TFIIA, TFIIB, TFIIE, and TFIIF for transcription by RNA polymerase II (23). To identify factors that associate with SNAPC and are potentially involved in human snRN A gene transcription, a biochemical fractionation of SNAPc from HeLa cell extracts was performed. We report that endogenous HEXIMl associates with SNAPC during chromatographic purification and furthermore, HEXIMl associates with human snRNA genes in vivo as detected by chromatin immunoprecipitation (ChIP). In vitro, recombinant HEXIMl stimulates promoter recognition by SNAPC and by TBP to their cognate cis elements within a 7SK-like probe, and in vivo HEXIMl activates 7SK snRNA, thus revealing a novel positive function for HEXIMl in regulating RNA polymerase III transcription. Other components of P-TEFb including cyclin T1 and cdk9 are dispensable for U6 and 7SK snRNA gene transcription by RNA polymerase III in vitro, but are required for U1 transcription by RNA polymerase 11, suggesting that the function of HEXIMl for RNA polymerase III transcription is separate from its role in regulating P-TEFb kinase activity. 127 Methods and Materials UV cross-linking. The conditions for UV cross linking are essentially as described (35), except that reactions were performed with highly purified SNAPc fractions. The purification of SNAPC was described previously (14). Electrophoretic mobility shift assays. EMSA were performed essentially as described (16) using recombinant HEXIMI expressed in E. coli as a N-terrninal GST-fusion protein (GST-HEXIMI). After protein expression, GST-HEXIMI was affinity-purified by binding to glutathione agarose beads followed by thrombin digestion to release the untagged full-length HEXIMI protein. As indicated, reactions also included recombinant full-length human TBP or mini-SNAPC containing SNAP190 (1-505), SNAP43, and SNAP50. The amounts of each protein are indicated in the figure legends. The radiolabeled DNA probes used contained a high affinity wild-type mouse U6 PSE and human U6 TATA box. Additional reactions were performed with probes that contain debilitating mutations in each element. Protein-DNA complexes were separated on a 5% polyacrylamide gel in 0.5X Tris-borate-EDTA (TBE) running buffer at 150 V. The HEXIMl-TBP complexes were resolved in running buffer containing Tris-glycine-EDTA-Mg2+ (TGEM). Chromatin immunoprecipitation assay. Normal human mammary epithelial cells (184B5) were maintained inDulbecco’s minimum essential media (Gibco) containing 5% fetal bovine serum (Gibco), and 128 penicillin-streptomycin at 37°C with 5% C02. ChIP assays were performed as described previously (18) using 184B5 cells that were grown to ~75% density. Cells were treated with 1% formaldehyde for 30 min at room temperature. After cell lysis, the DNA was fragmented into 500-800 bp fragments by sonication. Immunoprecipitation reactions were performed overnight at 4°C using chromatin from approximately 107 cells per reaction and approximately lug of each antibody. The anti-HEXIMI antibody (C8162) was generated in rabbits using a peptide (CSH419) corresponding to the C-terminal 18 amino acids of HEXIMI. The anti-SNAP43 (C848, (14)) and anti-TBP (SL2, (27)) antibodies were described previously. The anti-RNA polymerase III antibody (MIl'70) was raised in rabbits against the C-terminal 18 amino acids of hRPClSS as previously described (40). The RNA polymerase 11 antibody (8WG16) was purchased from Covance Research Products. The anti-cdk9 (SC-484) and anti-cych (SC-8127) antibodies were purchased from Santa Cruz Biotechnology. Immunoprecipitated DNA was analyzed by PCR using primers specific to the genes indicated. The primers used for amplification of each gene are as follows: U6 forward: 5’-AAGACGCGCAGGCAAAACG-3’ U6 reverse: 5’-CGGTGTTTCGTCCTTTC-3’ 7SK forward: 5’-T'ITI‘GGGAATAAATGATATTTG-3 ’ 7SK reverse: 5’- GAGGTACCCAGGCGGCGCACAAG-3’ U1 forward: 5’- CACGAAGGAGTI‘CCCGTG-3’ U1 reverse: 5’-CCCTGCCAGGTAAGTATG-3’ U1 upstream forward: S’-GAACTTACTGGGATCTGG-3’ 129 U1 upstream reverse: 5’-GAGACAACTGAGCCACTTG-3’ GAPDH forward: 5’-AGGTCATCCCTGAGCTGAAC-3’ GAPDH reverse: 5’- GCAATGCCAGCCCCAGCGTC-3’ PCR products were separated by 1.5 % agarose gel electrophoresis in 0.5X TBE buffer, stained with ethidium bromide, and visualized with Kodak imaging software. In vitro transcription. Antibody irnmunodepletion of HeLa cell nuclear extracts were performed as described (13) and the depleted extracts were used for in vitro transcription assays for 1hr at 30°C. In vitro transcription of human U6 snRNA, U1 snRNA, AdML, and AdVAI genes were performed as described (25, 35). 7SK transcription was analyzed by a riboprobe protection assay using the plasmid pBS-7 SK, containing the 7SK promoter from —245 to +1 fused to an inverted B-globin sequence, and conditions identical to those used for U6 in vitro transcription. Transcripts were separated by denaturing PAGE and visualized by autoradiography. Transient transfection assay. For the HEXIMl knockdown experiment, control siRNA and the HEXIMl-specific annealed siRNA (Si-RNA ID# 17952) were purchased from Ambion Biotechnology (5’- GGAUCCGAGCCGAGAUGUU-3’). HeLa cells were grown to 50-60% confluence in a six-well plate and transiently transfected with 0.2 nmol of the siRNA using Lipofectarnine 2000 (Invitrogen). After 8 h, the medium containing the transfection reagent was supplemented with Dulbecco’s modified Eagle’s medium with 5% fetal 130 bovine serum and antibiotics. Cells were also co-transfected with 300 ng of the pBS-7SK reporter plasmid containing an inverted B-globin sequence driven by the 7SK snRNA core promoter (-245 to +1). The pBS-Ul reporter plasmid was constructed by replacing the 7SK promoter region with that of U1. The pU6/I-Iae/Ra.2 has already been described (25). Cells were harvested 30 hr post transfection. For HEXIMl overexpression, HeLa cells were transiently transfected with 10 ng pCGN-HEXIMl plasmid or pCGN empty vector and 300 ng of the pBS-7SK reporter plasmid. Total RNA was extracted using TRIZOL reagent (Invitrogen). Equal amounts of total RNA were further used in an RNase protection assay with a radiolabeled probe specific to the inverted B-globin sequence. Results Endogenous HEXIMl co-purifies with SNAPC, Previous UV cross linking experiments performed with partially-purified SNAPC /PTF, hereafter referred to as SNAPC, revealed that two subunits, SNAP190/PTF01 and SNAP50/PTFB, were in close proximity to the PSE dtu‘ing DNA binding by SNAPC (12, 45). However, a third unidentified factor of 70 kDa apparent molecular weight was also covalently attached to the DNA in a PSE-specific manner (35). As no subunits within SNAPC correspond to this size, this observation suggested that an additional unknown factor could function in human snRN A gene transcription through interactions with the general transcription factor SNAPc and with DNA. Therefore, an extensive biochemical fractionation of SNAPC was employed to identity this associated factor. The purification scheme for endogenous SNAPC (Figure 4-1A) is the same as that previously described 131 (14). Typically, the SNAPC present in the last Mono-S step of fractionation has been purified approximately 104-fold to the point that individual proteins can be isolated and identified (data not shown). To determine whether a similar pattern of protein cross-linking to PSE probes was possible with highly purified SNAPc as was previously described for crude fractions, UV cross linking experiments were performed with fractions obtained after the Mono-S step of fractionation. DNA binding reactions were performed with radioactive wild type (wt) or mutant (mu) PSE probes containing bromodeoxyuridine. Reactions were also performed in the presence of excess unlabeled wt or mu PSE competitor DNA. After DNA binding, reactions were cross linked with UV light, digested with micrococcal nuclease, and the molecular weight of proteins cross linked to DNA were estimated by SDS-PAGE and autoradiography. As shown in Figure 4-1B, three proteins of approximately 70, 54, and 50 kDa were detected in reactions performed with the wt PSE probe either alone (lane 2) or with mu PSE competitor DNA (lane 5). No cross linking was observed in reactions using the mu PSE probe (lane 3) or with wt PSE probe plus wt PSE competitor DNA (lane 4). Thus, all three proteins are specifically bound to DNA in a PSE-dependent fashion. Cross linking of the 70 and 50 kDa proteins in this experiment is similar to that previously described using less purified SNAPc-enriched fractions (3 5). To determine the identity of the cross-linked proteins, the Mono-S fractions were separated by SDS-PAGE and proteins were detected by Coomassie blue staining. Subsequently, those proteins that were of similar molecular weight as the radiolabeled species observed in the UV cross-linking experiment were excised for lysyl endopeptidase digestion and N-terminal peptide sequencing. All peptides that were 132 Figure 4-1. Endogenous HEXIMl associates with SNAPC. (A) Schematic representation of the chromatographic steps used to purify SNAPc. (B) A polypeptide of ~ 70 kDa is cross-linked specifically to the PSE during DNA binding by SNAPC. DNA binding reactions were performed using Mono-S fractions enriched in SNAPC and homogeneously labeled probes that were substituted with bromodeoxyuridine and contained either a wild-type (wt) or mutated (mu) mouse U6 PSE (12). Reactions were performed with the wt or mu probes alone (lanes 2 and 3), or wt probe in the presence of excess wt (lane 4) or mu (lane 5) unlabeled competitor DNA. Proteins were cross-linked to DNA with UV light, digested with DNase I and micrococcal nuclease, and size fractionated by 12.5% SDS-PAGE. Proteins covalently linked to the remnants of the radiolabeled probe DNA were visualized by autoradiography. Lane 1 contains protein size markers. N-terrninal sequencing of the 50, 54, and 70 kDa proteins from untreated Mono-S fractions identified the proteins to be SNAP50 and HEXIMl, as indicated. (C) A minor population of cellular HEXlMl co-purifies with SNAPC. The phosphocellulose P-ll fractions were characterized for the presence of HEXIMI and SNAP43 by Western blot analysis. Increasing amounts of P1 l-A (lanes 1 to 3), P1 l-B (lanes 4 to 6), P11- C (lanes 7 to 9), and P1 l-D (lanes 10 to 12) were separated by SDSPAGE electrophoresis, and the presence of SNAPC and HEXIMl were detected by Western analysis. (D) HEXIMI co-fractionates extensively with SNAPC. Twenty mL of highly purified SNAPC fractions from the Mono-S stage of purification were analyzed by Western analysis using antibodies against HEXIMl (top panel) or SNAP43 (middle panel). Five mL of the same fractions were also analyzed by EMSA for PSE-binding activity (bottom panel) using radioactive probes containing a high-affinity mouse U6 PSE and TATA box. The positions of unbound probe (free probe) and SNAPC bound to DNA are indicated. The peak of SNAPC DNA binding activity is contained in fractions 61 to 66, whereas the peak of HEXIMI is found from fractions 58 to 69. This experiment was performed by R. William Henry. 133 A HeLa M whole cell extract 5 _ - Wt Mu Competitor E Wt Mu Wt Wt Probe 18-24% ammonium sulfate ppt. Phosphocellulose P-11 1 1 I A B C D 974 _ 66 _ — HEXIM1 Cibacron blue ' . _,HEX|M1' . — SNAP50 Mono-Q 45 _ Mono-S 30 _ c _ 1 2 3 4 5 Pn'fil 1311-3. P11-C I P11-BI w w* SNAP43 7 a... - HEXIM1 12345676 9101112 134 Mono-S fractions 0 $3 4: 52 54 56 58 so 62 64 66 68 70 72 74 76 g g §51 53 55 57 59 61 63 65 e7 69 71 77 " i” ‘ “'7 “’ HEXIM1 W -HEX1M1' . W -SNAPc Free probe 1 2 345" 6 7 8 9101112131415161718192021222324252627282930 135 obtained from the 70 and 54 kDa proteins correspond to HEXIM1 and peptides from the 50 kDa protein were found to belong to SNAP50, previously identified as contacting the PSE during DNA binding by SNAPC (12). For all three cases, no peptides from other proteins were obtained, including the recently identified HEXIM2 protein that shares extensive similarity with HEXIM1 (2, 44). We speculate that the smaller HEXIM1 protein (labeled HEXIM1*) is a proteolytic product of larger full length HEXIM1. Thus, HEXIM1 present in the SNAPc-enriched fractions is in close proximity to the PSE during DNA binding by SNAPC. Next, antibodies were generated against HEXIM1 to follow the fi‘actionation of HEXIM1 during SNAPc purification. As shown in Figure 4-1C, most SNAPc is detected in the P1 l-C fraction (lanes 7-9), as expected, whereas most HEXIM1 is present in the P1 1- B (lanes 4-6) and P1 l-D (lanes 10-12) fractions, with a similar but lesser amount detected in the P1 l-C fraction containing SNAPC. Neither SNAPC nor HEXIM1 were detected in the P1 l-A fraction (lanes 1-3). Therefore, a substantial but minor proportion of HEXIM1 co-fractionates with SNAPC during phosphocellulose chromatography. The population of HEXIM1 that does co-purify with SNAPc was more extensively analyzed as shown in Figure 1D. Fractions obtained from the Mono-S step of fractionation revealed an extensive co-purification of HEXIM1 (top panel) and SNAP43 (middle panel). The fractionation pattern for SNAP43 closely resembled the pattern of SNAPC DNA binding activity in electrophoretic mobility shift assay (EMSA; bottom panel). However, significant amounts of HEXIM1 were also detected in fractions that were devoid of SNAP43 and SNAPC DNA binding activity, indicating that even at this late stage of purification, HEXIM1 can be chromatographically separated from SNAPc. Only 136 modest amounts of HEXIM1 associated with SNAPc during co-irnmunoprecipitation from HeLa nuclear extracts or SNAPc-enriched fractions, and the SNAPc/DNA complex was not obviously super shifted using anti-HEXIM1 antibodies (data not shown), suggesting that HEXIM1 is not a stable component of endogenous SNAPc. Together, these data indicate that HEXIM1 and SNAPc may only loosely associate. HEXIM1 cooperates with SNAPC and TBP for snRNA gene promoter recognition The observation that HEXIM1 was cross linked to DNA in a PSE-specific fashion indicated that HEXIM1 could target these promoters through cooperative DNA binding with SNAPC or TBP, which also function for both RNA polymerase II and III transcription of snRN A genes. Therefore, EMSAs were performed to investigate whether HEXIM1 could cooperate with SNAPC and TBP for promoter recognition. In the following experiments, low levels of each factor were purposefully chosen to minimize DNA binding by each factor alone. As shown in Figure 4-2A, neither HEXIM1 nor recombinant mini-SNAPC alone bound strongly to thSE/thATA probe DNA (lanes 2 and 3, respectively), but inclusion of both factors resulted in cooperative complex formation (lane 4) that was not observed in reactions containing SNAPC plus comparable amounts of GST (lane 5). A similar pattern of cooperation between HEXIM1 and SNAPc continued with the thSE/muTATA box probe (lanes 11-15), but complex formation was not observed in reactions performed with probes containing mutations in the PSE, regardless of the TATA box status (lanes 6-10 and 16-20). The HEXIM1-SNAPC complex was super shifted by anti-HEXIM1 and anti-SNAP190 antibodies, but not by IgG (data not shown), indicating that both factors are components of this complex. 137 Figure 4-2. HEXIM1 cooperates with SNAPC and TBP for DNA binding. (A) HEXIM1 cooperates with SNAPC for DNA binding in a PSE-specific manner. Approximately 30 ng of mini-SNAPC (mSNAPc) containing SNAP190 (1-505), SNAP43, and SNAP50, was used for DNA binding either alone or with 100 ng of HEXIM1 or GST. Reactions containing only HEXIM1 were also performed as indicated. Reactions were performed with dsDNA probes containing combinations of wt and mu PSE with wt and mu TATA as shown schematically. Reactions containing only the dsDNA probes are shown in lanes 1, 6, 11, and 16. (B) HEXIM1 cooperates with TBP for DNA binding in a TATA box specific manner. Approximately 100 ng of TBP was tested for DNA binding either alone, or with 100ng of HEXIM1 or GST, as indicated. Reactions containing HEXIM1 alone were also performed, as indicated. The identity of each probe is shown schematically above the figure. (C) HEXIM1 stimulates preinitiation complex assembly on snRNA promoters. HEXIM1 stimulates SNAPC and TBP binding. Similar amounts of SNAPC, HEXIM1 and TBP as above were used and tested for DNA binding on probes containing combnations of wt or mu PSE with wt or mu TATA box. Reactions containing SNAPC and TBP are shown in lane 2. The presumptive identities of the various protein-DNA complexes are shown on the left. 138 WWW I].---+l----+----+----+GST ++---++---++---++- HEXIM1 -++-+-++-+-++-+-++mSNAPc Free Probe 12 3 4 5 6 7 8 91011121314151617181920 WWW ----+---- +----+'----+GST --++---++---++---++-HEX|M1 -+-++-+-++-+-++-+-++'l['BP 139 ESE-113E}- WWW fiflF—‘I l—_'—"'1 - 'A‘PA'i'A‘F'A'i'HEXIM‘l -+++-++-++-++-T3p ++++-++-++-++-mSNAPc SNAPC+TBP+HEXIM1 SNAPc+TBP TBP+HEXIM1 ’ w SNAPC+HEXIM}_, ,5 SNAPc Free Probe 1234567891011121314 140 Therefore, HEXIM1 and mini-SNAPC can cooperate for DNA binding in a PSE- dependent fashion. This result can also explain the PSE dependent cross linking of HEXIM1 to DNA previously observed in the UV cross linking experiments with highly purified SNAPc fractions (Figure 4-1). We next tested whether HEXIM1 can influence DNA binding by TBP (Figure 4- 28). Neither HEXIM1 nor TBP alone bound to the thSE/thATA box (lanes 2 and 3, respectively), but 000perative DNA binding was observed in reactions containing both factors (lane 4), and in this experiment, GST did not influence TBP binding (lane 5). In contrast to the PSE-dependent cooperation between HEXIM1 and SNAPc, the cooperative DNA binding by HEXIM1 and TBP was TATA box dependent, as PSE mutation did not affect complex formation (lanes 6-10), whereas no cooperative complex formation was observed in reactions where the probes contained mutations in the TATA box (lanes 11-20). Thus, HEXIM1 can cooperate with either SNAPC or TBP, and the ability of HEXIM1 to form complexes on DNA is dictated by the DNA binding specificity of its cooperating partner. TBP recruitment by SNAPc is thought to be part of the preinitiation complex assembly pathway. We therefore tested whether HEXIM1 can influence TBP recruitment by SNAPC. As shown in Figure 4-2C, inclusion of HEXIM1 in reactions that contain SNAPc and TBP caused a noticeable retardation in mobility of the SNAPc-TBP-DNA complex, consistent with the idea that higher order complexes are being formed (compare lanes 3 and 4 with lane 2). Interestingly, this effect was not observed on probes that contained muPSE/thATA, thSE/muTATA or muPSE/muTATA sequences. Only the HEXIM1-TBP complex was seen with the probe containing a muPSE/thATA box, 141 (lanes 6 and 7) while the HEXIM1-SNAPC complex was seen with the probe containing thSE/muTATA box (lane 9 and 10). It should be noted that SNAPc and TBP were both detected in the HEXIM1-SNAPc-TBP-DNA complex, but not HEXIM1, using antibody supershilt assays (data not shown). It is possible that HEXIM1 only transiently associates with the DNA and therefore was not detected in the HEXIM1-SNAPc-TBP-DNA complex. Nonetheless, together these observations suggest that HEXIM1 stimulates DNA binding by SNAPC and TBP. Differential snRNA promoter occupancy in vivo by P-TEFb subunits. The association between HEXIM1 and SNAPc observed during biochemical purification of SNAPC suggests that HEXIM1 may play a role in human snRNA gene transcription. Therefore, chromatin immunoprecipitation experiments were performed to determine whether endogenous HEXIM1 associates with various snRN A gene promoters, including the 7SK and U6 snRNA genes that are transcribed by RNA polymerase III, and the U1 snRNA gene that is transcribed by RNA polymerase II. In addition to HEXIM1, immunoprecipitation reactions were performed using antibodies directed against SNAPC, TBP, and RNA polymerases II and 111. As HEXIM1 is a component of P-TEFb, snRNA gene promoter association by the cyclinTl and cdk9 subunits of P-TEFb subunits were examined (Figure 4-3). As expected, the SNAPC and TBP immunoprecipitated samples were enriched for all snRNA gene promoters examined (lanes 6 and 7), relative to the IgG control (lane 5), whereas RNA polymerase III was present only at the U6 and 7SK genes (lane12), and RNA polymerase 11 only at the U1 snRNA gene promoter (lane 11). Interestingly, 7SK promoter DNA was enriched in all P-TEF b specific 142 Figure 4-3. HEXIM1 occupies endogenous snRNA promoters. Chromatin was harvested from human mammary epithelial cells (18435) as described previously (18) and immunoprecipitation reactions were performed using IgG (lane 5) or anti-SNAP43 (lane 6), anti-TBP (lane 7), anti-HEXIM1 (lane 8), anti-Cdk9 (lane 9), anti- Cyclin T1 (lane 10), anti -RNAP 11 (lane 11), and anti-RNAP III (lane 12) antibodies. Immunoprecipitated DNA was analyzed by PCR for enrichment of 7SK snRNA, U6 snRNA, and U1 snRNA core promoters using primers specific for each gene. Enrichment of GAPDH exon 2 and U1 upstream DNA were examined as negative controls. Amplification for a lO-fold serial dilution (10% to 0.01%) of input chromatin is shown in lanes 1-4. 143 a-RNA Polymerase ll a—RNA Polymerase lll ,— v- x _ % INPUT 2 95 1.1.1 ‘6 2. ‘_ 0 "9 *7 3F ‘9 9’ 2’ d 8 C5 5 t5 - u m ~ in.) all. ~““:~.‘) - 1. 7SKSI‘1RNA 397.15 A” 1165'”: tibia: Ni... .44. - - an. - ~ on: m ~ U1 SHRNA it...» rem.- M all- ill- -- , Ul upstream 3 4 5 6 7 8 9101112 144 immunoprecipitations using HEXIM1 (lane 8), cdk9 (lane 9), and cyclin T1 (lane 10) antibodies, whereas U6 promoter DNA was only enriched in the HEXIM1-specific reactions. U1 promoter recovery by P-TEFb antibodies resembled the U6 pattern, with strong recovery by HEXIM1 antibodies. The modest U1 promoter enrichment by cyclin T1 immunoprecipitation suggests that the cyclin/cdk component of P-TEFb could regulate U1 transcription by RNA polymerase II. In all immunoprecipitations, enrichment of the U1 upstream region and GAPDH exon 1 was not observed. These results indicate that multiple components of endogenous P-TEF b associate with the 7SK gene promoter in vivo and thus P-TEF b might regulate 7SK transcription by RNA polymerase 111. Furthermore, 7SK, U6, and U1 transcription may be differentially regulated by P-TEFb even though the 7SK and U6 promoters are highly similar and all genes share a requirement for the PSE, and thus SNAPC, for efficient transcription. The cyclin/cdk component of P-TEFb is not required for RNA polymerase III transcription. Whereas RNA polymerase II elongation is regulated by P-TEFb, which phosphorylates the CTD of the RNA polymerase 11 largest subunit, a direct role for the cyclin/cdk sub-complex of PTEF b in snRNA gene transcription by RNA polymerase II or III has not yet been established. RNA polymerase 111 does not contain a similar proven target for P-TEFb phosphorylation and thus it might be immune to P-TEFb action. Nonetheless, P-TEFb could participate in regulation of snRNA gene transcription by RNA polymerase 111 through an unanticipated mechanism. 145 To test whether P-TEFb is required for snRNA gene expression, P-TEFb was removed from HeLa nuclear extracts by irnmunodepletion using antibodies directed against individual P-TEFb subunits, and the effect on in vitro transcription was determined. As shown in Figure 4-4A, Western blot analysis revealed that treatment of extracts with antibodies against cdk9 (lanes 4-6) and cyclin T1 (lanes 7-9) resulted in an approximate 90% reduction of both cdk9 (top panel) and cyclin T1 (middle panel) relative to the IgG treated extracts (lanes 10-12) and untreated nuclear extracts (lanes 1- 3). Endogenous HEXIM1 levels were reduced by approximately 50% in the P-TEFb- depleted samples. Thus, multiple components of P-TEFb are effectively removed by either treatment. This result is also consistent with the idea that not all HEXIM1 is associated with the catalytic P-TEFb subcomplex. As shown in Figure 4-4B, U1 transcription by RNA polymerase II was markedly inhibited for both the cdk9 (lane 2) and cyclin T1 (lane 3) depleted extracts relative to transcription levels supported by the IgG depleted (lane 4) or mock treated extracts (lane 5). Under these conditions adenovirus major late (AdML) transcription was modestly affected only in the cdk9 immunodepleted extracts (data not shown). Thus, P-TEFb is required for U1 in vitro transcription by RNA polymerase 11, possibly for CTD phosphorylation, and is consistent with a previously postulated role for P-TEFb in 3’ end formation of U2 snRNA (21, 30). In contrast with RNA polymerase II snRNA gene transcription, neither 7SK nor U6 snRN A gene transcription was substantially affected by P-TEFb depletion, indicating that P-TEFb is not essential for in vitro snRNA gene transcription by RNA polymerase 111. Even though cdk9 and cyclin T1 associate with the 7SK snRNA gene promoter in vivo, the apparent bystander status for cyclin T1 and cdk9 during RNA polymerase III 146 Figure 4-4. P-TEFb is differentially used for snRNA gene transcription by RNA polymerases II and III in vitro. (A) Multiple P-TEFb subunits are removed from extracts by antibody irnmunodepletion. HeLa cell nuclear extracts were subjected to immunodepletion using anti-Cdk9 (lanes 4- 6), anti-Cyclin T1 (lanes 7-9), and IgG (lanes 10-12) antibodies that were covalently cross-linked to protein G agarose beads. Lanes 1-3 contain a titration of the untreated extract. A portion of the extracts was separated by 12.5% SDS-PAGE, and analyzed by Western blot analysis using antibodies directed against cdk9, cyclin T1, and HEXIM1, as indicated. (B) P-TEFb is required for U1 transcription by RNA polymerase II, but not for 7SK and U6 snRNA transcription by RNA polymerase III. ln-vitro transcription assays for U1, U6, and 7SK snRNA genes were performed with HeLa nuclear extract (lane 1) or extract that had been immunodepleted for P-TEFb components (lanes 2 and 3). Lanes 4 and 5 show transcription from extract that was mock depleted with IgG or beads, respectively. 147 Input or-Cdk9 or-CycT1 IgG 123456789101112 4;" i; a 431:." 511533 B 117:1 1 011.1 for l. a-Cdk9 N.E. or-Cyclin T1 N.E. Beads alone N.E. IgG N.E. 7322' RNAP ll RNAP III 148 transcription suggests that the 7SK gene might be a site for assembling the inactive P- TEFb ribonucleoprotein complex, perhaps as the 7SK snRNA is being transcribed. HEXIM1 activates 7SK transcription in vivo Next the effect of HEXIM1 over-expression on 7SK reporter gene expression was tested (Figure 4-SA). Over-expression of HEXIM1 resulted in a modest, but reproducible, increase in 7SK transcription (lane 4) relative to that seen in cells transfected with the 7SK reporter plasmid either alone (lane 2) or with an empty expression vector (lane 3). In this experiment, over-expression of HEXIM1 did not affect endogenous actin or rRNA levels. We note that HEXIM1 over-expression did not significantly affect 7SK transcription during nuclear run-on assays in HeLa cells (data not shown), suggesting that in vivo HEXIM1 levels are not limiting for 7SK transcription unless the 78K gene copy number is increased such as during transient transfection. We were unable to achieve a substantial reduction in endogenous HEXIM1 levels by anti-HEXIM1 irnmunodepletion (data not shown), and thus could not determine whether HEXIM1 contributes to snRN A gene transcription in vitro. Nonetheless, the observation that HEXIM1 is present at snRNA promoters in vivo suggests that this protein could directly regulate transcription. To examine the possibility that HEXIM1 regulates RNA polymerase III transcription of snRNA genes, the steady state levels of HEXIM1 were reduced by using siRNA specific for HEXIM1 and the effect on 7SK reporter gene expression was tested. As 7SK snRNA binds directly to HEXIM1, any change in 7SK snRNA levels may confound the assessment of HEXIM1 function, and therefore the 7SK snRNA encoding sequence in this reporter gene was replaced with an inverted B-globin sequence. As shown in Figure 149 Figure 4-5. HEIMl positively regulates 7SK snRNA transcription in vivo. (A) Over-expression of HEXIM1 increases 7SK snRNA transcription. HeLa cells were cotransfected with pBS-7SK reporter gene alone (lane 2) or with either pCGN (lane 3) or pCGN-HA-HEXIMI (lane 4). Cells were harvested 24 hr after transfection and processed for whole cells extract preparation and total RNA collection. Endogenous HEXIM1 and actin levels were measured by Western blot analysis. Endogenous 18S and 28S rRNA levels were monitored by agarose gel electrophoresis and ethidium bromide staining. 7SK snRNA reporter gene transcription was measured by RNase T1 protection assay and phosphoimager analysis. The experiment was performed five times and the average 7SK reporter gene activity is shown in the graph at the bottom. Statistical significance was estimated using a Student’s T-test and error bars are the standard deviation. (B) siRNA mediated reduction in HEXIM1 does not affect 7SK transcription. Hela cells were cotransfected with an inverted B-globin reporter construct driven by a human 7SK snRNA promoter (pBS-7SK) alone (lane 1) or along with either the control siRNA (lane 2) or siHisl (lane 3). 30 hr after transfection cells were harvested and processed for whole cell extract preparation and total RNA collection as in (A). This experiment was performed three times. 150 pCGN-HEXIM1’ - ' + pCGN I I + 7SK-q - + + + “ 1 HEX 'M' 7SK transcription Arbrtl’ary unlts 4 n=3.” “0'01 151 I I I + Si-His1 I I + I si-ctrl E] globin-reporter 1 + + + . - HEXIM1 ' ‘ ' '3'"=1".-:1‘~.>-1A‘:-;.:«‘.:. 4‘ br . \ ‘ ’ - ‘ .m’ - . . .. ~ ,-.-.‘- ‘ - gs."- -' ‘- r .o, 5:5,: =52.)- / ' ‘- "-"q ,"fi‘ . fiztvfl « . .. ,". 5,)1' ' :- n , ‘. " 1\1"~'I:-“> Iu- " . . _ 7SK- reporter . - . "'2 I . . < \. J ‘ ' - . .111 A ; / z \7 ‘ . _.. ‘ ‘ _. -.. . . . . ”.1 .U.“ 1 . 4 it ‘1‘ K. _‘ > .4 "f. (,5 “(.75 ‘4,- . 1:17 .' . .' ; v ~ 3. .' .11 ', .‘ ‘ . ‘ . ; . », .. 1 ~ ’ . ~ I . 7 ' ‘ x ‘ .1 'i'. A" ‘1 ‘ i. - 4 , a. ‘ ’ - o 1 ‘ .2 ' ' I‘- . \ I . 3 ‘. l '-'.','."\ v ‘- ' .' a- . . «a '~: PM, It, ~17 w / 7 z I a A. a s , ‘1 152 4-SB, transient transfection of the si-HEXIMlcaused 90% reduction of endogenous HEXIM1 (lane 4) relative to levels observed during transfection of the control siRN A (si- ctrl lane 3). Actin levels were similar for each treatment as were the steady state 188 and 28S rRNA levels. Interestingly, 7SK reporter gene transcription by RNA polymerase III was not affected with HEXIM1 knock down, suggesting that endogenous HEXIM1 is not essential for transcription. Interestingly, a lower migrating form of HEXIM1 (labeled as HEXIM1 *) was not affected in this knockdown. The sequence of this lower migrating form of HEXIM1 is not known and whether it plays a role in snRNA transcription is yet to be determined. Nonetheless, the over-expression and knockdown data suggests that HEXIM1 stimulates 7SK snRNA transcription although HEXIM1 is not essential for this process. Discussion As the products of human snRNA genes play critical roles in numerous steps of productive global gene expression, the rate of cellular grth and the ability to proliferate may be sensitive to steady state snRNA levels. In particular, global RNA polymerase II transcription may be sensitive to the cellular levels of U1 and 7SK snRNA that associate with the cyclin/cdk complexes, TFIIH and P-TEFb, respectively (24, 34, 42). Both of these complexes directly regulate RNA polymerase II transcription. Interestingly, HEXIM1 forms a cyclin/cdk inhibitor complex with 7SK snRNA to down regulate P- TEFb activity (31, 43). In this context, HEXIM1 is thought to play a negative role in RNA polymerase II transcription. The data presented herein reveal an unexpected 153 positive role for HEXIM1 in transcription of its 7SK snRNA co-repressor partner by RNA polymerase III. The role of HEXIM1 in snRNA gene transcription was first suggested because HEXIM1 associated with the general transcription factor SNAPc during chromatographic fractionation of HeLa cell extracts. As SNAPC is required for transcription of human snRNA genes including U1, U6, and 7SK, a role for HEXIM1 in transcription of all these genes was postulated. Whereas our results suggest a positive role for HEXIM1 in 7SK snRNA gene regulation, however, HEXIM1 function does not seem to be essential for snRN A gene transcription, even though endogenous HEXIM1 was resident at these gene promoters in vivo. While efficient HEXIM1 knockdown was observed, a shorter form of HEXIM1 (HEXIM1‘) was not affected. Interestingly, HEXIMI" and not HEXIM1 associates with the DNA-binding domain within SNAP50 (22), when this region of GST- SNAPSO was used as a bait to pull out interacting proteins from HeLa cell nuclear extract (data not shown). The sequence of HEXIM1 * is not known, but it seems likely that it may have internally deleted sequences, as both N- and C-terminal specific antibodies were able to recognize HEXIM1 * in a Western analysis (data not show). Thus it is likely that an unidentified splice variant of HEXIM1 exists in the cell and may play a role in the regulation of snRN A gene transcription. U6 transcription has been reconstituted in vitro with recombinant factors and highly purified RNAP III, and in this system no requirement for HEXIM1 was observed (5). Perhaps traces of HEXIM1 co-purify with RNAP III or with SNAPC purified from insect cells. It seems more likely; however, that the role of HEXIM1 can be bypassed in the in vitro transcription system, For example, if HEXIM1 serves to facilitate 154 preinitiation complex assembly, this function might be dispensable in vitro where the template is naked DNA and the transcription initiation factors are in excess (20), and thus HEXIM1 activity for U6 regulation may be restricted to a context that is not yet appreciated. Our data further indicate that only a minor proportion of HEXIM1 co-fractionates with SNAPC, suggesting that HEXIM1 partitions into multiple complexes consistent with the idea that HEXIM1 plays multiple independent roles in the cell (43). Indeed, the function of HEXIM1 for RNA polymerase III transcription is independent of P-TEFb kinase activity, as 7SK in vitro transcription is not sensitive to cyclinTl and cdk9 levels. Thus, it is interesting that cyclinTl and cdk9 were both found associated with the endogenous 7SK snRNA gene promoter. One possibility is that P-TEF b kinase activity is required for RNA polymerase III transcription in the cell, but this requirement is not revealed by in vitro assays. However, the catalytic subunits of P-TEFb were not detected at the highly related U6 snRN A gene promoter, and although the role of HEXIM1 for U6 transcription by RNA polymerase III is not known, this observation suggests that RNA polymerase III phosphorylation by P-TEFb is not essential for RNA polymerase III transcription. An alternative explanation is that the 78K snRN A gene serves as a site for P-TEFb assembly of the cyclinT/cdk9 kinase complex with the regulatory HEXIM1/7 SK snRNA complex, as the 7SK snRN A is being transcribed. As endogenous HEXIM1 associates with PSE-containing promoter sequences within the cell an important question remains as to how HEXIM1 is targeted to these genes. We observed that at protein concentrations where little DNA binding is detected by either SNAPC or HEXIM1 alone, HEXIM1 cooperates with SNAPC for PSE- 155 dependent promoter recognition. Furthermore, HEXIM1 also cooperated with TBP for TATA-box dependent binding, suggesting that HEXIM1 plays a more general role to assert its influence on 7SK snRNA gene transcription by facilitating cooperative preinitiation complex assembly at snRN A gene promoters. Human 7SK snRNA, along with other snRNAs, are expressed at high levels in the cell. In part, these levels are maintained by exceptional transcription efficiencies dictated by the typical promoter structure of snRNA genes (3 7). In fact, the relatively compact and powerful promoters of some snRN A genes have lent themselves to widespread use in biotechnology and medical applications to drive high-level expression of effector RNA molecules. However, snRN A gene promoters are sensitive to complex regulatory control that may have downstream effects on global RNA production and cellular proliferation. It is especially intriguing that 7SK snRNA gene transcription is regulated by its functional partner HEXIM1. We speculate that P-TEFb stimulates HEXIM1 production by RNA polymerase II, and in turn, HEXIM1 stimulates 7SK snRNA gene transcription in a process that is expected to down regulate P-TEFb activity as increased levels of the HEXIM1/7SK snRNA complex are formed. Thus, one possibility is that HEXIM1 levels may function as a barometer for cellular gene expression levels via feedback regulation of its co-repressor partner. It is further possible that HEXIM1 contributes to cell growth control in specialized contexts. For example, an intriguing idea is that HEXIM1 antagonizes HIV-1 transcription by directly maintaining P-TEFb in an inactive state and ensuring that the levels of its 7SK snRNA co-repressor partner are adequate for this function. Indeed, a recent model was proposed wherein the HIV-1 Tat protein competes with HEXIM1 for binding to the cyclin T1 component of P-TEFb to increase levels of 156 active P-TEF b and HIV-1 gene transcription (39). It will be important to determine the context, timing, and cell-type specificity for HEXIM1 regulation of 7SK transcription. 157 References 10. ll. Akoulitchev, S., T. P. Makela, R. A. Weinberg, and D. Reinberg. 1995. Requirement for TFIIH kinase activity in transcription by RNA polymerase 11. Nature 377:557-60. Byers, S. A., J. P. Price, J. J. Cooper, Q. Li, and D. H. Price. 2005. HEXIM2, a HEXIM1-related protein, regulates positive transcription elongation factor b through association with 7SK. J Biol Chem 280: 16360-7. Cairns, C. A., and R. J. White. 1998. p53 is a general repressor of RNA polymerase III transcription. Embo J 17:3112-23. Chesnokov, I., W. M. Chu, M. R. Botchan, and C. W. Schmid. 1996. p53 inhibits RNA polymerase III-directed transcription in a promoter-dependent manner. Mol Cell Biol 16:7084-8. Chang, S. S., P. Hu, and N. Hernandez. 2001. Reconstitution of transcription from the human U6 small nuclear RNA promoter with eight recombinant polypeptides and a partially purified RNA polymerase III complex. J Biol Chem 276:20727-34. Dahlberg, J. E., and E. Lund. 1998. The genes and transcription of the major small nuclear RNAs., p. 38-70. In M. Birnstiel (ed.), Structure and function of major and minor small nuclear ribonucleoprotein particles. Springer Verlag, Berlin. Dvir, A., K. P. Garrett, C. Chalut, J. M. Egly, J. W. Conaway, and R. C. Conaway. 1996. A role for ATP and TFIIH in activation of the RNA polymerase II preinitiation complex prior to transcription initiation. J Biol Chem 271:7245-8. Fong, Y. W., and Q. Zhou. 2001. Stimulatory effect of splicing factors on transcriptional elongation. Nature 414: 929- 33. Gomez-Roman, N., C. Grandori, R. N. Eisenman, and R. J. White. 2003. Direct activation of RNA polymerase III transcription by c-Myc. Nature 421:290— 4. Gridasova, A. A., and R. W. Henry. 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 25:3247-60. Henry, R. W., E. Ford, R. Mital, V. Mittal, and N. Hernandez. 1998. Crossing the line between RNA polymerases: transcription of human snRNA genes by RNA polymerases II and 111. Cold Spring Harb Symp Quant Biol 63:111-20. 158 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. Henry, R. W., B. Ma, C. L. Sadowski, R. Kobayashi, and N. Hernandez. 1996. Cloning and characterization of SNAP50, a subunit of the snRNA- activating protein complex SNAPc. Embo J 15:7129-36. Henry, R. W., V. Mittal, B. Ma, R. Kobayashi, and N. Hernandez. 1998. SNAP19 mediates the assembly of a functional core promoter complex (SNAPc) shared by RNA polymerases II and III. Genes Dev 12:2664-72. Henry, R. W., C. L. Sadowski, R. Kobayashi, and N. Hernandez. 1995. A TBP-TAF complex required for transcription of human snRN A genes by RNA polymerase II and 111. Nature 374:653-6. Hernandez, N. 1992. Transcription of vertebrate snRNA genes and related genes, p. 281-313, Transcriptional Reguation., In S. McKnight and K. Yamamoto ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor. Hinkley, C. S., H. A. Hirsch, L. Gu, B. LaMere, and R. W. Henry. 2003. The small nuclear RNA-activating protein 190 Myb DNA binding domain stimulates TATA box-binding protein-TATA box recognition. J Biol Chem 278:18649-57. Hirsch, H. A., L. Gu, and R. W. Henry. 2000. The retinoblastoma tumor suppressor protein targets distinct general transcription factors to regulate RNA polymerase III gene expression. Mol Cell Biol 20:9182-91. Hirsch, H. A., G. W. Jawdekar, K. A. Lee, L. Gu, and R. W. Henry. 2004. Distinct mechanisms for repression of RNA polymerase III transcription by the retinoblastoma tumor suppressor protein. Mol Cell Biol 24:5989-99. Hu, P., K. Samudre, S. Wu, Y. Sun, and N. Hernandez. 2004. CK2 phosphorylation of del executes cell cycle-specific RNA polymerase III transcription repression. Mol Cell 16:81-92. Hu, P., S. Wu, and N. Hernandez. 2003. A minimal RNA polymerase III transcription system from human cells reveals positive and negative regulatory roles for CK2. Mol Cell 12:699-709. Jacobs, E. Y., I. Ogiwara, and A. M. Weiner. 2004. Role of the C-terminal domain of RNA polymerase II in U2 snRNA transcription and 3' processing. Mol Cell Biol 24:846-855. Jawdekar, G. W., A. Hanzlowsky, S. L. Hovde, B. Jelencic, M. Feig, J. H. Geiger, and R. W. Henry. 2006. The unorthodox SNAP50 zinc finger domain contributes to co-operative promoter recognition by human SNAPc. J Biol Chem. Kuhlman, T. C., H. Cho, D. Reinberg, and N. Hernandez. 1999. The general transcription factors IIA, IIB, IIF, and [IE are required for RNA polymerase II transcription from the human U1 small nuclear RNA promoter. Mol Cell Biol 19:2130-41. 159 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. Kwek, K. Y., S. Murphy, A. Furger, B. Thomas, W. O'Gorman, H. Kimura, N. J. Proudfoot, and A. Akoulitchev. 2002. U1 snRNA associates with TFIIH and regulates transcriptional initiation. Nat Struct Biol 9:800-5. Lobo, S. M., and N. Hernandez. 1989. A 7 bp mutation converts a human RNA polymerase II snRNA promoter into an RNA polymerase III promoter. Cell 58:55-67. Lobo, S. M., and N. T. Hernandez. 1994. Transcription of snRNA genes by RNA polymerases II and III., p. 127-159. In R. C. a. C. Conaway, J.W. (ed.), Transcription: Mechanisms and Regulation. Raven Press, Ltd., New York. Lobo-Ruppert, S., V. McCulloch, M. Meyer, C. Bautista, M. Falkowski, H. G. Stunnenberg, and N. Hernandez. 1996. Monoclonal antibodies directed against the amino-terminal domain of human TBP cross-react with TBP from other species. Hybridoma 15:55-68. Mancebo, H. S., G. Lee, J. Flygare, J. Tomassini, P. Luu, Y. Zhu, J. Peng, C. Blau, D. Hazuda, D. Price, and O. Flores. 1997. P-TEFb kinase is required for HIV Tat transcriptional activation in vivo and in vitro. Genes Dev 11:2633-44. Marshall, N. F., J. Peng, Z. Xie, and D. H. Price. 1996. Control of RNA polymerase II elongation potential by a novel carboxyl-terminal domain kinase. J Biol Chem 271:27176—83. Medlin, J. E., P. Uguen, A. Taylor, D. L. Bentley, and S. Murphy. 2003. The C-terminal domain of pol II and a DRB-sensitive kinase are required for 3' processing of U2 snRNA. Embo J 22:925-34. Michels, A. A., A. Fraldi, Q. Li, T. E. Adamson, F. Bonnet, V. T. Nguyen, S. C. Sedore, J. P. Price, D. H. Price, L. Lania, and O. Bensaude. 2004. Binding of the 7SK snRNA turns the HEXIM1 protein into a P-TEFb (CDK9/cyclin T) inhibitor. Embo J 23:2608-19. Murphy, S., J. B. Yoon, T. Gerster, and R. G. Roeder. 1992. Oct-1 and Oct-2 potentiate functional interactions of a transcription factor with the proximal sequence element of small nuclear RNA genes. Mol Cell Biol 12:3247-61. Myslinski, E., A. Krol, and P. Carbon. 1998. ZNF76 and ZNF143 are two human homologs of the transcriptional activator Staf. J Biol Chem 273:21998- 2006. Nguyen, V. T., T. Kiss, A. A. Michels, and O. Bensaude. 2001. 7SK small nuclear RNA binds to and inhibits the activity of CDK9/cyclin T complexes. Nature 414:322-5. Sadowski, C. L., R. W. Henry, S. M. Lobo, and N. Hernandez. 1993. Targeting TBP to a non-TATA box cis-regulatory element: a TBP-containing 160 fl“ r-«e “war:- 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. complex activates transcription from snRNA promoters through the PSE. Genes Dev 7:1535-48. Schaub, M., E. Myslinski, C. Schuster, A. Krol, and P. Carbon. 1997. Staf, a promiscuous activator for enhanced transcription by RNA polymerases II and III. Embo J 16:173-81. Schramm, L., and N. Hernandez. 2002. Recruitment of RNA polymerase III to its target promoters. Genes Dev 16:2593-620. Schramm, L., P. S. Pendergrast, Y. Sun, and N. Hernandez. 2000. Different human TFIIIB activities direct RNA polymerase III transcription from TATA- containing and TATA-less promoters. Genes Dev 14:2650-63. Schulte, A., N. Czudnochowski, M. Barboric, A. Schonichen, D. Blazek, B. M. Peterlin, and M. Geyer. 2005. Identification of a cyclin T-binding domain in Heximl and biochemical analysis of its binding competition with HIV-1 Tat. J Biol Chem 280:24968-77. Sepehri, S., and N. Hernandez. 1997. The largest subunit of human RNA polymerase III is closely related to the largest subunit of yeast and trypanosome RNA polymerase III. Genome Res 7 :1006-1019. Teichmann, M., Z. Wang, and R. G. Roeder. 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: 14200-14205. Yang, Z., Q. Zhu, K. Luo, and Q. Zhou. 2001. The 7SK small nuclear RNA inhibits the CDK9/cyclin T1 kinase to control transcription. Nature 414:317-22. Yik, J. H., R. Chen, R. Nishimura, J. L. Jennings, A. J. Link, and Q. Zhou. 2003. Inhibition of P-TEFb (CDK9/Cyc1in T) kinase and RNA polymerase II transcription by the coordinated actions of HEXIM1 and 7SK snRNA. Mol Cell 12:971-82. Yik, J. H., R. Chen, A. C. Pezda, and Q. Zhou. 2005. Compensatory contributions of HEXIM1 and HEXIM2 in maintaining the balance of active and inactive positive transcription elongation factor b complexes for control of transcription. J Biol Chem 280:16368-76. Yoon, J. B., S. Murphy, L. Bai, Z. Wang, and R. G. Roeder. 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 15:2019-27. 161 46. Zhu, Y., T. Pe'ery, J. Peng, Y. Ramanathan, N. Marshall, T. Marshall, B. Amendt, M. B. Mathews, and D. H. Price. 1997. Transcription elongation factor P-TEF b is required for HIV-1 tat transactivation in vitro. Genes Dev 11:2622-32. 162 CHAPTER 5 SUMMARY Non-translated RNAs synthesized by RNA polymerase III contribute to the biosynthetic growth capacity of a cell (6). Deregulated transcription of these RNAs may cause unrestricted grth and therefore it is important to regulate RNA polymerase III transcription. Interestingly, the RB and p53 tumor suppressor proteins (2, 9) as well as the CK2 and Myc oncoproteins (1, 3) that are important for regulating cell proliferation, also regulate transcription of these genes. This observation indicates that transcriptional regulation of these RNA polymerase III transcribed genes may play an important role in controlling cellular proliferation. Genes transcribed by RNA polymerases 111 include those encoding SS rRNA, tRN A and U6 snRNA that are molecular components of the cellular machinery governing multiple steps in the flow of genetic information in cells. Of these genes, the human U6 snRN A gene is interesting because subtle changes in the core promoter architecture can switch transcription from RNA polymerase III to II (5). Thus the U6 snRNA gene and related family members provide a good model system to study the molecular mechanism of polymerase preference and activity during both normal and deregulated growth. All human snRN A genes contain a proximal sequence element (PSE) located in the core promoter region that is recognized by the general transcription factor called SNAPC. SNAPC plays a pivotal role in snRNA gene transcription by providing core promoter recognition and coordinating TBP activity as part of nucleating the preinitiation 163 complex assembly for both RNA polymerase II and III. In contrast to other promoter recognition complexes such as SL1 and TFIIIC that specialize in transcription by a single RNA polymerase, SNAPC is more functionally versatile due to its role in both RNA polymerase II and III transcription. SNAPC is composed of at least five subunits namely, SNAP19, SNAP43, SNAP45, SNAP50, and SNAP190. Although SNAP190 has been shown to contain a Myb domain in its sequence that contributes to DNA binding by SNAPC, it is not sufficient for this process (10). Indeed DNA binding by SNAPc is a cooperative event wherein SNAP190, SNAP50, and SNAP43 are all required ((7) and Figure 3-1A). These observations suggested that additional contacts are necessary for DNA binding. A role for SNAP50 in this process was suggested because cross-linking experiments showed that SNAP50 is in close proximity to the DNA (4). My mutagenesis studies revealed that SNAP50 does indeed play a role in SNAPC DNA binding through a highly conserved zinc finger domain in its C-terminus, that functions for preinitiation complex assembly for both RNA polymerase II and III transcription. As SNAPC binding to the PSE is a crucial early event in the preinitiation complex assembly at snRNA promoters, it is a target for regulatory factors. Indeed the Retinoblastoma (RB) tumor suppressor protein, which was shown to repress of U6 snRNA gene transcription, does interact with the SNAP50 subunit of SNAPC (8), This observation suggested that the RB-SNAPSO interaction could disable DNA binding by SNAPC thus explaining the mechanism for RB repression. However, subsequent studies have shown that RB does not affect DNA binding by SNAPC, indicating that RB may use some other mechanism. For example, it may recruit co-factors to repress U6 transcription. For other RB target genes such as the RNA polymerase II transcribed E2F- 164 regulated genes required for progression through the cell cycle, RB has been shown to recruit histone deacetylases (HDAC3) and components of the SWI/SNF ATP-remodeling complexes (12). Therefore, I examined the role of these RB co-factors during repression of U6 transcription. My studies revealed a role for HDACs and SWI/SNF in RB repression of U6 snRN A transcription. My data further indicates that endogenous HDACs and SWI/SNF proteins associate with the U6 promoters in 18435 cells that retain RB function but not in HeLa cells in which the RB firnction is compromised. These observations suggest that recruitment of co-repressor to the U6 snRN A promoter may be dependent upon RB ftmction. As a first step to address whether RB recruits co-repressor proteins to the U6 promoter in vivo I have successfully established RB overexpression in HeLa cells using transient transfection assays. Interestingly, my preliminary data suggests that one other RB co-factor called DNA methyl transferase 1 (DNMTl) associates with a U6 promoter in vivo only in the RB-transfected cells but not in a control empty vector- transfected or mock-treated cells as assayed by transient transfection assay followed by ChIP experiments. These results suggest that HDAC, SWI/SNF, DNMTl could be involved in RB-mediated repression of U6 transcription. My results further indicate that RB repression and HDAC activity are biochemically separable. Using this system, it will be possible to determine whether RB co-factors are recruited sequentially to the U6 promoter and whether chromatin affects this process. For example, RB can form different repressor complexes that contain HDACs or HDACs plus SWI/SNF. Indeed a RB-HDAC-SWI/SNF complex and the RB- SWI/SNF complex have been shown to repress cyclin E and cyclin A, respectively, whose genes products are required for cell cycle progression The sequential disassembly 16S of the RB-SWI/SNF/HDAC complex to one lacking HDAC activity is proposed as one mechanism governing RB repression of different genes at distinct phases of the cell cycle (11). It is possible that RB may utilize a similar mechanism wherein it associates with multiple corepressor complexes. My studies indicate that RB stimulates U6 promoter association by HDAC2 and the Brgl component of SW1/SNF complex (Chapter 2), Thus, an important future avenue of research will be to determine if and how RB coordinates the activity of multiple co-repressor proteins for repression of U6 transcription. RB repression of U6 snRNA transcription is a good model system to understand the mechanistic details of RB repression, as we have established in vitro repression assays using naked- as well as chromatin-DNA templates, and in vivo assays to study promoter association by RB and its cofactors. A link between the tumor suppressor function of RB and U6 repression has been suggested due to the observation that the region encompassing the AB pocket and the C-terminus of RB are the same regions required for tumor suppression and U6 repression by RB (9). These studies will help clarify the mechanism of RB repression as an important tool to understand RB activity during tumor suppression. 166 References 10. 11. Felton-Edkins, Z. A., N. S. Kenneth, T. R. Brown, N. L. Daly, N. Gomez- Roman, C. Grandori, R. N. Eisenman, and R. J. White. 2003. Direct regulation of RNA polymerase III transcription by RB, p53 and c-Myc. Cell Cycle 2:181-4. Gridasova, A. A., and R. W. Henry. 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 25:3247-60. Gu, L., W. J. Esselman, and R. W. Henry. 2005. Cooperation between small nuclear RNA-activating protein complex (SNAPC) and TATA-box-binding protein antagonizes protein kinase CK2 inhibition of DNA binding by SNAPC. J Biol Chem 280:27697-704. Henry, R. W., B. Ma, C. L. Sadowski, R. Kobayashi, and N. Hernandez. 1996. Cloning and characterization of SNAP50, a subunit of the snRNA- activating protein complex SNAPc. Embo J 15:7129-36. Hernandez, N. 2001. Small nuclear RNA genes: a model system to study fundamental mechanisms of transcription. J Biol Chem 276:26733-6. Hernandez, N. 1992. Transcription of vertebrate snRNA genes and related genes, p. 281-313, Transcriptional Reguation., In S. McKnight and K. Yamamoto ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor. Hinkley, C. S., H. A. Hirsch, L. Gu, B. LaMere, and R. W. Henry. 2003. The small nuclear RNA-activating protein 190 Myb DNA binding domain stimulates TATA box-binding protein-TATA box recognition. J Biol Chem 278:18649-57. Hirsch, H. A., L. Gu, and R. W. Henry. 2000. The retinoblastoma tumor suppressor protein targets distinct general transcription factors to regulate RNA polymerase III gene expression. Mol Cell Biol 20:9182-91. Hirsch, H. A., G. W. Jawdekar, K. A. Lee, L. Gu, and R. W. Henry. 2004. Distinct mechanisms for repression of RNA polymerase III transcription by the retinoblastoma tumor suppressor protein. Mol Cell Biol 24:5989-99. Wong, M. W., R. W. Henry, B. Ma, R. Kobayashi, N. Klages, P. Matthias, M. Strubin, and N. Hernandez. 1998. The large subunit of basal transcription factor SNAPc is a Myb domain protein that interacts with Oct-1. Mol Cell Biol 18:368- 77. Zhang, H. S., M. Gavin, A. Dahiya, A. A. Postigo, D. Ma, R. X. Luo, J. W. Harbour, and D. C. Dean. 2000. Exit from G1 and S phase of the cell cycle is 167 l2. regulated by repressor complexes containing HDAC-Rb-hSWI/SNF and Rb- hSWI/SNF. Cell 101:79-89. Zhu, L. 2005. Tumour suppressor retinoblastoma protein Rb: a transcriptional regulator. Eur J Cancer 41:2415-27. 168 APPENDIX A MULTIPLE SUBUNITS OF SNAPC CO-EXPRESSED IN E. COLI ARE ACTIVE FOR TRANSCRIPTION BY HUMAN RNA POLYMERASE 11 AND 1111 DNA binding by SNAPC is a crucial early event during preinitiation complex assembly for both RNA polymerase II and III transcribed human snRNA genes. Although mini-SNAPC (mSNAPc) that assembled by mixing the individual subunits namely SNAP190 (1-505), SNAP50, and SNAP43 is able to function in DNA binding, it is crippled for in vitro transcription (1). One possible explanation is that a post-translational modification of SNAPC is crucial for its activity and that this event is not successfully recapitulated in E. coli cells. This might also explain the observation that SNAPC expressed in a Baculaovirus expression system functions for transcription (2). Another possibility is that SNAPC does not efficiently obtain its fully active, native conformation when its subunits are expressed separately and reassembled. To test this hypothesis we devised a co-expression system wherein the individual subunits of mSNAPc were simultaneously expressed in the same E. coli cell. The co-expressed SNAPC is referred to as mSNAPcy4 hereafter. lFigures A-lB and A-lC used in this document were published in the following manuscript: Andrej Hanzlowsky, Blanka Jelencic, Gauri W. Jawdekar, Craig S. Hinkley, James H. Geiger, and R. William Henry (2006) Co-expression of multiple subunits enables recombinant SNAPC assembly and function for transcription by human RNA polymerase 11 and 111. Protein Expression and Purification Expression Vol.48; pp.215-223 169 mSNAPcy4 is functional for DNA binding and TBP recruitment to a U6 snRNA promoter We wanted to determine whether mSNAPcy4 was functional. The composition of mSNAPcy4 obtained by using a co-expression system is shown in Figure A-1A. The ability of mSNAPcy4 to bind DNA in an electrophoretic mobility shift assay was tested as shown in Figure A-lB. Increasing amounts of mSNAPcy4 was able to bind thSE/thATA probe DNA (lanes 2 and 3) and thSE/muTATA probe DNA (lanes 10 and 11), however failed to bind to a muPSE/thATA DNA probe (lanes 6 and 7) and muPSE/muTATA DNA probe (lanes 14 and 15). The mSNAPcy4 was functional for TBP recruitment to a probe containing thSE/thATA DNA (lane 4) but not to a thSE/muTATA DNA (lane 12). These results show that the mSNAPcy4 obtained using the co-expression system does indeed behave similarly to mSNAPc for DNA binding and TBP recruitment. mSNAPcy4 is able to reconstitute human U1 and U6 snRNA transcription in vitro We next wanted to test whether mSNAPcy4 was able to reconstitute in vitro transcription initiated from a plasmid containing the human U1 snRNA promoter as shown in Figure A-2A. HeLa cell nuclear extract was either mock depleted with pre- immune rabbit sera or depleted with anti-SNAP43 to remove endogenous SNAPC. In the absence of endogenous SNAPC, U1 transcription is reduced considerably but not in the mock depleted extract (compare lanes 1 and 2 with lane 3). When increasing amounts of mSNAPcy4 were added to the transcription reaction U1 transcription was restored (lanes 4 to 10). Similarly, as shown in Figure A-2B, U6 snRNA transcription was also restored 170 by increasing amounts of mSNAPcy4 (lanes 3 to 9), but not by a non-specific protein like GST (lane 10). These results show that mSNAPcy4 is indeed functional in an in vitro transcription assay. Thus we have been able to establish a system for co-purification of functional mSNAPcy4 that contains SNAP190 (1-505), SNAP10, SNAP43, and SNAP19. This system is overall far superior as it yields nearly a pure and homogenous complex, and the quantity of protein obtained is suitable for further structure-function characterization of this multi-protein transcription factor. 171 Figure A-l. mSNAPcy4 is competent for DNA binding and TBP promoter recruitment (A) Purified recombinant mSNAPcy4 was separated by 15% SDS-PAGE and visualized by staining with Coomassie blue (lane 2). Lane 1 contains a protein size markers. (B) Increasing amounts of mSNAPcy4 (3 ng and 10 ng) were added to EMSA reactions containing dsDNA probes harboring a wt PSE and wt TATA, mu PSE and wt TATA, wt PSE and mu TATA, or mu PSE and mu TATA box, as indicated. Lanes 4, 8, 12, and 16 contain approximately 50 ng of recombinant human TBP in addition to IOng of mSNAPcy4. Reactions containing only the DNA probe are shown in lanes 1, 5, 9, and 13. 172 A - g. s 5 <0 (I) 2 E 68" - — an- 100— u 75 —— . MW 1 -SNAP190 (1-505) 50 — C m —SNAP50 37 —- a w —-SNAP43 25 - o 20 — a 15 __ ~ —SNAP19 10 — u ' 1 2 Coomassie Blue staining B PROBE M— W W 11 II P' I TBP ---+---+---+---+ mSNAP’Y4- ‘-‘ -4-A ........... . ............ ,1," ‘4'??? Free . probe 12 3 4 5 6 7 8 910111213141516 EMSA 173 Figure A-2. mSNAPcy4 supports human snRNA gene transcription in vitro by both RNA polymerase II and III (A) The HeLa cell nuclear extract used for human U1 in vitro transcription assay was either mock depleted with a preimmune rabbit sera or anti-SNAP43 antisera to immunodeplete endogenous SNAPC. Reduction of the U1 signal upon removal of endogenous SNAPC is shown in lane 3. Increasing amounts of mSNAPcy4 (0.3 ng, 1 ng, 3 ng, 10 ng, 30 ng, 100 ng, or 300 ng) was able to reconstitute correctly initiated transcription from a human U1 promoter as shown in lanes 4 to 10. Lanes 1 and 2 show the U1 signal obtained from either untreated or mock-depleted reactions. RT represents a read-through transcript. (B) In vitro transcription of human U6 snRNA was carried out using HeLa cell nuclear extract that was treated as before in. Increasing amounts of mSNAPcy4 (0.3 ng, 1 ng, 3 ng, 10 ng, 30 ng, 100 ng, or 300 ng) was able to reconstitute correctly initiated transcription from a human U6 promoter as shown in lanes 3 to 9. Lane 2 shows the reduced U6 signal upon removal of endogenous SNAPC. Approximately 300ng of GST was added to the transcription reaction instead of mSNAPCy4 as shown in lane 10. 174 a-SNAPc depleted l ‘ 11"» ' .V ij'“ " 7:3 Anny», ..~-._.\, ‘~ . '. In .. 't . .1 » . v '. ’1/2 ‘.:.;.\~.fr“.v , ‘1' , " ,‘ ,‘ ‘ ., 1‘ ‘ , K- I ‘ 4' LI. '1 ' z m‘ u ‘ \ V/flifl. 47...; .1 ”1,1:- ”‘9? A»? -:-‘~,:;'.;.f x; fig»; by: 1 . ‘ . V ,‘ I V . . J . ‘ x I,” .-.‘ ‘('-;‘.T;*\- -. W .‘ '1' . . 1 RI. depleted a—SNAPc depleted l . ., 1 -- ~1:»' ,, . . ‘ ‘ . v t A ‘ J'é esr “~gg;-ueg 175 References Hanzlowsky, A., B. Jelencic, G. Jawdekar, C. S. Hinkley, J. H. Geiger, and R. W. Henry. 2006. Co-expression of multiple subunits enables recombinant SNAP(C) assembly and function for transcription by human RNA polymerases II and 111. Protein Expr Purif 48:215-23. Henry, R. W., V. Mittal, B. Ma, R. Kobayashi, and N. Hernandez. 1998. SNAP19 mediates the assembly of a functional core promoter complex (SNAPc) shared by RNA polymerases II and III. Genes Dev 12:2664-72. 176 APPENDIX B DIFFERENTIAL U6 snRNA PROMOTER ASSOCIATION IN VIVO BY RB FAMILY MEMBERS Pocket proteins associate with a U6 sNRNA promoter in vivo. The Retinoblastoma (RB) tumor suppressor protein is thought to contribute to growth control through its regulation of RNA polymerase III transcription Therefore to test whether RB function at U6 snRNA promoters is linked to growth, chromatin immunoprecipitations assay was performed from normal mammary epithelial cells (1 84B5) harvested at increasing cell densities. Populations of cells were harvested at low (~25%), intermediate (~75%), and high (~100%) cell densities. As shown in Figure B- 1A, both U6 (top panel) and U2 (middle panel) promoter DNA were significantly enriched in chromatin immunoprecipitations performed with anti-SNAP43 antibodies (lane 6) at all cell densities as compared the GAPDH exon2 negative control (bottom panel). Interestingly, measurable levels of RB were detected at the U6 snRNA promoter only in those cells harvested at intermediate cell density, but not at the low or high densities (lane 7). The pattern for RB enrichment in these experiments suggests that RB may regulate these genes when cells are actively growing but not when the cells have exited the cell cycle. Therefore, to characterize the relative percentage of cells in each phase of the cell cycle, cells grown at the different densities were analyzed for DNA content by flow cytometry. Indeed, the relative number if cells in GO/Gl increases as the cell density increases, indicating that when grown to 100% confluence most cells have 177 Figure B-l. U6 snRNA promoter occupancy by RB and RB family members is cell density dependent. (A.) U6 promoter occupancy by RB is sensitive to the cell density. Chromatin was harvested form normal human mammary epithelial cells (184B5) grown to approximately 25%, 75%, and 100% cell density. Immunoprecipitation reactions from each chromatin sample were performed using rabbit pre-immune sera (lane 5), anti-SNAP43 (lane 6), or anti-RB antibodies (lane 7). Immunoprecipitated material was analyzed for U6 snRNA and U2 snRN A promoter DNA (upper and middle panels, respectively) or GAPDH exon 2 DNA (bottom panel) by PCR using primers specific to each gene. Lanes 14 represent 10-fold serial dilution (10% to 0.001%) of input chromatin. Cells grown to similar density were processed for propidium iodide staining and the relative percentages of cells in 60/01, S, or 62 phase of the cell cycle were determined using FACS analysis. (B) Cell density influences distinct RB family member association with the human U6 promoter. Chromatin was collected from human 18485 cells that were grown to either intermediate or high densities. Immunoprecipitation reactions were performed using IgG (lane 2), anti-RB (lane 3), anti-p130 (lane 4), or anti-p107 (lane 5) antibodies. Immunoprecipitated DNA was analyzed for U6 snRNA (upper panel) and Cyclin A (lower panel) promoter DNA by PCR. Lane 1 indicates the input DNA. 178 WM! E E < 8 " E O %lnput égg 25%;: 74:: ‘ o. a as E 2’ 8 is is 1,: "l— -- ~— -- 125% F u. .. _. intermediate 4--- U6 snRNA t-..- us snRNA L- -— i.-. .. .. 75% g, __ huh 25:. f. .. .._.. .._.. .._. 110070 : ."1. b, . 'I' - intermediate 9-75: ”EL; an. m - 12570 CYCIII'IA .. ..- - high ”‘ 02 snRNA may... .._.... 75% ' ‘1 "2 '3‘ “"2" "5 b 25% ._ ]75% 5:: - -« ltoov. 5»...- 1 2 3 4 5 6 7 C )5'1' Cell density 25% 75% 100% GO/G1‘67 71 E s 25 2o 12 62 8 9 3 179 likely exited the cell cycle. However, the difference in cell cycle profile for cells grown at low and intermediate densities is subtle, and thus other contributing factors may govern whether RB regulates U6 transcription. One possible explanation could be that at lower cell-density most of the RB protein is in the hyperphosphorylated form and as the cell density increases there is a shift to the hyphosphorylated form of RB, which is believed to be transcriptionally active. Indeed there has been a report showing that as cell density increased from 13% to 43% there was an increase in hyperphosphorylated-RB as observed by Western blot analysis. However, as the cell density reached 75% and 100% there was more hypophosphorylated-RB (1). Prior studies have suggested that all three members of the RB family can regulate RNA polymerase III transcription (3, 5). Therefore, I wanted to determine whether p107 and p130 also associate with the U6 snRNA promoter. Chromatin immunoprecipitation assays were performed from cells grown to intermediate or high cell density and using antibodies for each RB family member. Enrichment of the cyclin A promoter was examined as a positive control for the p107 and p130 immunoprecipitations (6). As shown in Figure B-1B, the U6 snRNA promoter was enriched in the a-RB and a-p107 immunoprecipitations at the intermediate cell density conditions, but not when cells were grown at high densities. RB was not present at the cyclin A promoter during either growth condition whereas the cyclin A promoter was enriched in the p107 immunoprecipitation from cells grown at both intermediate and high density. In contrast, p130 associates with both promoters only at high cell densities. None of the RB pocket proteins was detected at the U6 promoter at low cell density nor was enrichment of the GAPDH exon 2 DNA observed (data not shown). Together these results suggest that all 180 three RB family members may participate in regulation of U6 snRNA expression but under different grth conditions. Data from our lab indeed shows that p107 and p130 also repress U6 snRNA transcription in vitro by RNA polymerase III (Xianzhou Song, unpublished data). Thus my data has revealed that both RB and p107 associate with U6 snRNA promoter DNA when cells are actively dividing. When cells have exited the cell cycle due to contact inhibition, neither RB nor p107 occupy a U6 snRNA promoter, but instead p130 now associates with the U6 promoter. Previously, p130 was shown to be mostly active in the GO/Gl phase while p107 is active in the S-phase (reviewed in (4). These observations are consistent with previous reports that both p107 and p130 can regulate RNA polymerase III transcription (5) My data indicates that RB and p107 may regulate RNA polymerase III activity during active growth and transition through the cell cycle whereas p130 may be active during growth arrest, and suggest that pocket protein family members regulate hmnan U6 snRNA gene transcription at distinct phases of cell growth. Methods and Materials Human mammary epithelial 184BS cells were used for the cell density experiments. The cells were seeded as follows: 0.15x107 cells into 40 plates (25%), 0.6x107 cell into 10 plates (75%), and 1.2x107 cells into 5 plates (100%). After 48 hrs cells were harvested form each density pool and processed for collecting chromatin as described before (2). A portion of the chromatin corresponding to 1x107 cells was then used for each immunoprecipitation reaction using anti-SNAP43 (C848), anti-RB (SC-1538) antibodies, or an irrelevant preimmune serum. For the ChIP experiment shown in Figure B-lB, anti- 181 p107 (SC-318), anti-p130 (SC-317) antibodies were additionally used. Immunoprecipitated DNA was examined for enrichment of U6 snRNA, U2 snRN A, and Cyclin A promoter DNA or GAPDH exon2 DNA using PCR amplification. For the FACS analysis done Figure B-lA, a portion of the harvested cells were washed and fixed with ice cold 70% ethanol. Subsequently, cells were stained with Propidium Iodide and the DNA content was analyzed using a FACS Vantage flow cytometer. 182 References 1. Hannan, K. M., B. K. Kennedy, A. H. Cavanaugh, R. D. Hannan, I. J. Hirschler-Laszkiewicz, L. S. Jefferson, and L. I. Rothblum. 2000. RNA polymerase I transcription in confluent cells: Rb downregulates rDNA transcription during confluence-induced cell cycle arrest. Oncogene. 2. Hirsch, H. A., G. W. Jawdekar, K. A. Lee, L. Gu, and R. W. Henry. 2004. Distinct mechanisms for repression of RNA polymerase III transcription by the retinoblastoma tumor suppressor protein. Mol Cell Biol 24:5989-99. 3. Larminie, C. G., C. A. Cairns, R. Mital, K. Martin, T. Kouzarides, S. P. Jackson, and R. J. White. 1997. Mechanistic analysis of RNA polymerase III regulation by the retinoblastoma protein. Embo J 16:2061-71. 4. Mulligan, G., and T. Jacks. 1998. The retinoblastoma gene family: cousins with overlapping interests. Trends Genet 14:223-9. 5. Sutcliffe, J. E., C. A. Cairns, A. McLees, S. J. Allison, K. Tosh, and R. J. White. 1999. RNA polymerase III transcription factor IIIB is a target for repression by pocket proteins p107 and p130. Mol Cell Biol 19:4255-61. 6. Takahashi, Y., J. B. Rayman, and B. D. Dynlacht. 2000. Analysis of promoter binding by the E2F and pRB families in vivo: distinct E2F proteins mediate activation and repression. Genes Dev 14:804-16. 183 APPENDIX C RB INTERACTS WITH MULTIPLE COMPONENTS OF THE U6 snRNA-SPECIFIC GENERAL TRANSCRIPTION MACHINERY FOR PROMOTER RECRUITMENTl RB interacts with the U6 snRNA-specific general transcription machinery. For RB to enact repression of U6 snRNA transcription RB might be recruited to the promoter either directly by binding to specific DNA control elements or through interactions with the general transcription factors. As RB does not have a DNA-binding domain, RB most likely is recruited to the DNA via interactions with SNAPC and TFIIIB. Therefore, GST pulldown experiments were performed to determine the region(s) of SNAP50 that is required for interaction with RB. The truncated SNAP50 mutant proteins used are represented in Figure C-lA. GST-RB (379-928) and the 35S-labeled SNAP50 proteins were expressed as before (4). GST-RB (379-928) interacted with the full-length SNAP50 (1-411) protein and this interaction was specific, as no interaction was seen with GST (lanes 2 and 3). SNAP50 (1-300) and SNAP50 (301-411) proteins interacted with GST-RB (379-928) suggesting that there might be two regions in SNAP50 involved in RB interaction. A bigger C-terminal deletion of SNAP50 containing amino acids 1-199 also interacted with RB, however the region in SNAP50 containing amino acids 1-124 and 123-199 did not interact with RB suggesting that the region in SNAP50 around amino acids 123/124 could be important for RB interaction. Interestingly, a LxCxE motif ' Figure C-2 used in this document was published in the following manuscript: Heather A. Hirsch, Gauri W. Jawdekar, Kang-Ae Lee, LiPing Gu, and R. William Henry (2004) Distinct mechanisms for repression of RNA polymerase III transcription by the Retinoblastoma tumor suppressor protein. Molecular and Cellular Biology Vol.24; pp.5989-5999 184 is present within this region, amino acid residues 109-113. The LxCxE sequence, which is a known motif for RB binding (3), may be important for SNAP50 interaction with RB. However, a truncation of SNAP50 containing amino acids 123-300, which lacks the LxCxE motif, also interacts strongly with RB, suggesting that a region outside the LxCxE motif may provide a region for interaction with RB. Whether the LxCxE motif can contribute to interactions with RB is not known. Other amino-terminal truncations tested, including SNAP50 (301-411), SNAP50 (123-411), SNAP50 (200-411) lacking the LxCxE motif interacted weakly with RB. Together these data provide evidence that the region of SNAP50 between amino acids 1-199, 123-300, and 301-411 may be important for RB interaction and provide a basis for finther mutational analysis. The region(s) in deI that are important for RB interaction were mapped in a GST pulldown assay. 0 and N- terminal deletion mutants of del were cloned and expressed as 35S-labeled proteins. The de1 protein contains a SAN T domain in the N- terminal amino acids. Though the exact function of the SANT domain is not known, in other proteins it has been implicated in DNA binding (1). de1 contains a series of repeats in the C-terminal two-thirds of the protein with potential phosphorylation sites. As shown in Figure C-1B, full length del (1-1338) protein, de1 (471-1338) protein, and de1 (823-1338) protein interacted strongly with GST-RB (379-928) (lane 2) but not with GST or beads alone (lanes 3 and 4). However, the N-terminal region containing amino acids 1-470 did not interact with GST-RB (379—928), suggesting that the C- terminal two-thirds of del is important for RB interaction and the amino acids between 823-1338 are sufficient for this interaction. RB could potentially be recruited to the U6 promoter via these interactions. 185 Figure C-l. Characterization of the regions in SNAP50 and de1 that are required for RB interaction. (A) The RB pocket domain interacts with at least three regions in SNAP50. Schematic representation of SNAP50 proteins containing the indicated deletions is shown in the left panel. GST pulldown analysis was performed to map the regions in SNAP50 that interact with RB. SNAP50 proteins containing deletions were expressed In vitro using rabbit reticulocyte lysate and labeled with 35 S methionine. Equal amounts of the SNAP50 proteins were incubated with approximately 1 ug GST- RB (379- 928) (lane 3) and 1 pg GST (lane 2). Complexes were collected with glutathione agarose beads for 2 hrs at 4°C. Bound proteins were washed and eluted in Laemmlie buffer. Proteins were separated by SDS-PAGE and visualized by autoradiography. Lane 1 shows 10% of the input. (B) The C- terminal region of del is important for RB interaction. Schematic representation of the de1 proteins harboring the indicated deletions are shown In the lefi panel. GST pulldown assays were carried out with3 5S-methionine-labeled de1 proteins and GST-RB (379- 928) as in (A). 186 a N 0} d. [x '5 £9. ‘2 E \o ('3 P" .32 o 8 1 LxCx_E . [ SNAP50 (1-411) ' SNAP50 (1 —300) ' SNAP50(301-411) 1 199 ~ SNAP50 (1.199) 1 124 L :1] SNAP50 (1-124) 123 199 ‘ SNAP50 (123.199) 123 300 SNAP50 (123-300) SNAP50 (123-411) rm l a” W l l “ l SNAP50 (ZOO-411) L________e 1 2 3 187 10 % INPUT 3 6: 5. m a; 1.. U) (5 GST 15.15: Beads de1 (1-1338) l>>>>>>>>>>>§l de1(1-470) ' 33;}- f .1, de1 (471 -1338) " "-‘ —. j]: : h I D>>>>>>>>>>>i>3§l 873 1338 , . : " sill de1 (873-1338) »>>>>>>>>»>>>>:| ;' 2731;; ~ " ' *3 SNAP50 188 To further understand the mechanism of RB repression of U6 transcription, the RB domains that are required for repression of U6 transcription were characterized. Therefore, RB proteins containing the A domain but lacking additional regions of the carboxy terminus were analyzed. Previous observations show that RB interacts with SNAPC (4) and TFIIIB (2, 6). That RB does not occupy a human U1 snRNA gene promoter even though SNAPC is required for transcription of U1 by RNA polymerase II, suggests that RB interaction with SNAPC and TFIIIB represents a mechanism by which RB is specifically recruited to the U6 snRNA promoter to repress transcription. GST pull down experiments were performed to identify the regions of RB necessary for interaction with components of SNAPC and TFIIIB. Each component of the basal transcription machinery was individually expressed and labeled with 35S-methionine in rabbit reticulocyte lysate. Expression of these labeled proteins is shown in Figure C-2B (lane 1). Equivalent amounts of GST-RB (3 79-928) or GST-RB proteins containing the indicated truncations were incubated with the labeled proteins. Strong interactions between GST- RB (379-928) and two components of SNAPC, SNAP43 and SNAP50 were observed. Specific interactions of GST-RB (379-928) with the TBP, Brfl, and de1 components of TFIIIB were also observed, although the interactions with TFIIIB appear to be weaker as compared to those with SNAPC components. These interactions are specific because neither GST nor beads alone bound to any of the SNAPC or TFIIIB proteins (lanes 6 and 7). In contrast GST-RB (379-928) did not interact with Brf2 or Oct-l in these assays (lane 2). More interestingly, GST-RB (379-870), which repressed U6 transcription, maintained the ability to interact with components of SNAPC and TFIIIB (lane 3), but GST-RB (3 79-772), which failed to repress U6 transcription, showed 189 Figure C-2. The A/B pocket domain and the C-terminal region of RB are required for interactions with RNA polymerase III-general transcription machinery. (A) Schematic representation of the GST-RB proteins containing the indicated deletions. (B) Characterization of the RB regions required for interactions with RNA polymerase HI basal transcription machinery. GST-pull down analysis was performed to determine the region of RB that can interact with each component of RNA polymerase III basal machinery. SNAP43, SNAP50, TBP, Brfl, Br12, del, and Oct-1 were expressed in vitro and labeled with 35S-methionine. Lane 1 shows 10% of each protein that was added to the reaction. The various GST-RB proteins containing deletions were incubated with each 35S-methionine labeled protein (lanes 2-5). GST or beads alone were used as controls (lanes 6 and 7). The stable protein complexes were purified using glutathione sepharose. The beads were extensively washed and bound proteins were separated by SDS-PAGE. Associated proteins were visualized by autoradiography. 190 re re:- I IO.--’J. , , . l TBP " J Brfl ' . 7": ,3. n, . j SNAP50 GST-RB (379—870) GST-RB (1379-772) GST-RB (379-577) GST-RB Protein ID U6 snRNA repression SNAP43 SNAP50 TBP de1 Ber GST-RB (379-928) GST-RB (379-870) GST-RB (379-772) GST-RB (379-577) 191 reduced interactions with SNAP43, TBP, del, and Brfl (lane 4). Thus these interactions might be critical for RB repression of U6 snRN A transcription (summarized in the table). Although GST-RB (379-772) interacted strongly with SNAP50, this interaction alone is not sufficient to maintain RB repression. Finally, GST-RB (379-577), which contains only the A domain, was not able to interact with any of the SNAPC or TFIIIB proteins (lane 5) consistent with its inability to repress transcription. These observations show that the NB pocket domain and the C-terminal region of RB are important for interactions with the U6 snRNA-specific general transcription factors. It is possible that RB is recruited to the promoter DNA via multiple protein-protein interactions with SNAPc and TFIIIB. Sequential ChIP experiments indeed show that RB co-occupies the same U6 snRNA gene promoter with SNAPC and TFIIIB in vivo (5). Interestingly, proteins harboring progressively increasing C-terminal deletions lose their ability to repress transcription, indicating that the NB pocket domain and the C region are necessary for repression of RNA polymerase III transcription (5). 192 J“ W. us. “A: References 1. Aasland, R., A. F. Stewart, and T. Gibson. 1996. The SANT domain: a putative DNA-binding domain in the SWI-SNF and ADA complexes, the transcriptional co-repressor N-CoR and TFIIIB. Trends Biochem Sci 21:87-8. 2. Chu, W. M., Z. Wang, R. G. Roeder, and C. W. Schmid. 1997. RNA polymerase III transcription repressed by Rb through its interactions with TFIIIB and TFIIIC2. J Biol Chem 272:14755-61. 3. Harbour, J. W., and D. C. Dean. 2000. Chromatin remodeling and Rb activity. Curr Opin Cell Biol 12:685-9. 4. Hirsch, H. A., L. Gu, and R. W. Henry. 2000. The retinoblastoma tumor suppressor protein targets distinct general transcription factors to regulate RNA polymerase III gene expression. Mol Cell Biol 20:9182-91. 5. Hirsch, H. A., G. W. Jawdekar, K. A. Lee, L. Gu, and R. W. Henry. 2004. Distinct mechanisms for repression of RNA polymerase III transcription by the retinoblastoma tumor suppressor protein. Mol Cell Biol 24:5989-99. 6. Larminie, C. G., C. A. Cairns, R. Mital, K. Martin, T. Kouzarides, S. P. Jackson, and R. J. White. 1997. Mechanistic analysis of RNA polymerase III regulation by the retinoblastoma protein. Embo J 16:2061-71. 193 AR l11111111111111111111