LIBRARY Mlchlgan State Unlverslty 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 6/01 c:/CIRCJDaIeDue.p65-p.15 REGULATION OF U6 SMALL NUCLEAR RNA TRANSCRIPTION BY THE RETINOBLASTOMA TUMOR SUPPRESSOR PROTEIN By Heather Anne Hirsch A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Program in Cell and Molecular Biology 2003 ABSTRACT REGULATION OF U6 SMALL NUCLEAR RNA TRANSCRIPTION BY THE RETINOBLASTOMA TUMOR SUPPRESSOR PROTEIN By Heather Anne Hirsch The Retinoblastoma tumor suppressor protein (RB) participates in many cellular functions including cell cycle progression, apoptosis, differentiation, and growth control. Key to the ability to participate in each of these processes is RB’s ability to regulate gene expression. Postulated repression mechanisms suggest that RB directly blocks pre- initiation complex assembly or recruits additional co-factors such as histone deacetylases or ATP-dependent chromatin remodeling machines whose activities impair RNA polymerase II access to promoters. RB also represses transcription of non-translated genes that are transcribed by RNA polymerases I and III, potentially to control cell growth. To further understand how RB regulates gene expression, we examined RB repression of two classes of RNA polymerase III transcribed genes, the Adenovirus 2 VAI gene (similar to tRNA genes in promoter architecture and factor requirements) and the U6 snRNA gene. Herein we demonstrate that RB associates with the endogenous U6 snRNA promoter in viva, an important step in the repression of a target gene. RB did not however, associate with RNA polymerase II transcribed snRNA genes (U1 and U2) or repress transcription of the U1 snRNA gene. We also demonstrate that the general transcription factors snRNA activating protein complex (SNAPc) and TFIIIB are important for RB repression of human U6 snRNA gene transcription by RNA polymerase III. RB interacts with these basal factors that are required for RNA polymerase III transcription providing a potential mechanism for RB recruitment to target genes. Together, SNAPc and TFIIIB act cooperatively to recruit RB to a U6 snRNA promoter in vitro and TFIIIB acts as a selectivity factor specifically recruiting RB to RNA polymerase III transcribed snRNA genes. Additionally, we show that RB co-occupies the U6 promoter with RNA polymerase III in vivo and RB repression in vitro does not preclude RNA polymerase III recruitment. These results suggest a novel mechanism wherein RB represses transcription at steps subsequent to RNA polymerase recruitment to gene promoters. AKNOWLEDGMENTS I would like to thank Dr. R. William Henry for his mentorship and guidance. I wish to thank my thesis guidance committee: Dr. David Arnosti, Dr. Susan Conrad, Dr. Ronald Patterson, and Dr. Steven Triezenberg. I would also like to thank members of the Henry laboratory past and present, especially Dr. Craig S. Hinkley and Gauri Jawdekar. I would also like to thank the graduate students from various programs for hours of scientific discussion, numerous reagents, and emotional support especially Kirsten F ertuck, Fransisco Herrera, Pete Davidson, Rich Grey, Saren Ottosen, Meghana Kulkami, and Paolo Struffi. I also thank my family for years of undying love and support. Finally, I would also like to thank my husband, Jim Mann, for being my unyielding pillar of support and center of my sanity. iv TABLE OF CONTENTS LIST OF FIGURES .............................................................................. viii KEY TO SYMBOLS AND ABBREVIATIONS ............................................. x CHAPTER 1: Introduction .......................................................................................... 1 CHAPTER 2: The retinoblastoma tumor suppressor protein targets distinct general transcription factors to regulate RNA polymerase [11 gene expression Abstract ................................................................................ 40 Introduction ........................................................................... 41 Materials and Methods ............................................................... 46 Results ................................................................................. 50 Discussion ............................................................................. 73 References ............................................................................. 82 CHAPTER 3: Retinoblastoma protein and RNA polymerase III concurrently occupy a repressed human U6 promoter Abstract ................................................................................ 89 Introduction ........................................................................... 90 Materials and Methods ............................................................... 93 Results ................................................................................. 98 Discussion ........................................................................... 1 20 References ........................................................................... 125 CHAPTER 4: The intact Large A/B pocket domain and C-terminus of R8 is required for efficient repression of RNA polymerase III transcription Abstract .............................................................................. 129 Introduction .......................................................................... 1 29 Materials and Methods ............................................................. 131 Results ................................................................................ 135 Discussion ........................................................................... 1 59 References ........................................................................... 165 CHAPTER 5: Summary .......................................................................................... 167 APPENDIX A: Recruitment of Basal Factors to the U6 snRNA promoter ................................... 179 Results and Discussion ............................................................. 180 Materials and Methods .............................................................. 191 References ........................................................................... 194 APPENDIX B: Potential co-factors that regulate snRNA gene expression ................................... 195 Results and Discussion ............................................................. 195 Actin association with potentially active U6 snRNA promoters ....... 195 HMG-l association with snRNA promoters ................................. 198 p70: a potential co-repressor for RB? ....................................... 201 Materials and Methods .............................................................. 205 References ........................................................................... 208 APPENDIX C: Subcellular localization of SNAPc and T F [113 components ................................. 209 Results and Discussion ............................................................. 209 Materials and Methods .............................................................. 212 References ........................................................................... 217 vi Figure 1-1: Figure I-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: Figure4-l: Figure 4-2: LIST OF FIGURES Representation of RNA polymerase III transcribed promoters. Factor requirements for RNA polymerase III transcribed genes. RB represses in vitro transcription by RNA polymerase III Different factors reconstitute adenovirus VAI and human U6 snRNA gene transcription in GST-RB (379-928) treated extracts. Endogenous RB is associated with SNAPC. Endogenous RB co-fractionates with SNAPc during chromatographic purification. RB interacts with two components of SNAPc Model for RB repression of human U6 snRNA gene transcription. RB selectively occupies U6 snRNA promoters in vivo and specifically represses U6 snRNA transcription in vitro. RB interacts with components of TFIIIB complexes TFIIIB acts as a selectivity factor to recruit RB specifically to RNA polymerase III transcribed snRNA promoters Contribution of individual components of TFIIIB to RB recruitment to a U6 snRNA promoter: RB co-occupies the same endogenous U6 snRNA promoter as SNAPC, TFIIIB and RNA polymerase III. RB and RNA polymerase III co-occupy the same repressed U6 snRN A promoter in vitro. Amino acids 379-870 of RB contain the minimal region RB occupancy of RNA polymerase III transcribed promoters in vitro and in vivo vii 53 57 63 67 72 79 101 105 108 111 115 118 137 140 Figure 4-3: Figure 4-4: Figure 4-5: Figure 4-6: Figure 4-7: Figure 4-8: Figure A-l: Figure A-2: Figure A-3: Figure 8-1: Figure B-Z: Figure B-3: SNAPc can recruit RB to promoter DNA. Addition of or-RB antibodies precludes interaction with SNAPc. Characterization of the region of RB necessary for interaction with SNAPc bound to U6 promoter DNA. Characterization of the region in RB that can interact with RNA polymerase III basal machinery. SNAP43 (234-368) is the minimal region required for interaction with RB RB repression of class 2 and class 3 RNA polymerase III transcribed genes. SNAPc assembly and binding to U6 snRNA promoter DNA in vitro: Mini-SNAPc can recruit Brf2 to a U6 snRNA promoter. SNAPc and TFIIIB assembly at a U6 snRNA promoter in vitro. Actin and SNAPc or RNA polymerase III co-occupy the same U6 snRNA promoter in vivo. HMG-l occupies snRNA promoters in vivo. p70 associates with the U6 snRNA promoter in vivo and interacts with RB. Figure C- l : Subcellular Localization of components of SNAPc and TFIIIB. Figure C-2: Cell density affects subcellular localization of de1. viii 145 148 151 154 158 164 182 186 190 197 200 203 211 214 ATP de1 Brf- l Brf-1_v2 Brf-2 Cch CDK ChIP Co-IP DHFR DNA EMSA GAPDH HAT HDAC HMT HP 1 IC R MCM MEF mS NAPc KEY TO ABBREVIATIONS Adenosine triphosphate B double prime TFIIB related protein 1 TFIIIB related protein 1 splice variant 2 TFIIB related protein 2 Cell division cycle 2 protein Cyclin dependent kinase Chromatin immunoprecipitation Co-immunoprecipitation Dihydrofolate reductase Deoxyribonucleic acid Electrophoretic mobility shift assay Glyceraldehyde — 3 —- phosphate dehydrogenase Histone acetyl transferase Histone deacetylase Histone methyltransferase Heterochromatin protein 1 Internal control region Mini-chromosome maintenance protein Mouse embryonic fibroblast Mini-SNAPC ix NTP PcG PCNA PCR RB RT-PCR RRMZ SNAPc SnRNA SuVAR39H 1 TBP TFIIA TFIIB TFIID TFIIF TFIIIA TFIIIB TFIIIC TK TS Nucleotide triphosphate Polycomb group complex Proliferating cell nuclear antigen Polymerase chain reaction Retinoblastoma tumor suppressor protein Ribonucleic acid Reverse transcriptase polymerase chain reaction ribonucleotide reductase M2 Small nuclear RNA activating protein complex Small nuclear RNA Suppressor of varigation TATA binding protein Transcription factor II A Transcription factor II B Transcription factor II D Transcription factor II F Transcription factor 111 A Transcription factor 111 B Transcription factor 111 C Thymidine kinase Thymidylate synthase I r I ‘1‘, flit: Chapter 1 Introduction RNA Polymerase III In eukaryotic organisms, nuclear genes are transcribed by three highly related RNA polymerases: RNA polymerase I, II, and 111. Each of these polymerases is responsible for transcription of a distinct subset of genes. RNA polymerase I synthesizes a single tandemly arrayed set of non-translated structural RNAs that assemble into ribosomes. RNA polymerase II transcribes a diverse set of genes including protein-encoding genes and some non-translated uridine rich snRNA genes. RNA polymerase III produces an assorted collection of non-translated structural and catalytic RNAs including small RNAs that are usually shorter in length than 400 nucleotides (36, 1 13). Structure of genes transcribed by RNA polymerase III The genes transcribed by RNA polymerase [11 fall into four classes characterized by promoter architecture (Figure I-l). Class 1 and 2 genes, represented by the SS ribosomal RNA (SS rRNA) and transfer RNA (tRNA) genes respectively, contain required gene internal promoter elements. Class 3 genes are characterized by external promoter elements and are represented by the U6 snRNA gene and the 78K gene. Class 4 genes exhibit a combination of the elements found in the first three classes. Figure I-l: Representation of the three classes of genes transcribed by RNA polymerase 111. Class 1 genes such as SS rRNA genes contain gene internal promoter elements including A and C boxes that are separed by an intermediate element (IE). Together, these elements comprise the internal control region (ICR) that is required for gene expression. Class 2 genes such as the mammalian tRNA gene also contain gene internal cis-acting elements referred to as the A and B boxes. Class 3 genes (U6 snRNA) are characterized by gene external promoter elements including the distal sequence element (DSE), the proximal sequence element (PSE) and a TATA element. Class 4 genes such as the vault RNA genes or EBER gene from the Epstein Barr virus, contain a mixture of all three classes of promoter elements. ICR l—P l'_‘——I Class1 {HID—- ss rRNA A IE C Class 2 F’U—D—- tRNA A B Class 3 m/l—fl—Dr, us snRNA DSE PSE TATA fl—flv’fl—{I-Ij—H— Vault DSE DSE PSE TATA A B Class 4 DZ-H— seem TATA A B SS rRNA genes (class 1) contain an A box, an intermediate element (IE), and a C box that is highly conserved through many species. These sequence elements combined constitute the internal control region (ICR) that is required for transcription (5, 95-97). For example, in Xenopus laevis, the A box is located at +50 to +64, the IE between +67 and + 72, and the C box is located from +80 to +97 (96). The spacing of these elements is important, as effective transcription of SS rRNA genes is intolerant of changes in spacing (96). In S. cerevisiae, only the C box is required for efficient expression of 58 genes (10). Class 2 gene promoters, Ad2 VAI (adenovirus gene product similar to tRNA) and tRNA genes, consist of an A box (e.g. X. laevis leucine tRNA +8 to +19) and B box (+52 to +62) region downstream of the start site of transcription (35, 119). These intragenic sequences are extremely well conserved from species to species perhaps in part because these regions encode the T and D loop regions that are critical for tRNA function. In contrast, the spacing between the A and B boxes is variable. However, recently it has been shown that some extragenic sequences may be necessary for class 2 gene transcription. For example, yeast U6 snRNA genes appear to have an extragenic TATA like element (7). Similarly, some tRNA genes in D. melanogastor also appear to contain TATA elements upstream of the start site of transcription that may be important for efficient gene expression (127). Additionally, the —30 to —25 bp region of C elegans tRNA genes may also contain sequences required for the transcription of these genes (68). Class 3 genes as represented by the mammalian U6 snRNA and 78K genes, are characterized by gene external core promoter elements. The enhancer regions of these genes contain a distal sequence element (DSE) that includes octamer sites, as well as sites for other transcriptional activators such as Spl and Staf (69, 112). The DSE activates transcription from the core promoter of Class 3 genes and can be located at variable positions upstream of the core promoter from species to species. In mammalian systems the DSE is located approximately 239 bp upstream of the start site of transcription (121, 150). The core promoter regions of class 3 genes consist of two essential elements: the proximal sequence element (PSE) and a TATA element. These cis-acting elements are located at approximately -46 and -20 bp upstream of the start site of transcription, respectively. The spacing of these two elements is invariant in human genes. Class 4 genes represent a mix of characteristic sequence elements found in the other three classes of RNA polymerase 111 genes. These class 4 genes include vault genes (130, 132), selenocysteine tRNA, EBER2 (Epstein-Barr viral product) (47, 48), MRP RNase (involved in maturation of mitochondrial DNA replication), Y1 and Y3 (unknown function). In the case of vault RNAs and selenocysteine tRNA, the promoters of these genes contain gene external PSEs as well as gene internal control elements. Additionally, many human vault RNA genes contain distal sequence elements similar to those found in snRNA genes. The Epstein-Barr gene, EBER contains a TATA element as well as gene internal A and B box. RNA polymerase III Basal Transcription Machinery Just as each class of RNA polymerase III transcribed gene contains characteristic promoter sequences, each class of RNA polymerase III transcribed genes has distinct, specific basal machinery requirements, as discussed in more detail in the following sections (figure I-2). SS rRNA (class 1) genes require the binding of TFIIIA to the ICR of the promoter followed by recruitment of TFIIIC. Interaction of TFIIIB with TFIIIC allows TFIIIB to associate with the promoter, subsequently recruiting RNA polymerase III. For preinitiation complex formation to occur at tRNA (class 2) genes, TFIIIC must bind to both the A and B box of the promoters followed by interaction with TFIIIB. Positioning of TFIIIB near the start site of transcription allows for RNA polymerase III recruitment. U6 snRNA genes (class 3) require different factors. This set of genes requires cooperative interactions between Oct-1, SNAPc (snRNA activating protein complex), and a variant TFIIIB. At this time, the factor requirements for class 4 genes are unclear. One possibility is that a blend of class 2 and class 3 transcription factors are required, depending on specific promoter structure (47, 48, 130, 132). TFIIIA: TFIIIA is a basal factor required to recruit TFIIIC specifically to SS rRNA genes. A founding member of C2H2 family of zinc finger proteins, Xenopus laevis TFIIIA was the first eukaryotic transcription factor to be purified to homogeneity and to have its cDNA Figure I-2: Factor requirements for RNA polymerase III transcribed genes. Both class 1 and class 2 RNA polymerase III transcribed genes require the TFIIIC complex and a TFIIIB complex consisting of del, TBP, and Brfl. Class 1 genes have an additional requirement for TFIIIA which provides DNA binding specificity during pre-initiation complex formation. Class 3 genes have very different factor requirements. Transcriptional activators such as Oct-l bind to the DSE to activate transcription. A multi—protein complex called SNAPc binds to the PSE to nucleate pre-initiation complex assembly and is important for subsequent gene expression. An alternative TFIIIB complex consisting of del, Brf2, and del associates with the TATA element and may also play an important role in RNA polymerase III recruitment. O de1 erc cram TBP 819;",5 SSrRNA ' CHwZ tRNA In. ,..'L-.-. 'nm)‘ ‘ v 45. ' ' TBP de Chea IEEEII lBsmNA /é/ cloned (37). When TFIIIA binds to the promoter of the SS RNA gene, its nine zinc fingers are aligned over the ICR with the C terminus of the protein near the 5’ end of the promoter and its N-terminus at the 3’ end of the promoter. The C block of the promoter is recognized by the three end most zinc fingers at the N terminus and the A block is recognized by zinc fingers 7-9 (18, 33, 89). The middle three fingers adopt a completely different conformation in order to span the intervening DNA. After binding to SS rRNA genes, TFIIIA provides a platform for the recruitment of TFIIIC, which has little affinity for the SS rRNA gene alone. TFIIIC TFIIIC is a required basal transcription factor that enables pre-initiation complex formation to occur at all RNA polymerase III transcribed promoters in yeast and the 5S and tRNA genes in mammalian systems. The mammalian U6 snRNA gene does not require TFIIIC but rather another basal factor, SNAPc, which is discussed in detail below. Productive recruitment of TFIIIC to SS rRNA promoters requires interaction with TFIIIA, whereas recruitment to tRNA promoters and yeast U6 snRNA promoters is mediated through sequence specific DNA contacts by TFIIIC. Yeast TFIIIC consists of six subunits (36, 113) whereas human TFIIIC includes 5 subunits. The TFIIIC complex can be separated into two domains separated by a flexible linker: TA which binds weakly to the A box and 1:3 which binds with high affinity to the B box (78). The linker between the domains confers a flexibility to the TFIIIC protein complex that allows for binding to tRNA promoters that have varied distances between the A and B boxes. Stable association of TFIIIC with promoters allows for recruitment and correct positioning of TF 111B. TFIIIC has also been shown to contact RNA polymerase III itself (50) suggesting that although TFIIIB alone is sufficient to recruit polymerase, contacts with TF [11C may also be important. SNAPc Mammalian class 3 RNA polymerase III transcribed genes have slightly different factor requirements, perhaps in part due to the presence of unique gene external promoter elements. The QRNA activating protein complex (SNAPc) also called the proximal element transcription factor (PTF) (2, 146, 147) is required for snRNA expression including the U6 snRNA gene which is representative of Class 3 genes. SNAPc is a multi-protein complex consisting of at least five subunits as designated by apparent molecular mass: SNAP190 (PTFor) (32, 145), SNAPSO (PTFB) (2, 41), SNAP45 (PTFS) (108, 147), SNAP43 (PTFy) (43, 147), and SNAP19 (42). SNAPc is specifically recruited to the PSE of snRNA genes and is important for nucleation of pre-initiation complex formation (41-43, 72, 109, 145). Further characterization of the SNAP complex demonstrated that the minimal complex (mSNAPc) necessary for DNA binding and transcriptional activation consists of SNAP190 (l-SOS), SNAP43, and SNAPSO (44, 74, 83). SNAPc interacts with TBP and cooperatively binds to DNA with TBP (82) suggesting that SNAPc plays a role in recruitment of TFIIIB to U6 promoters. TFIIIB TFIIIB plays a central role in the establishment of a pre-initiation complex at the promoters of RNA polymerase III transcribed genes. TFIIIB is recruited to promoters lO through interactions with TFIIIC (58-60) or SNAPc (13, 44). Subsequent to recruitment, TFIIIB is responsible for bringing RNA polymerase to promoters to initiate transcription (57, 133, 144). Three proteins compose TFIIIB complexes: the TATA binding protein (TBP), TFIIB related factor (Brf), and B” (de) (58-60, 64, 100, 117, 129). Most of the information currently available for how TFIIIB complexes are assembled and how they function for RNA polymerase III transcription comes from studies in S. cerevisiae. In yeast, TBP and Brfl form a very stable complex (designated B’) from which the less tightly associated B” is separable by chromatography (58). The Brfl subunit plays a central role in maintaining TFIIIB complex integrity through extensive contacts with TBP and de1. Less is known about the TFIIIB complexes that function in human systems. It appears that multiple variant complexes may exist that function at different classes of RNA polymerase III transcribed genes (81, 114, 125). Human Brfl and yeast Brf are highly homologous, especially in the TFIIB related N-terminal half (133). Human del is a very large protein that has some similarity (~40%) to yeast B” along a 400 amino acid stretch that encompasses the indispensable SANT domain (1 14). At the human U6 promoter, the TATA box directly recruits TBP, which is likely to be important for recruiting additional BRF-related factor(s), and del (114). In contrast to most RNA polymerase III-transcribed genes, human U6 snRNA gene transcription does not require Brfl (79, 81). While the TFIIIB complex containing Brfl and TBP clearly does not function for human U6 transcription (81), the exact nature of the TBP complex 11 that does function at human U6 promoters is currently unclear. Two proteins analogous to Brfl have been shown to be important for U6 snRNA gene expression. Brf2 associates with U6 promoters in vivo (114) and is necessary for gene expression as determined by immunodepletion-reconstitution experiments (13, 114). Additionally, in vitro transcription experiments performed using highly purified recombinant SNAPc, TBP, del, Brf2 and chromatographic fractions enriched for RNA polymerase 111 support U6 snRNA transcription, suggesting that these proteins define the minimal set of factors required for U6 transcription (13). However, a splice variant of Brfl called Brfl_v2 has also been shown to participate in U6 gene expression in immunodepletion experiments (79). Compared to Brfl, Ber has a conserved zinc finger and core domains and a divergent C-terminus. Brfl_v2 lacks the zinc finger and lst repeat domain conserved in Brfl and TFIIB. The role that each of these proteins plays in U6 snRNA gene expression is unclear. However, both proteins may be in a complex that is required for U6 snRNA transcription. RNA polymerase III enzyme RNA polymerase III has been well defined in S. cerevisiae and has been shown to have 17 subunits. RNA polymerase 111 contains ten subunits that are unique to RNA polymerase III and are designated the C subunits. The two common subunits shared by RNA polymerase I and III are designated the AC subunits. RNA polymerase III also contains five subunits common to all three polymerases (ABC subunits) (46). Human RNA polymerase III has been characterized both by chromatography (133, 134) and from cell lines expressing epitope tagged subunits (51, 133, 134). All yeast subunits except 12 RPC37 have been disrupted and shown to be essential for viability (1 1). All of the human RNA polymerase III subunits have now been characterized by mass spectrometry after purification fi'om human cell lines contained double epitope tagged HsRPC4 (51). This analysis found human orthologues to all of the yeast RNA polymerase III subunits except for RPCIO, which may not have been detected due to small size. The Retinoblastoma Tumor Suppressor Protein The Retinoblastoma tumor suppressor protein (RB) is a critical regulator of important cellular processes including cell cycle progression (24, 25, 39, 40, 56, 80, 86, 101, 111, 135, 149), DNA replication (105), growth regulation (118, 140), differentiation (12, 38, 90, 99, 118, 126, 148), and apoptosis (49, 148). RB is frequently mutated in a variety of human malignancies and tumors. Approximately 30% of all human cancers are deficient for RB activity, and the RB signal transduction pathway is disrupted upstream in 50% of human cancers. RB was originally identified as a result of frequent mutation in the rare pediatric eye tumor (34, 67). Cancers that have been shown to be deficient in RB activity include osteosarcomas, small cell lung cancers, bladder, prostate, and breast cancer (17). RB belongs to a family of pocket proteins that also includes p107 and p130 (135). These proteins were originally characterized by the presence of a large A/B pocket domain that was identified by the ability to bind to viral oncoproteins, including SV40 Large T antigen, Adenovirus Ela, and Papilloma virus E7 (27, 28). The Large A/B pocket extends from amino acids 379-869 and is required for tumor suppression. This region can be 13 further subdivided into small A/B pocket (393-768) and the C pocket (768-869) (138). The small A/B pocket is sufficient for repression of transcription (14). Classically, RB has been studied in the context of cell cycle progression. RB mediates the Gl/S transition of the cell cycle by regulating the expression of genes required to enter into S phase. Additionally, RB is a phosphoprotein that is phosphorylated in a cell cycle dependent manner. The hypophosphorylated form of the protein, considered the active form of the protein, binds to and regulates target cellular factors. Phosphorylation by cylin D/cdk4 (30, 61) during late Gl or by Cyclin E/cdk2 (22, 23, 26, 87) during early Gl/S phase inactivates RB such that it can no longer bind to target factors. RB plays an essential role in development A powerful tool for dissecting the function of genes of interest is the technology that allows for targeted gene disruption (knock out) or ectopic gene expression (knock in) within a model organism. “Knock-out” or “knock-in” mice provide a wealth of valuable information about the functions of RB family members in an in vivo and physiologically relevant setting. Transgenic mice over-expressing RB show dwarfism by day 15 embryonic development (E15), indicating that RB regulation is required for proper development (4). However the lack of RB also adversely affects development. Gene targeted Rb-/- embryos die between E13.5 and E15.5 (16, 53, 65). Additionally, increased apoptosis is observed in the nervous system as early as E115 and is particularly evident in the hindbrain, spinal cord, and trigeminal and dorsal root ganglia. Ectopic mitoses are also observed especially in the hindbrain. Rb-/- embryos exhibit defective hematopoiesis, 14 manifested as an increased number of immature nucleated erythrocytes. Introduction of a RB transgene into knockout mice fully rescues developmental defects indicating the importance of this gene in normal development (4). Unlike human retinoblastoma patients, mouse RB chimeras develop pituitary gland tumors rather than retinoblastomas (75, 143). A similar phenotype is observed in Rb+/- mice, in that tumors develop in the brain and pituitary gland. These tumors exhibit loss of heterozygosity of the remaining wild type allele, demonstrating that RB is a tumor suppressor in mice as well as in humans. Although Rb+/- mice are cancer prone, they do not accurately reiterate the tumor spectrum observed in human retinoblastoma patients. This phenomenon may be explained by the fact that other RB family members could compensate for the loss of RB in the eye. Mice homozygous for p107 and p130 knock outs are viable, fertile and healthy (19, 66). p107-p130 double knock out mice experience neonatal lethality and most Rb+/- p107-/- mice are growth retarded, and increased mortality of these mice is observed in the first three weeks after birth. Although Rb+/-p107—/- pups that survive to adulthood do not show increased cancer predisposition compared to Rb+/— mice, they develop multiple dysplasic lesions of the retina. Unlike RB, p107 and p130 are not required for embryonic development, and p107+/-, p130+/—, p130-/-, p107-/- mice do not exhibit increased incidence of tumor development. Triple knockout embryonic fibroblasts have normal growth characteristics but impaired differentiation capacity (21, 110). Triple knockout mouse embryonic fibroblasts have a shorter cell cycle and can spontaneously immortalize. These cells are also resistant to G1 arrest following DNA damage, contact inhibition or serum starvation. 15 Targets for RB regulation Originally, genes proposed to be repressed by RB were known E2F responsive genes. It had been shown that RB could convert a positive acting E2F site to a negatively acting element in transient transfection experiments (137). Based on the data presented in this study, genes that had E2F binding sites or were shown genetically or biochemically to be dependent on E2F were postulated RB targets. Proposed target genes include S phase cyclins and cdks, EZF family members, other pocket protein family members, genes that function for DNA replication and nucleotide biosynthesis pathways (1, 56, 84, 86, 118, 136). Many studies have been performed to identify the true physiological targets of RB. RT- PCR and Northern blot analysis using mouse embryonic fibroblasts in which each individual RB family member is knocked out were used to study relative amounts of target gene expression in the absence of RB family members (52). This study showed a de-repression of cyclin E and p107 at the GO/Gl check point in the absence of RB. Additionally, b-myb, cch, E2F-1, thyrnidylate sythase (TS), ribonucleotide reductase M2 (RRMZ), cyclin A2 and dihydroxyfolate reductase (DHFR) were shown to be de- repressed at the 60/61 interface in the absence of both p107 and p130. Another group of researchers employed DNA micro-array chips to study global changes in gene expression under conditions in which a constitutively active RB was over-expressed in tissue culture cells (77). This study identified 341 targets that were reproducibly repressed at least 1.7 fold in multiple cell lines. These targets fell into four categories including: (1) DNA 16 '- II 2'. replication, (2) DNA repair, (3) chromatin structure and transcription, and (4) G2/M progression. While many of the genes identified in this study have previously been shown to be E2F dependent, several novel targets identified have not been previously linked to E2F. The micro-array analysis pinpointed several genes also identified previously by RT-PCR analysis (52) including cyclin A, RRM2, thymidine kinase, TS, cyclin E, cch and DHFR. Additionally, this screen also indicated that MCM6, MCM7, DNA polymerase 5, topoisomerase IIor, PCNA, Cdk2, cyclin BI, and HMGZ were repressed by the ectopically expressed RB. Together these two studies provide a pool of candidate genes regulated by RB family members. However, these studies do not distinguish between direct and indirect regulation of gene expression. In order to determine which genes RB family members directly target, it is important to study RB on the level of target gene promoter occupancy. Several groups have undertaken this question by using chromatin immunoprecipitation (ChIP) to analyze RB family member promoter occupancy. ChIP analysis provides a powerful tool for understanding direct gene targets for proteins of interest because it relies on in vivo endogenous proteins bound to genes in a physiologically relevant chromatin structure rather than on over-expression and reporter gene assays. The results of these studies however, are contradictory. Wells and co-workers (139) report that RB and RB family members occupy the TK promoter during G0; DHFR, cdc2, and cyclin B during GO and Gl/S; and b-myb in asynchronous cell populations (139). A second group, Takahashi and co-workers (124), were unable to detect RB at any promoter studied. They instead observed p130 at E2F1, cdc6, p107, b-myb, and cyclin A promoters during GO and 17 GO/Gl suggesting that p130 is the major player in cell cycle progression. One explanation for this discrepancy may be that these two groups worked in two different systems: mouse and human, respectively. RB family members may regulate cell cycle progression and growth control differently in these two species. Additionally, the experiments were performed from different cell types indicating that there may be tissue specific regulation by RB family members. Mechanisms for RB repression of RNA polymerase II transcribed genes The function of RB as a tumor suppressor is linked to its ability to regulate gene expression. Therefore, to fully understand the contribution of RB to cellular proliferation observed during carcinogenesis, it is important to determine the mechanisms that RB uses to regulate gene activity. Direct repression of transcription by RB implicates protein- protein interactions between RB and transcriptional activators belonging to the E2F family. E2F binds to enhancer regions of E2F responsive gene promoters to stimulate transcription by RNA polymerase II. By binding to E2F at gene promoters, RB can potentially directly block activation function of E2F by obscuring the trans-activation domain that facilitates pre-initiation complex assembly, or RB can recruit co-regulatory factors that alter chromatin structure surrounding these genes to repress transcription. Disruption of pre-initiation complex formation One critical step in the activation of transcription for most RNA polymerase II 18 transcribed genes is the establishment of a stable preinitiation complex (PIC) (93). Preinitiation complexes are thought to assemble in a step-wise fashion (93, 103, 104). First the TBP containing factor, TFIID, binds to TATA elements, nucleating preinitiation complex formation and recruiting additional basal factors. Next, TFIIA and TFIIB are recruited to the start site of transcription, followed by association of TFIIF in complex with RNA polymerase. Potentially, RB may repress transcription by interfering with one of these steps. Elegant studies using purified factors in an in vitro transcription assay show that RB does in fact repress transcription through disruption of pre—initiation complex formation (107). Specifically, RB inhibits the formation of the TFIID-TFIIA (DA) complex. However, if the DA complex is allowed to bind to DNA prior to the addition of RB, it becomes resistant to RB repression. Together, these results suggest that RB represses transcription by blocking pre-initiation complex formation thereby not allowing for RNA polymerase II association and subsequent gene expression. RB and histone modification: HDA Cs Covalent modification of histones and change chromatin structure is critical for efficient gene regulation (3, 55). Histone tails in nucleosomes at the promoters of active genes are generally acetylated on lysine residues. This acetylation neutralizes the positive charge of the lysine that binds tightly to the surrounding negatively charged DNA. The resulting unwrapping of the histone tails loosens the chromatin structure, allowing access to transcription factors and RNA polymerase. 19 a a K. M.‘ \‘u'n 1“ One potential mechanism that RB may use to repress transcription of target RNA polymerase II transcribed genes is the recruitment of histone deacetylase (HDAC) activities. HDACs are co-repressor complexes that enzymatically remove the acetyl groups from the lysines of histone tails. Several groups have shown that RB interacts with class I HDACs and that a deacetylase activity co-purifies with RB (6, 31, 73, 76, 99, 101, 149). Potentially, this HDAC activity could deacetylate histone tails at RB target genes, causing a net compaction of surrounding chromatin. This alteration of chromatin structure, resulting in a loss of accessibility to transcription factors and RNA polymerase, would then effectively repress transcription of the gene. Nonetheless, HDAC activity does not account for all of RB’s repression capability. The addition of HDAC inhibitors only partially reverses RB repression, suggesting that RB has HDAC independent as well as HDAC dependent repression mechanisms (6, 76). The HDAC independent repression may include mechanisms such as masking of the E2F trans-activation domain or preinitiation complex disruption. Additionally, there appears to be some promoter selectivity for RB repression through histone deacetylases (73). The true target of RB associated deacetylation activity is unclear. There is no direct in vivo evidence that the HDACs recruited by RB repress transcription by deacetylation of the histone tails in the promoters of target genes. While a corresponding lack of acetylation has been observed, it may be an indirect effect perhaps caused by failure to recruit the proper HAT complex. Another possibility is that the true target of RB- associated deacetylase activity is not histone tails but rather other acetylated factors 20 important for gene expression. In fact, one group studying RB repression of RNA polymerase I transcription demonstrated that acetylation of the required factor, UBF, was important for active transcription of rRNA genes (9). Moreover, this group also showed that deacetylation of UBF by RB associated HDACs results in loss of gene expression, suggesting that HDACs may have substrates in addition to histone tails. Altogether, studies to date indicate that HDAC activity is important for the regulation of select promoters even if the mechanism of repression is still as yet unclear. RB and chromatin remodeling: S WI/SNF Chromatin remodeling is another fundamental process in the regulation of gene expression. Many chromatin-remodeling complexes reposition nucleosomes that create repressive barriers within gene promoters. Repositioning these nucleosomes alters the local chromatin structure and increases promoter access to factors necessary for active transcription of the gene (85). Chromatin remodeling is also another key process that RB may influence in order to repress transcription. Several groups have demonstrated that RB can interact with mammalian orthologues of the Drosophila Brahma protein: hBRM and Brg-l (120, 122, 128, 149). These paralogous human proteins are the large ATPase subunits of the human SWI/SNF complexes. SWI/SNF is an ATP-dependent chromatin remodeling complex that is required for the activation of many RNA polymerase II transcribed genes (94). SWI/SNF appears to be required for RB regulation of cell cycle progression (149). In contrast to the activation activity normally associated with Chromatin remodeling, when SWI/SNF is recruited to target genes by RB, the overall effect is negative, perhaps by repositioning nucleosomes in a fashion that would obscure 21 the promoter. Interestingly, RB may recruit both SWI/SNF and HDACs to the same promoter to repress transcription through a combination of histone modification and chromatin remodeling. RB appears to regulate exit from G] phase of the cell cycle through the recruitment of both HDACs and SWI/SNF, whereas exit from S phase is regulated through RB recruitment of SWI/SNF alone. Again these results suggest multiple mechanisms of RB repression that may be dependent on promoter selectivity. RB and co-repressor complexes: Polycomb Additionally, RB may regulate gene expression and consequently cell cycle through recruitment of Polycomb (PcG) complexes. Members of the Polycomb family have roles in the negative regulation of Hox genes during development (98, 115) and also serve as critical regulators of cell cycle progression (54, 70). Polycomb complexes bind to Polycomb response elements (PRE) in target genes to establish a silenced chromatin state (98) and also contribute to transcriptional memory such that once silenced in a cell, a gene will also be silenced in progeny cells (8). RB has been shown to associate with a PcG complex containing HPC2, Ringl, and Bmi- 1 in an interaction mediated by CtBP (20). Specifically, this HDAC-independent mechanism appears to arrest cells in the GZ phase of the cell cycle by repressing the expression of cyclin A and cdc2. This PcG complex also associates with the cyclin A promoter under conditions of cell cycle arrest. The authors of this study suggest that this mechanism of repression may occur under conditions of irreversible grth arrest 22 whereas HDAC-dependent RB repression may occur in the case of reversible cell cycle arrest. RB and histone methylation: HP] — Su Var39 RB may also regulate gene expression by recruitment of HPl and SUV39H1 (88, 131). SUV39H1 specifically methylates lysine 9 on histone H3 tails. (91, 102). The resulting methylation on K9 of H3 tails induces the formation of a high affinity-binding site on the chromatin for proteins of the heterochromatin protein 1 (HPl) family, which are involved in heterochromatin silencing (29). RB associates with SUV39H1 in co-immunoprecipitation experiments and directly interacts with HP] (88). A histone methyltransferase activity also co-purifies with RB. Furthermore, HPI and RB are both recruited to a methylated H3 peptide, suggesting that HP] can recognize RB and methylated histone tails simultaneously. In vivo chromatin IP data demonstrates that HPl is present at the cyclin E promoter in RB+/+ cells and that the H3 tails at these promoters are methylated, whereas HPl does not associate with this promoter in the absence of RB and the H3 tails exhibit lower methylation. Collectively the results suggest that RB recruits the SUV39H1 enzyme to target promoters where it methylates K9H3, providing a binding site for HP]. Since methylation cannot occur at a lysine residue that is acetylated (102), the deacetylase activity associated with RB may be required as a preceding step to SUV39H1 mediated methylation. Potentially the role for HPl in this repression is not to promote the formation of heterochromatin but rather to mask the methylated lysine 9 in histone H3 tails such that it cannot be de-methylated and 23 subsequently acetylated. These results indicate that the SUV39Hl-HP1 complex is not only involved in heterochromatic silencing but also has a role in repression of euchromatic genes by RB and perhaps other co-repressor proteins. RB and DNA methylation: DMN T 1 DNA methylation at CpG dinucleotides is essential for embryonic development and is catalyzed by three DNA methyltransferases, DMNTl, DMNT3a and DMNT3b, which have differential capacities for maintenance and de novo methylation (71, 92). During chromatographic purification of the DMNTl methyltransferase activity, RB was shown to co-purify in a complex with DMNTI (106) that also includes E2F and HDAC]. The methyltransferase activity that is isolated by immunoprecipitation for RB and DNMTl appears to work co—operatively with RB and HDACl to repress transcription. Potentially, the DNA binding domains of the associated E2F proteins provide the necessary specificity to recruit the repression activities to target genes. The results of this study establish a link between DNA methylation, sequence specific DNA binding activity, histone deacetylation as well as 3 grth regulatory pathway this is disrupted in the majority of cancer cells. RB regulation of RNA polymerase III In addition to regulating genes expressed by RNA polymerase 11, RB also regulates genes transcribed by RNA polymerase III. RB globally represses RNA polymerase III transcripts both in vivo and in vitro, including RNA polymerase III transcribed snRNA genes (15, 45, 62, 116, 123, 142). Interestingly, nuclear run-on assays in RB-/- mouse 24 embryonic fibroblasts suggest that while global RNAP III activity is elevated in these cells, RNAP II transcription is largely unaffected (142). Regulation of RNA polymerase III transcribed genes may be important for controlling the growth potential of the cell. Genes transcribed by RNA polymerase III encode small RNAs that act as metabolic machinery and thus contribute to the biosynthetic capacity of the cell. Thus, cells lacking RB would be expected to have increased proliferation potential as a result of increased available biosynthetic machinery, and this potential may contribute to unregulated growth during tumor formation. How RB regulates RNA polymerase III activity in the cell is not clear. RNA polymerase III transcriptional activity is under cell cycle control, with higher levels observed in the late G l, S and G2 phases of the cell cycle than in GO and early G1. The increase in RNA p01ytnerase III activity correlates with an increase in phosphorylated RB (inactive form) during the G1 phase of the cell cycle, suggesting a link between RB regulation and RNA pOIanerase III activity (116, 141). Over-expression of RB during transient transfection assays in RB-/- SaOS-2 osteosarcoma cells results in the repression of tRNA genes as well as Ad2 VAI genes. One proposed mechanism for RB repression of SS rRNA and tRNA transcription by RNA polymerase III is the disruption of interactions between basal transcription factors I'tl‘luired for pre-initiation complex formation. RB has been shown to associate with both TFIIIB (63) and TFIIIC2 (15). Specifically, RB associates with the Brfl and TBP Sllbunits of TFIIIB in co-immunoprecipitation experiments and co-purifies with TFIIIB 25 biochernically. RB can also interact with TFIIIC2 in GST pull-down experiments performed in HeLa cell nuclear extracts (15). RB however does not appear to interact with TFIIIA (63) or RNA polymerase III. Sutcliffe and collaborators (123) demonstrated that RB interaction with TFIIIB precludes TFIIIB interaction with TFIIIC. Since TFIIIB can no longer associate with promoters through interactions with TFIIIC, RNA polymerase III is not recruited to the start site of transcription, effectively abolishing gene expression. Less is known about how RB regulates the expression of class 3 RNA polymerase III transcribed genes. RB represses transcription of the human U6 gene by RNA polymerase 111 both in vivo and in vitro. However, U6 snRNA genes differ significantly from other genes transcribed by RNA polymerase III. Importantly, these genes contain snRNA- specific promoter elements and do not employ the same TFIIIB and TF 111C complexes implicated in repression of tRNA genes. One suggestion is that the factors recognizing these sequence elements are important for regulating U6 transcription specifically with respect to RB. Because the promoter elements are simple and well defined and the factor requirements are known, RNA polymerase 111 genes provide an excellent model system for understanding regulation of eukaryotic gene expression. We therefore wished to exploit this model system to better understand how the Retinoblastoma tumor suppressor protein functions to repress transcription. 26 References 10. 11. Adnane, J., Z. Shao, and P. D. Robbins. 1995. The retinoblastoma susceptibility gene product represses transcription when directly bound to the promoter. J Biol Chem 270:8837-43. Bai, L., Z. Wang, J. B. Yoon, and R. G. Roeder. 1996. Cloning and characterization of the beta subunit of human proximal sequence element-binding transcription factor and its involvement in transcription of small nuclear RNA genes by RNA polymerases II and 111. Mol Cell Biol 16:5419-26. Berger, S. L. 2002. Histone modifications in transcriptional regulation. Curr Opin Genet Dev 12: 142-8. Bignon, Y. J., Y. Chen, C. Y. Chang, D. J. Riley, J. J. Windle, P. L. Mellon, and W. H. Lee. 1993. Expression of a retinoblastoma transgene results in dwarf mice. Genes Dev 7 :1654-62. Bogenhagen, D. F. 1985. The intragenic control region of the Xenopus 5 S RNA gene contains two factor A binding domains that must be aligned properly for efficient transcription initiation. J Biol Chem 260:6466-71. Brehm, A., E. A. Miska, D. J. McCance, J. L. Reid, A. J. Bannister, and T. Kouzarides. 1998. Retinoblastoma protein recruits histone deacetylase to repress transcription. Nature 391 :597-601 . Burnol, A. F., F. Margottin, P. Schultz, M. C. Marsolier, P. Oudet, and A. Sentenac. 1993. Basal promoter and enhancer element of yeast U6 snRNA gene. J Mol Biol 233:644-58. Busturia, A., C. D. Wightman, and S. Sakonju. 1997. A silencer is required for maintenance of transcriptional repression throughout Drosophila development. Development 124:4343-50. Cavanaugh, A. H., W. M. Hempel, L. J. Taylor, V. Rogalsky, G. Todorov, and L. I. Rothblum. 1995. Activity of RNA polymerase I transcription factor UBF blocked by Rb gene product. Nature 374: 177-80. Challice, J. M., and J. Segall. 1989. Transcription of the 5 S rRNA gene of Saccharomyces cerevisiae requires a promoter element at +1 and a 14-base pair internal control region. J Biol Chem 264:20060-7. Chedin, S., M. L. Ferri, G. Peyroche, J. C. Andrau, S. Jourdain, O. Lefebvre, M. Werner, C. Carles, and A. Sentenac. 1998. The yeast RNA polymerase III transcription machinery: a paradigm for eukaryotic gene activation. Cold Spring Harb Symp Quant Biol 63:381-9. 27 12. l3. 14. 15. l6. 17. 18. 19. 20. 21. 22. 23. Chen, P. L., D. J. Riley, Y. Chen, and W. H. Lee. 1996. Retinoblastoma protein positively regulates terminal adipocyte differentiation through direct interaction with C/EBPs. Genes Dev 10:2794-804. Chong, 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 276:20727-20734. Chow, K. N., and D. C. Dean. 1996. Domains A and B in the Rb pocket interact to form a transcriptional repressor motif. Mol Cell Biol 16:4862-8. Chu, W. M., Z. Wang, R. G. Roeder, and C. W. Schnrid. 1997. RNA polymerase III transcription repressed by Rb through its interactions with TFIIIB and TFIIIC2. J Biol Chem 272:14755-61. Clarke, A. R., E. R. Maandag, M. van Roon, N. M. van der Lugt, M. van der Valk, M. L. Hooper, A. Berns, and H. te Riele. 1992. Requirement for a functional Rb-l gene in murine development. Nature 359:328-30. Classon, M., and E. Harlow. 2002. The retinoblastoma tumour suppressor in development and cancer. Nat Rev Cancer 2:910-7. Clemens, K. R., X. Liao, V. Wolf, P. E. Wright, and J. M. Gottesfeld. 1992. Definition of the binding sites of individual zinc fingers in the transcription factor IIIA-SS RNA gene complex. Proc Natl Acad Sci U S A 89:10822-6. Cobrinik, D., M. H. Lee, G. Harmon, G. Mulligan, R. T. Bronson, N. Dyson, E. Harlow, D. Beach, R. A. Weinberg, and T. Jacks. 1996. Shared role of the pRB-related p130 and p107 proteins in limb development. Genes Dev 10: 1633- 44. Dahiya, A., S. Wong, 8. Gonzalo, M. Gavin, and D. C. Dean. 2001. Linking the Rb and polycomb pathways. Mol Cell 8:557-69. Dannenberg, J. H., A. van Rossum, L. Schuijff, and H. te Riele. 2000. Ablation of the retinoblastoma gene family deregulates G(1) control causing irnmortalization and increased cell turnover under growth-restricting conditions. Genes Dev 14:3051-64. DeGregori, J., T. Kowalik, and J. R. Nevins. 1995. Cellular targets for activation by the E2F1 transcription factor include DNA synthesis- and 01/8- regulatory genes. Mol Cell Biol 15:4215-24. DeGregori, J., G. Leone, K. Ohtani, A. Miron, and J. R. Nevins. 1995. E2F-1 accumulation bypasses a G1 arrest resulting from the inhibition of G1 cyclin- dependent kinase activity. Genes Dev 9:2873-87. 28 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. Dunaief, J. L., B. E. Strober, S. Guha, P. A. Khavari, K. Alin, J. Luban, M. Begemann, G. R. Crabtree, and S. P. Goff. 1994. The retinoblastoma protein and BRGl form a complex and cooperate to induce cell cycle arrest. Cell 79:119- 30. Dynlacht, B. D. 1997. Regulation of transcription by proteins that control the cell cycle. Nature 389: 149-52. Dynlacht, B. D., O. Flores, J. A. Lees, and E. Harlow. 1994. Differential regulation of E2F transactivation by cyclin/cdk2 complexes. Genes Dev 8: 1772- 86. Dyson, N ., K. Buchkovich, P. Whyte, and E. Harlow. 1989. Cellular proteins that are targetted by DNA tumor viruses for transformation. Princess Takamatsu Symp 20:191-8. Dyson, N ., P. M. Howley, K. Munger, and E. Harlow. 1989. The human papilloma virus-16 E7 oncoprotein is able to bind to the retinoblastoma gene product. Science 243:934-7. Eissenberg, J. C., and S. C. Elgin. 2000. The HPl protein family: getting a grip on chromatin. Curr Opin Genet Dev 10:204-10. Ewen, M. E., H. K. Sluss, C. J. Sherr, H. Matsushime, J. Kato, and D. M. Livingston. 1993. Functional interactions of the retinoblastoma protein with mammalian D-type cyclins. Cell 73:487-97. Ferreira, R., L. Magnaghi-Jaulin, P. Robin, A. Harel—Bellan, and D. Trouche. 1998. The three members of the pocket proteins family share the ability to repress E2F activity through recruitment of a histone deacetylase. Proc Natl Acad Sci U S A 95:10493-8. Ford, E., M. Strubin, and N. Hernandez. 1998. The Oct-l 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. Foster, M. P., D. S. Wuttke, I. Radhakrishnan, D. A. Case, J. M. Gottesfeld, and P. E. Wright. 1997. Domain packing and dynamics in the DNA complex of the N-terminal zinc fingers of TFIIIA. Nat Struct Biol 4:605-8. Fung, Y. K., A. L. Murphree, A. T'Ang, J. Qian, S. H. Hinrichs, and W. F. Benedict. 1987. Structural evidence for the authenticity of the human retinoblastoma gene. Science 236: 1657-61 . Galli, G., H. Hofstetter, and M. L. Birnstiel. 1981. Two conserved sequence blocks within eukaryotic tRNA genes are major promoter elements. Nature 294:626-31. 29 36. 37. 38. 39. 40. 41. 42. 43. 45. 46. 47. Geiduschek, E. P., and G. A. Kassavetis. 2001. The RNA polymerase III transcription apparatus. J Mol Biol 310:1-26. Ginsberg, A. M., B. 0. King, and R. G. Roeder. 1984. Xenopus SS gene transcription factor, TFIIIA: characterization of a cDNA clone and measurement of RNA levels throughout development. Cell 39:479-89. Gu, W., J. W. Schneider, G. Condorelli, S. Kaushal, V. Mahdavi, and B. Nadal-Ginard. 1993. Interaction of myogenic factors and the retinoblastoma protein mediates muscle cell commitment and differentiation. Cell 72:309-24. Harrington, E. A., J. L. Bruce, E. Harlow, and N. Dyson. 1998. pRB plays an essential role in cell cycle arrest induced by DNA damage. Proc Natl Acad Sci U S A 95:11945-50. Hatakeyama, M., and R. A. Weinberg. 1995. The role of RB in cell cycle control. Prog Cell Cycle Res 1:9-19. Henry, R. W., B. Ma, C. L. Sadowski, R. Kobayashi, and N. Hernandez. 1996. Cloning and characterization of SNAPSO, 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 firnctional core promoter complex (SNAPc) shared by RNA polymerases II and III. Genes Dev 112:2664-72. 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. Hinkley, C. S., H. A. Hirsch, L. Cu, B. lLaMere, 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. Hossenlopp, P., J. Sumegi, and P. Chambon. 1978. Transcription in vitro of adenovirus-2 DNA by RNA polymerases class C purified from uninfected and adenovirus-infected HeLa cells. Eur J Biochem 90:615-31. Howe, J. G., and M. D. Shu. 1989. Epstein-Barr virus small RNA (EBER) genes: unique transcription units that combine RNA polymerase 11 and III promoter elements. Cell 57:825-34. 30 48. 49. 50. 51. 52. 53. 54. 55. S6. 57. 58. Howe, J. G., and M. D. Shu. 1993. Upstream basal promoter element important for exclusive RNA polymerase III transcription of the EBER 2 gene. Mol Cell Biol 13:2655-65. Hsieh, J. K., F. S. Chan, D. J. O'Connor, S. Mittnacht, S. Zhong, and X. Lu. 1999. RB regulates the stability and the apoptotic function of p53 via MDM2. Mol Cell 3:181-93. Hsieh, Y. J., T. K. Kundu, Z. Wang, R. Kovelman, and R. G. Roeder. 1999. The TFIIIC90 subunit of TFIIIC interacts with multiple components of the RNA polymerase III machinery and contains a histone-specific acetyltransferase activity. Mol Cell Biol 19:7697-704. Hu, P., S. Wu, Y. Sun, C. C. Yuan, R. Kobayashi, M. P. Myers, and N. Hernandez. 2002. Characterization of human RNA polymerase III identifies orthologues for Saccharomyces cerevisiae RNA polymerase III subunits. Mol Cell Biol 22:8044-55. Hurford, R. K., Jr., D. Cobrinik, M. H. Lee, and N. Dyson. 1997. pRB and p107/p130 are required for the regulated expression of different sets of E2F responsive genes. Genes Dev 11: 1447-63. Jacks, T., A. Fazeli, E. M. Schnritt, R. T. Bronson, M. A. Goodell, and R. A. Weinberg. 1992. Effects of an Rb mutation in the mouse. Nature 359:295-300. Jacobs, J. J., K. Kieboonr, S. Marino, R. A. DePinho, and M. van Lohuizen. 1999. The oncogene and Polycomb-group gene bmi-l regulates cell proliferation and senescence through the ink4a locus. Nature 397: 164-8. Jenuwein, T., and C. D. Allis. 2001. Translating the histone code. Science 293: 1074-80. Johnson, D. G., K. Ohtani, and J. R. Nevins. 1994. Autoregulatory control of E2F1 expression in response to positive and negative regulators of cell cycle progression. Genes Dev 8: 15 14-25. Kassavetis, G. A., B. Bartholomew, J. A. Blanco, T. E. Johnson, and E. P. Geiduschek. 1991. Two essential components of the Saccharomyces cerevisiae transcription factor TFIIIB: transcription and DNA-binding properties. Proc Natl Acad Sci U S A 88:7308-12. Kassavetis, G. A., B. Bartholomew, J. A. Blanco, T. E. Johnson, and E. P. Geiduschek. 1991. Two essential components of the Saccharomyces cerevisiae transcription factor TFIIIB: transcription and DNA-binding properties. Proc Natl Acad Sci U S A 88:7308-12. 31 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. Kassavetis, G. A., B. R. Braun, L. H. Nguyen, and E. P. Geiduschek. 1990. S. cerevisiae TFIIIB is the transcription initiation factor proper of RNA polymerase III, while TFIIIA and TFIIIC are assembly factors. Cell 60:235-45. Kassavetis, G. A., D. L. Riggs, R. Negri, L. H. Nguyen, and E. P. Geiduschek. 1989. Transcription factor IIIB generates extended DNA interactions in RNA polymerase III transcription complexes on tRNA genes. Mol Cell Biol 9:2551-66. Kato, J., H. Matsushime, S. W. Hiebert, M. E. Ewen, and C. J. Sherr. 1993. Direct binding of cyclin D to the retinoblastoma gene product (pr) and pr phosphorylation by the cyclin D-dependent kinase CDK4. Genes Dev 7 :331-42. Larminie, C. G., H. M. Alzuherri, C. A. Cairns, A. McLees, and R. J. White. 1998. Transcription by RNA polymerases I and III: a potential link between cell growth, protein synthesis and the retinoblastoma protein. J Mol Med 76:94-103. 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. Lassar, A. B., P. L. Martin, and R. G. Roeder. 1983. Transcription of class 111 genes: formation of preinitiation complexes. Science 222:740-8. Lee, E. Y., C. Y. Chang, N. Hu, Y. C. Wang, C. C. Lai, K. Herrup, W. H. Lee, and A. Bradley. 1992. Mice deficient for Rb are nonviable and show defects in neurogenesis and haematopoiesis. Nature 359:288-94. Lee, M. H., B. 0. Williams, G. Mulligan, S. Mukai, R. T. Bronson, N. Dyson, E. Harlow, and T. Jacks. 1996. Targeted disruption of p107: functional overlap between p107 and Rb. Genes Dev 10:1621-32. Lee, W. H., R. Bookstein, F. Hong, L. J. Young, J. Y. Shew, and E. Y. Lee. 1987. Human retinoblastoma susceptibility gene: cloning, identification, and sequence. Science 235: 1394-9. Lee, Y., and R. N. N azar. 1997. Ribosomal SS rRNA maturation in Saccharomyces cerevisiae. J Biol Chem 272: 15206-12. Lescure, A., G. Tebb, I. W. Mattaj, A. Krol, and P. Carbon. 1992. A factor with Spl DNA-binding specificity stimulates Xenopus U6 snRNA in vivo transcription by RNA polymerase 111. J Mol Biol 228:387-94. Lessard, J., A. Schumaeher, U. Thorsteinsdottir, M. van Lohuizen, T. Magnuson, and G. Sauvageau. 1999. Functional antagonism of the Polycomb- Group genes eed and Bmil in hemopoietic cell proliferation. Genes Dev 13:2691- 703. 32 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. Li, E., T. H. Bestor, and R. J aenisch. 1992. Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69:915-26. Lobo, S. M., M. Tanaka, M. L. Sullivan, and N. Hernandez. 1992. A TBP complex essential for transcription from TATA-less but not TATA-containing RNA polymerase III promoters is part of the TFIIIB fraction. Cell 71:1029-40. Luo, R. X., A. A. Postigo, and D. C. Dean. 1998. Rb interacts with histone deacetylase to repress transcription. Cell 92:463-73. 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. Maandag, E. C., M. van der Valk, M. Vlaar, C. Feltkamp, J. O'Brien, M. van Roon, N. van der Lugt, A. Berns, and H. te Riele. 1994. Developmental rescue of an embryonic-lethal mutation in the retinoblastoma gene in chimeric mice. EMBO J 13:4260-8. Magnaghi-Jaulin, L., R. Groisman, I. Naguibneva, P. Robin, S. Lorain, J. P. Le Villain, F. Troalen, D. Trouche, and A. Harel-Bellan. 1998. Retinoblastoma protein represses transcription by recruiting a histone deacetylase. Nature 391:601-5. Markey, M. P., S. P. Angus, M. W. Strobeck, S. L. Williams, R. W. Gunawardena, B. J. Aronow, and E. S. Knudsen. 2002. Unbiased analysis of RB-mediated transcriptional repression identifies novel targets and distinctions from E2F action. Cancer Res 62:6587-97. Marzouki, N., S. Camier, A. Ruet, A. Moenne, and A. Sentenac. 1986. Selective proteolysis defines two DNA binding domains in yeast transcription factor tau. Nature 323: 1 76-8. McCulloch, V., P. Hardin, W. Peng, J. M. Ruppert, and S. M. Lobo-Ruppert. 2000. Alternatively spliced hBRF variants function at different RNA polymerase III promoters. EMBO J 19:4134-43. Mihara, K., X. R. Cao, A. Yen, S. Chandler, B. Driscoll, A. L. Murphree, A. T'Ang, and Y. K. Fung. 1989. Cell cycle-dependent regulation of phosphorylation of the human retinoblastoma gene product. Science 246: 1300-3. Mital, R., R. Kobayashi, and N. Hernandez. 1996. RNA polymerase III transcription from the human U6 and adenovirus type 2 VAI promoters has different requirements for human BRF, a subunit of human TFIIIB. Mol Cell Biol 16:7031-42. Mittal, V., and N. Hernandez. 1997. Role for the arnino-terminal region of human TBP in U6 snRNA transcription. Science 275:1136-40. 33 83. 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. 94. 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. Muller, H., J. Lukas, A. Schneider, P. Warthoe, J. Bartek, M. Eilers, and M. Strauss. 1994. Cyclin D1 expression is regulated by the retinoblastoma protein. Proc Natl Acad Sci U S A 91:2945-9. N arlikar, G. J., H. Y. Fan, and R. E. Kingston. 2002. Cooperation between complexes that regulate chromatin structure and transcription. Cell 108:475-87. Neuman, E., E. K. Flemington, W. R. Sellers, and W. G. Kaelin, Jr. 1995. Transcription of the E2F-l gene is rendered cell cycle dependent by E2F DNA- binding sites within its promoter. Mol Cell Biol 15:4660. Nevins, J. R., G. Leone, J. DeGregori, and L. Jakoi. 1997. Role of the Rb/E2F pathway in cell grth control. J Cell Physiol 173:233-6. Nielsen, S. J., R. Schneider, U. M. Bauer, A. J. Bannister, A. Morrison, D. O'Carroll, R. F irestein, M. Cleary, T. Jenuwein, R. E. Herrera, and T. Kouzarides. 2001. Rb targets histone H3 methylation and HP] to promoters. Nature 412:561-5. Nolte, R. T., R. M. Conlin, S. C. Harrison, and R. S. Brown. 1998. Differing roles for zinc fingers in DNA recognition: structure of a six-finger transcription factor IIIA complex. Proc Natl Acad Sci U S A 95:2938-43. Novitch, B. G., G. J. Mulligan, T. Jacks, and A. B. Lassar. 1996. Skeletal muscle cells lacking the retinoblastoma protein display defects in muscle gene expression and accumulate in S and G2 phases of the cell cycle. J Cell Biol 135:441-56. O'Carroll, D., H. Scherthan, A. H. Peters, S. Opravil, A. R. Haynes, G. Laible, S. Rea, M. Schnrid, A. Lebersorger, M. Jerratsch, L. Sattler, M. G. Mattei, P. Denny, S. D. Brown, D. Schweizer, and T. Jenuwein. 2000. Isolation and characterization of Suv3 9h2, a second histone H3 methyltransferase gene that displays testis-specific expression. Mol Cell Biol 20:9423-33. Okano, M., D. W. Bell, D. A. Haber, and E. Li. 1999. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99:247-57. Orphanides, G., T. Lagrange, and D. Reinberg. 1996. The general transcription factors of RNA polymerase II. Genes Dev 10:2657-83. Peterson, C. L., and J. L. Workman. 2000. Promoter targeting and chromatin remodeling by the SWI/SNF complex. Curr Opin Genet Dev 10: 187-92. 34 95. 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. Pieler, T., B. Appel, S. L. Oei, H. Mentzel, and V. A. Erdmann. 1985. Point mutational analysis of the Xenopus laevis 5S gene promoter. EMBO J 4: 1 847-53. Pieler, T., J. Hamnr, and R. G. Roeder. 1987. The SS gene internal control region is composed of three distinct sequence elements, organized as two functional domains with variable spacing. Cell 48:91-100. Pieler, T., S. L. Oei, J. Hamm, U. Engelke, and V. A. Erdmann. 1985. Functional domains of the Xenopus laevis 5S gene promoter. EMBO J 4:3751-6. Pirrotta, V. 1998. Polycombing the genome: PcG, trxG, and chromatin silencing. Cell 93:333-6. Puri, P. L., S. Iezzi, P. Stiegler, T. T. Chen, R. L. Schiltz, G. E. Muscat, A. Giordano, L. Kedes, J. Y. Wang, and V. Sartorelli. 2001. Class I histone deacetylases sequentially interact with MyoD and pr during skeletal myogenesis. Mol Cell 8:885-97. Purrello, M., C. Di Pietro, A. Rapisarda, V. Amico, V. Giunta, H. Engel, S. Stevens, Y. Hsieh, M. Teichman, Z. Wang, G. Sichel, R. Roeder, and K. H. Grzeschik. 2001 . Genes for human general transcription initiation factors TFIIIB, TFIIIB-associated proteins, TFIIIC2 and PTF/SNAPC: functional and positional candidates for tumour predisposition or inherited genetic diseases? Oncogene 20:4877-83. Rayman, J. B., Y. Takahashi, V. B. Indjeian, J. H. Dannenberg, S. Catchpole, R. J. Watson, H. te Riele, and B. D. Dynlacht. 2002. E2F mediates cell cycle- dependent transcriptional repression in vivo by recruitment of an HDACl/mSin3B corepressor complex. Genes Dev 16:933-47. Rea, S., F. Eisenhaber, D. O'Carroll, B. D. Strahl, Z. W. Sun, M. Schmid, S. Opravil, K. Mechtler, C. P. Ponting, C. D. Allis, and T. Jenuwein. 2000. Regulation of chromatin structure by site-specific histone H3 methyltransferases. Nature 406:593-9. Reinberg, D., M. Horikoshi, and R. G. Roeder. 1987. Factors involved in specific transcription in mammalian RNA polymerase 11. Functional analysis of initiation factors IIA and IID and identification of a new factor operating at sequences downstream of the initiation site. .1 Biol Chem 262:3322-30. Reinberg, D., and R. G. Roeder. 1987. Factors involved in specific transcription by mammalian RNA polymerase II. Purification and functional analysis of initiation factors [18 and 11E. J Biol Chem 262:3310-21. Ren, B., H. Cam, Y. Takahashi, T. Volkert, J. Terragni, R. A. Young, and B. D. Dynlacht. 2002. E2F integrates cell cycle progression with DNA repair, replication, and G(2)/M checkpoints. Genes Dev 16:245-56. 35 106. 107. 108. 109. 110. 111. 112. 113. 114. 115. 116. 117. Robertson, K. D., S. Ait-Si-Ali, T. Yokochi, P. A. Wade, P. L. Jones, and A. P. Wolffe. 2000. DNMTl forms a complex with Rb, E2F1 and HDAC] and represses transcription from E2F-responsive promoters. Nat Genet 25:338-42. Ross, J. F., X. Liu, and B. D. Dynlacht. 1999. Mechanism of transcriptional repression of E2F by the retinoblastoma tumor suppressor protein. Mol Cell 3: 195-205. 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 snRNA 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 snRNA promoters through the PSE. Genes Dev 7:1535-48. Sage, J., G. J. Mulligan, L. D. Attardi, A. Miller, S. Chen, B. Williams, E. Theodorou, and T. Jacks. 2000. Targeted disruption of the three Rb-related genes leads to loss of G(l) control and immortalization. Genes Dev 14:3037-50. Sanchez, 1., and B. D. Dynlacht. 1996. Transcriptional control of the cell cycle. Curr Opin Cell Biol 8:318-24. 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 T ATA-less promoters. Genes Dev 14:2650-63. Schumaeher, A., and T. Magnuson. 1997. Murine Polycomb- and trithorax- group genes regulate homeotic pathways and beyond. Trends Genet 13:167-70. Scott, P. H., C. A. Cairns, J. E. Sutcliffe, H. M. Alzuherri, A. McLees, A. G. Winter, and R. J. White. 2001. Regulation of RNA polymerase III transcription during cell cycle entry. J Biol Chem 276: 1005-14. Segall, J., T. Matsui, and R. G. Roeder. 1980. Multiple factors are required for the accurate transcription of purified genes by RNA polymerase 111. J Biol Chem 255:] 1986-91. 36 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. Sellers, W. R., B. G. Novitch, S. Miyake, A. Heith, G. A. Otterson, F. J. Kaye, A. B. Lassar, and W. G. Kaelin, Jr. 1998. Stable binding to E2F is not required for the retinoblastoma protein to activate transcription, promote differentiation, and suppress tumor cell growth. Genes Dev 12:95-106. Sharp, 8., D. DeFranco, M. Silberklang, H. A. Hosbach, T. Schmidt, E. Kubli, J. P. Gergen, P. C. Wensink, and D. 8011. 1981. The initiator tRNA genes of Drosophila melanogaster: evidence for a tRNA pseudogene. Nucleic Acids Res 9:5867-82. Strobeck, M. W., K. E. Knudsen, A. F. Fribourg, M. F. DeCristofaro, B. E. Weissman, A. N. Imbalzano, and E. S. Knudsen. 2000. BRG-l is required for RB-mediated cell cycle arrest. Proc Natl Acad Sci U S A 97 :7748-53. Stunkel, W., I. Kober, and K. H. Seifart. 1997. A nucleosome positioned in the distal promoter region activates transcription of the human U6 gene. Mol Cell Biol 17:4397-405. Sudarsanam, P., and F. Winston. 2000. The Swi/Snf family nucleosome- remodeling complexes and transcriptional control. Trends Genet 16:345-51. Sutcliffe, J. E., T. R. Brown, S. J. Allison, P. H. Scott, and R. J. White. 2000. Retinoblastoma protein disrupts interactions required for RNA polymerase III transcription. Mol Cell Biol 20:9192-9202. 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. Teichmann, M., Z. Wang, and R. G. Roeder. 2000. A stable complex of a novel transcription factor IIB- related factor, human TF 111B50, and associated proteins mediate selective transcription by RNA polymerase III of genes with upstream promoter elements. Proc Natl Acad Sci U S A 97: 14200-5. Thomas, D. M., S. A. Carry, D. M. Piscopo, J. S. Lee, W. F. Wang, W. C. Forrester, and P. W. Hinds. 2001. The retinoblastoma protein acts as a transcriptional coactivator required for osteogenic differentiation. Mol Cell 8:303- 16. Trivedi, A., L. S. Young, C. Ouyang, D. L. Johnson, and K. U. Sprague. 1999. A TATA element is required for tRNA promoter activity and confers TATA- binding protein responsiveness in Drosophila Schneider-2 cells. J Biol Chem 274:1 1369-75. Trouche, D., C. Le Chalony, C. Muchardt, M. Yaniv, and T. Kouzarides. 1997. RB and hbrrn cooperate to repress the activation functions of E2F1. Proc Natl Acad Sci U S A 94:11268-73. 37 129. 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. Van Dyke, M. W., and R. G. Roeder. 1987. Multiple proteins bind to VA RNA genes of adenovirus type 2. Mol Cell Biol 7:1021-31. van Zon, A., M. H. Mossink, M. Schoester, G. L. Scheffer, R. J. Scheper, P. Sonneveld, and E. A. Wiemer. 2001. Multiple human vault RNAs. Expression and association with the vault complex. J Biol Chem 276:37715-21. Vandel, L., E. Nicolas, O. Vaute, R. F erreira, S. Ait-Si-Ali, and D. Trouche. 2001. Transcriptional repression by the retinoblastoma protein through the recruitment of a histone methyltransferase. Mol Cell Biol 21 :6484-94. Vilalta, A., V. A. Kickhoefer, L. H. Rome, and D. L. Johnson. 1994. The rat vault RNA gene contains a unique RNA polymerase III promoter composed of both external and internal elements that function synergistically. J Biol Chem 269:29752-9. Wang, Z., and R. G. Roeder. 1995. Structure and function of a human transcription factor TFIIIB subunit that is evolutionarily conserved and contains both TFIIB- and high- mobility-group protein 2-related domains. Proc Natl Acad Sci U S A 92:7026-30. Wang, Z., and R. G. Roeder. 1997. Three human RNA polymerase III-specific subunits form a subcomplex with a selective function in specific transcription initiation. Genes Dev 11:1315-26. Weinberg, R. A. 1995. The retinoblastoma protein and cell cycle control. Cell 81:323-30. Weintraub, S. J., K. N. Chow, R. X. Luo, S. H. Zhang, S. He, and D. C. Dean. 1995. Mechanism of active transcriptional repression by the retinoblastoma protein. Nature 375:812-5. Weintraub, S. J., C. A. Prater, and D. C. Dean. 1992. Retinoblastoma protein switches the E2F site from positive to negative element. Nature 358:259-61. Welch, P. J., and J. Y. Wang. 1993. A C-terminal protein-binding domain in the retinoblastoma protein regulates nuclear c-Abl tyrosine kinase in the cell cycle. Cell 75:779-90. Wells, J., K. E. Boyd, C. J. Fry, S. M. Bartley, and P. J. Farnham. 2000. Target gene specificity of E2F and pocket protein family members in living cells. Mol Cell Biol 20:5797-807. White, R. J. 1997. Regulation of RNA polymerases I and III by the retinoblastoma protein: a mechanism for growth control? Trends Biochem Sci 22:77-80. 38 141. 142. 143. 144. 145. 146. 147. 148. 149. 150. White, R. J., T. M. Gottlieb, C. S. Downes, and S. P. Jackson. 1995. Cell cycle regulation of RNA polymerase III transcription. Mol Cell Biol 15:6653-62. White, R. J., D. Trouche, K. Martin, S. P. Jackson, and T. Kouzarides. 1996. Repression of RNA polymerase III transcription by the retinoblastoma protein. Nature 382:88-90. Williams, B. O., E. M. Schnritt, L. Remington, R. T. Bronson, D. M. Albert, R. A. Weinberg, and T. Jacks. 1994. Extensive contribution of Rb-deficient cells to adult chimeric mice with limited histopathological consequences. EMBO J 13:4251-9. Willis, I. M. 1993. RNA polymerase III. Genes, factors and transcriptional specificity. Eur J Biochem 212:1-11. 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. Yoon, J. B., S. Murphy, L. Bai, Z. Wang, and R. G. Roeder. 1995. Proximal sequence elernent-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. Yoon, J. B., and R. G. Roeder. 1996. Cloning of two proximal sequence element-binding transcription factor subunits (gamma and delta) that are required for transcription of small nuclear RNA genes by RNA polymerases II and III and interact with the TATA-binding protein. Mol Cell Biol 16:1-9. Zacksenhaus, E., Z. Jiang, D. Chung, J. D. Marth, R. A. Phillips, and B. L. Gallie. 1996. pr controls proliferation, differentiation, and death of skeletal muscle cells and other lineages during embryogenesis. Genes Dev 10:3051-64. 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 regulated by repressor complexes containing HDAC-Rb-hSWI/SNF and Rb- hSWI/SNF. Cell 101:79-89. Zhao, X., P. Shannon Pendergrass, and Nouria Hernandez. 2001. A positioned nucleosome in the human U6 promoter allows recruitment of SNAPc by the Oct-1 POU domain. Mol Cell 7:539-549. 39 Chapter 2 The retinoblastoma tumor suppressor protein targets distinct general transcription factors to regulate RNA polymerase III gene expression'. Abstract The retinoblastoma protein (RB) represses RNA polymerase III transcription effectively both in vivo and in vitro. Here we demonstrate that the general transcription factors SNAPc and TBP are important for RB repression of human U6 snRNA gene transcription by RNA polymerase III. RB is associated with SNAPc as detected by both co- immunoprecipitation of endogenous RB with SNAPc and co-fractionation of RB and SNAPc during chromatographic purification. RB also interacts with two SNAPc subunits, SNAP43 and SNAPSO. TBP or a combination of TBP plus SNAPc restores efficient U6 transcription from RB-treated, extracts indicating that TBP is also involved in RB regulation. In contrast, the TBP-containing complex TFIIIB restores adenovirus VAI but not human U6 transcription in RB-treated extracts suggesting that TFIIIB is important for RB regulation of tRNA-like genes. These results suggest that different classes of RNA polymerase III-transcribed genes have distinct general transcription factor requirements for repression by RB. This work was published as the following manuscript: Hirsch, H. A., Gu, L., and Henry, R. W. (2000). The Retinoblastoma tumor suppressor protein targets distinct general transcription factors to regulate RNA polymerase III gene expression. Mol Cell Biol 20, 9182-91. 40 Introduction RB is a tumor suppressor that controls cell growth by influencing cell-cycle progression (8, 12, 44), differentiation (5, 15, 44), and apoptosis (23, 68). Mutations in the gene encoding RB are associated with diverse human cancers (21, 22, 27, 45). RB function is also compromised in other human malignancies through disruption of upstream control pathways or downstream targets of RB (reviewed in 58). The function of RB as a tumor suppressor is linked to its ability to regulate gene expression. Therefore, to fully understand the contribution of RB to cellular proliferation observed during carcinogenesis, it is important to determine the mechanisms that RB uses to regulate gene activity. An understanding of RB firnction in gene regulation was revealed through its role as a modulator of E2F transcription factor activity (16, 24, 25, 59). However, RB controls additional cellular firnctions beyond regulating E2F activity. The intracellular concentration of RB exceeds the concentration of E2F (58) and interactions between RB and other transcription factors have been described (10, 34, 51). Thus, further activities performed by RB involve regulation of other genes besides E2F-responsive genes. Interestingly, RB is not limited to regulating mRNA production by RNA polymerase II but also inhibits the synthesis of ribosomal RNAs by RNA polymerase I (4) and of 5S rRNA, tRNA, and U6 snRNA by RNA polymerase III (63). It was proposed that loss of control of these genes is an important step in tumor progression because the products of 41 genes transcribed by RNA polymerases 1 and III are important determinants of biosynthetic capacity (reviewed in 61). Repressed synthesis of non-translated RNAs is expected to inhibit cell proliferation presenting a significant hurdle to unregulated cell growth. Therefore, control of RNA polymerase I and III transcriptional activity may represent an essential component of growth regulation by RB. How RB regulates RNA polymerase III activity in the cell is not clear. RNA polymerase III transcriptional activity is under cell cycle control with higher levels observed in the late G1, S and G2 phases of the cell cycle than G0 and early G1 (62). The increase in RNA polymerase III activity correlates with an increase in phosphorylated RB during the G1 phase of the cell cycle. This is important because the firnction of RB is controlled by phosphorylation (6, 38). Hypo-phosphorylated RB can interact with potential target proteins to regulate their activity whereas hyper-phosphorylated RB cannot interact and therefore is inactive (58). RNA polymerase III activity is maximal during the cell cycle when RB is inactive. This implies that hypo-phosphorylated RB may target factors that function in RNA polymerase III transcription. The correlation between RB levels and RNA polymerase III activity has been further demonstrated in vivo by transient transfection assays of adenovirus (Ad) VAI gene transcription. Transcription of this gene by RNA polymerase III is elevated in a human osteosarcoma cell line (SAOS2) that is RB-deficient compared to an osteosarcoma cell line (UZOS) that contains functional RB. Over-expressing RB in SAOSZ cells represses RNA polymerase III transcription whereas RNA polymerase II transcription from the HIV LTR is unaffected. Furthermore, in nuclear run-off assays, RNA polymerase III-specific transcription is diminished in nuclei 42 isolated from wildtype mouse embryonic fibroblasts compared to nuclei isolated from mouse RB-/- embryonic fibroblasts whereas wholesale RNA polymerase II activity is unchanged in RB+/+ and RB-/- embryonic fibroblasts (63). These experiments suggest that RNA polymerase III activity in vivo is regulated by RB. We have focused on understanding the contribution of RB to repression of RNA polymerase III activity. Genes transcribed by RNA polymerase 111 can be subdivided into four classes. Class 1 and class 2 genes contain gene-intemal promoter elements exemplified by the 5S rRNA and tRNA genes, respectively. Class 3 genes contain gene external promoter elements exemplified by the human U6 snRNA genes. A fourth class exemplified by the Vault RNA genes contain both external and internal promoter elements (54). RNA polymerase III-transcribed genes also have distinct general transcription factor requirements consistent with their different promoter architectures. SS rRNA genes require TFIIIA, TFIIIB, and TFIIIC, whereas tRNA gene transcription only requires a subset of these factors, TFIIIB and TFIIIC, for full activity (50, 52, 64). In contrast, human U6 gene transcription requires the snRNA activating protein complex or SNAPc (48), which is also known as PTF (42). While TFIIIC is not required, the requirement for TFIIIB is controversial (39, 57). In addition to these general transcription factors, other transcription activator proteins including Oct-1 (3), Spl (30), and STAF (43, 49) positively regulate U6 snRNA gene transcription. The mechanism that RB utilizes to repress RNA polymerase III transcription is not known. However, potential targets for regulating RNA polymerase III activity include 43 TFIIIB and the TFIIIC2 form of TFIIIC (7, 29). Human TFIIIB consists of the TATA box binding protein (TBP) and a tightly associated factor called TFIIB-related factor or BRF (39, 57). By analogy with yeast TFIIIB (26), a loosely associated factor referred to as B" may also be a component of human TFIIIB. BRF and TBP associate with RB during chromatographic fractionation of cellular extracts and during co- immunoprecipitation experiments (29). TFIIIC2 is a multi-protein complex containing five proteins (67). RB can also interact with TFIIIC2 in GST-pulldown experiments from HeLa nuclear extracts (7). Together, these data indicate that RB can interact with the RNA polymerase 111 general transcription machinery, and this may be important for RB repression of RNA polymerase III-specific gene transcription. It is not known whether RB targets similar factors to regulate all classes of RNA polymerase III transcribed genes. In contrast to the clear requirement of TFIIIB and TFIIIC2 for RNA polymerase III transcription of genes containing gene-internal promoter elements, neither TFIIIC (55) nor a TFIIIB complex of BRF and TBP is required for human U6 snRNA gene transcription in vitro (17, 39). Potentially, a different form of TFIIIB may function both for U6 gene transcription and regulation by RB. Other alternative spliced forms of BRF have been identified and one form referred to as hBRF 2 functions for human U6 transcription in vitro (37). It is also possible that RB targets other factors to regulate human U6 snRNA genes. One potential target is SNAPc. SNAPc is a multi-protein complex composed of at least five proteins SNAP19 (19), SNAP43/PTFy (20, 66), SNAP45/PTF5 (47, 66), SNAPSO/PTFB (l, 18) and SNAP190/PTFOt (65). In 44 addition, SNAPc associates with TBP (20). SNAPc binds to the proximal sequence element (PSE) contained in the core promoter regions of human U6 snRNA genes and interacts with TBP bound to the TATA box. Together SNAPc and TBP act cooperatively to facilitate transcription by RNA polymerase III (40). The binding of these factors to the promoter is a crucial early step in pre-initiation complex assembly at these genes and therefore SNAPc and TBP are attractive targets for regulating human snRNA gene transcription. Our results suggest that RB regulates different RNA polymerase III-transcribed genes by targeting different components of the general transcription machinery. The general transcription factor TFIIIB, composed of BRF and TBP, functionally restores Ad VAI gene transcription but not human U6 snRNA gene transcription in RB-treated extracts. In contrast, a combination of the general transcription factors SNAPc and TBP act cooperatively to reconstitute U6 snRNA gene transcription after RB treatment indicating that these factors are also important for RB regulation of RNA polymerase III activity. Depleting extracts with GST-RB (379-928) resulted in a reduction in SNAP43 levels consistent with the idea that RB is targeting SNAPc. In HeLa cell nuclear extracts, a sub- population of RB is associated with SNAPc and this association may be direct because RB interacts effectively with two components of SNAPc. These data indicate that the general transcription factors SNAPc and TFIIIB provide an important targeting mechanism governing RB function for different classes of RNA polymerase III- transcribed genes. 45 ”'3 C") Materials and Methods Expression and purification of recombinant proteins: The region of RB corresponding to amino acids 379 to 928 was amplified by polymerase chain reaction and cloned into a pETl lc-based expression vector to generate pGST-RB (379-928). This contains an N- terrninal GST tag fused in frame with RB (379-928). Both SNAP43 (1-368) and SNAPSO (1-411) were constructed in a similar manner to generate pGST-SNAP43 (1-368) and pGST-SNAPSO (1-411). GST-fusion proteins were expressed in E. coli BL21 DE3 and extracts were prepared by sonication. Proteins were purified by binding to glutathione agarose beads (Sigma) followed by extensive washing in HEMGT-150 buffer (20 mM Hepes pH 7.9; 0.5 mM EDTA; 10 mM MgC12; 10% glycerol; 0.1% Tween-20) containing protease inhibitors (0.1 mM PMSF, 1 mM sodium bis-sulfate, 1 mM benzamidine, 1 uM pepstatin A) and 1 mM DTT. Bound proteins were then used directly for GST pulldown assays. For in vitro transcription experiments, GST-RB (379-928) and GST were eluted from beads in HEMGT-ISO buffer containing 50 mM glutathione for 1 hr at 4°C. Eluted proteins were dialyzed against Dignam buffer D (9) and concentrated by centrifugation through YMlO centricon columns (Millipore) to give a final concentration of at least 200 ng/ul. Protein expression levels, efficiency of binding to glutathione agarose beads, and protein concentrations were monitored by SDS-PAGE and staining with Coomassie blue. 46 .11, kr.’ 1,1,) .T' In vitro transcription assays: In vitro transcription of the adenovirus VAI and human U6 snRNA genes were performed essentially as described (31, 32, 48). For repression assays depicted in Figure 1, HeLa cell nuclear extracts were pre-incubated with purified recombinant proteins at 30°C for 30 minutes. The amounts of each protein used are indicated in the figure legend. Twenty microliter transcription reactions were initiated by addition of 0.25 ug DNA templates (pBSM13+VAI; pU6/Hae/RA.2), rNTPs, and transcription buffer. Transcription reactions were performed for 1 hour at 30° C. Transcripts were separated by denaturing PAGE and visualized by autoradiography or by phosphoimager analysis (Molecular Dynamics). RB afl‘mity depletion assays: For human U6 snRNA gene repression assays shown in Figures 2A, 8 uL HeLa cell nuclear extract (7.5 ug/ uL) was pre-incubated with 1, 2, or 3 uL of purified GST-RB (379-928) or GST (each at 1 ug/ uL) for 30 min at 30°C. GST- RB (379-928) and GST were removed by affinity purification with glutathione sepharose beads (Pharmacia) added at a 1:1 ratio of beads to extract. Samples were then incubated at 4°C for 4 hours and centrifuged to remove associated proteins. One half of each supernatant (4.5, 5.0, and 5.5 uL of the l, 2, and 3 ug treated samples, respectively) was used for in vitro transcription assays as described above. For the experiment shown in Figure 2B, the affinity depletion reactions were scaled up to include 32 uL nuclear extract (7.5 ug/ LIL) plus 10 ug GST-RB (379-928) (200 ng/ uL). For Ad VAI gene repression assays shown in Figure 2C, 24 uL HeLa cell nuclear extract (7.5 ug/ uL) was 47 pre-incubated with approximately 14 ug purified GST-RB (379-928) (200 ng/ uL). Similar pre-incubation reactions were performed with either Dignam buffer D (mock depleted) or GST protein at equivalent amounts as used for GST-RB (379-928). After affinity depletion, supematants were used immediately in adenovirus VAI and human U6 transcription reactions as described previously. Transcription reactions were also supplemented with chromatographic fractions containing SNAPc (Mono-Q peak fraction; approx. 0.3 mg/ml protein; ref. 20) or TFIIIB (PII-B; approx. 0.6 mg/ml; ref. 32). For the experiment presented in Figure 28, 10 ng of recombinant human TBP (Promega) was also added as indicated. For the experiment presented in Figure 2D, 20 uL HeLa nuclear extract was treated with 6 ug of purified GST-RB (379-928) or GST as described above. 15 uL of each supernatant and proteins bound to the beads after extensive washing were separated by 12.5 % SDS-PAGE and tested by western blot analysis using rabbit anti- SNAP43 antiserum (CS48; ref. 20) or mouse monoclonal antibodies (SL2; ref. 33). RB/SNAPc co-immunoprecipitation: Approximately 300 uL HeLa cell nuclear extract was incubated with 2 ug mouse anti-RB (clone G3-245; Phanningen) or anti- Haemaglutinin (12CA5) antibodies overnight at 4°C. Samples were diluted with 1 mL of HEMGT-ISO containing protease inhibitors and 20 uL Protein-G agarose beads (Gibco- BRL) were added to each reaction. Samples were further incubated at 4°C for 4 hours. Antibody beads were washed in HEMGT-lSO containing protease inhibitors (3 x 1 mL), bound proteins were eluted by boiling in 1x Laernmli buffer prior to size fractionation by 12.5% SDS-PAGE. SNAP43 was detected by western blot analysis using antibodies 48 specific to SNAP43 (C848; ref. 20). To perform the reciprocal immunoprecipitations, approximately 300 uL of HeLa cell nuclear extract was incubated with 50 11L protein-A agarose beads (Boehringer Mannheim) pre-coupled with either rabbit anti-SNAP43 or pre-immune antibodies. Reactions were incubated for 2 hr at 4°C with mixing. Beads were washed extensively in Dignam buffer D (100 mM KCl) containing protease inhibitors and bound proteins were then competitively eluted in 200 uL Dignam buffer D containing either a specific peptide (CSH375: ref. 20) or non-specific peptide each at 1 mg/mL. Eluted samples were precipitated with trichloroacetic acid. Precipitates were re- dissolved in 1x Laernmli buffer and size fractionated by 12.5% SDS-PAGE. Full length RB was detected by western blot analysis using mouse monoclonal antibodies directed against an epitope contained within amino acids 300-380 (G3-245; Pharmingen). Protein Chromatography and EMSA analysis: Nuclear extracts were prepared from HeLa cells by the method of Dignam et al. (1983). SNAPc- and TFIIIB-containing fractions were generated essentially as described previously (20, 32, 48). The SNAPc fractions used are from the Mono-Q step of purification. The TFIIIB fractions used are from the P1 l-B step of purification. PSE-specific DNA binding by SNAPc was assayed by EMSA as described (48). GST-pulldown assays: Individual SNAPc subunits and full-length RB were individually expressed in vitro using rabbit reticulocyte lysates (TNT-Promega) and proteins were labeled with 35S-methionine. GST-pulldown reactions were performed using 20 uL 49 glutathione agarose beads containing approximately 1 ug of GST-RB (379-928), GST- SNAPSO (1-411), GST-SNAP43 (1-368) and GST or beads alone. These were individually incubated with 10 [IL of 35S-labelled proteins for 2 hours at 4°C in 1 mL HEMGT-ISO containing protease inhibitors and 1 mM DTT. The specific combinations of proteins used are indicated in the figure legend. Beads were washed extensively in HEMGT-ISO and bound proteins were separated by 17% SDS-PAGE. Proteins were stained with Coomassie blue to ensure equivalent loading of GST tagged proteins in each sample. Associated radioactive proteins were detected by autoradiography. EMSA experiments: See appendix A Results RB represses RNA polymerase III transcription RB is an important regulator of cellular growth and its ability to perform this function can partially be attributed to regulation of RNA polymerase III activity. RB contains 928 amino acids and can be divided into at least three regions: the N-terminal region from amino acids 1-378, the NB region from 393-772, and the C region from 768-869. Most functions ascribed to RB including tumor suppressor activity and interactions with regulatory target proteins require either the NB and/or C regions (56, 60). To determine the function of RB in regulating RNA polymerase III activity, recombinant RB containing the A/B and C regions was tested for its ability to repress in vitro 50 transcription by RNA polymerase III. Specifically, in vitro transcription assays of the Ad VAI gene and a human U6 snRNA gene were performed to compare RB regulation of RNA polymerase III transcription for genes containing gene-internal (class 2) and gene- extemal (class 3) promoter elements. A schematic representation of the core-promoters of the genes used for this study is shown in Figure 2-1A. The Ad VAI gene contains gene- intemal A and B box control elements that are also characteristic of human tRNA genes. The core-promoter regions of human U6 snRNA genes contain a PSE and a TATA box. In addition, the U6 gene contains a DSE that recruits Oct-l to activate U6 transcription. The GST-RB (379-928) and GST proteins typically used for these experiments are shown in Figure 2-lB. GST-RB (379-928) and GST were each expressed in E. coli and were purified to homogeneity by affinity purification using glutathione agarose beads. In each case, the full-length protein is the most prevalent species observed. To determine the effect of these proteins on RNA polymerase III transcription, increasing amounts of purified GST-RB (379-928) and GST were added to HeLa cell nuclear extracts and these were tested for ability to support Ad VAI transcription. As shown in Figure 2-1C, GST- RB (379-928) inhibited transcription of the Ad VAI gene (top panel: lanes 2-5) compared to levels observed for the untreated extract (lane 1). This repression appears to be specific because addition of equivalent amounts of the GST control protein had no significant effect on Ad VAI transcription (lanes 6-9). The repression observed for RB is not limited to "classical" RNA polymerase III transcribed genes containing gene internal promoter 51 Figure 2-1: RB represses in vitro transcription by RNA polymerase III. (A) Schematic representation of the adenovirus VAI and human U6 snRNA promoters. (B) Analysis of GST-RB (379-928) and GST proteins used in transcription reactions. GST—RB (379-928) (lane 2) and GST (lane 3) were expressed in E. coli and purified by affinity chromatography using glutathione agarose beads and competitive elution with glutathione. After dialysis against Dignam buffer D proteins were separated by SDS- PAGE and visualized by staining with Coomassie blue. Lane 1 contains a protein size standard. (C) GST-RB (379-928) represses adenovirus VAI transcription by RNA polymerase III. Approximately 2 uL of HeLa cell nuclear extract (approx. 7.5 ug/uL) was incubated with 200, 400, 800, and 1200 ng of GST-RB (379-928) (lanes 2-5) or GST protein (lanes 6-9) at 30°C for 30 minutes. In vitro transcription of the Ad VAI gene (top panel) was then initiated by addition of template, cold rNTPs, [a32P]-CTP, and transcription buffer. Lane 1 shows the level of transcription with the untreated extract. Sample handling was monitored by a non-specific RNA handling control transcript (bottom). (D) GST-RB (379-928) represses human U6 snRNA gene transcription by RNA polymerase III. Approximately 2 uL of HeLa cell nuclear extract was incubated with 100, 250, and 500 ng of GST-RB (379-928) (lanes 2-4) or GST protein (lanes 5-7). Lane 1 shows the level of transcription with the untreated extract. Correctly initiated transcripts from the U6 promoter (labeled U6 5') were detected by RNase T1 protection essentially as described (31). 52 nomvmwe mmfiormvmwrw be: Ii. .g _o:coo _H i l . I. I ' .50 88.2.8 . mm.th D _<>_H : i . m N P _| _ p . I ' r ”.3 .50 83.2.9 0 ' r «.3 macho I .II - S I - .. .. JEEIE :— .Il. ' I am ommemEzod I r «3 (D (9 z , H a - U6 5' 1 2 3 4 or-RNAP III Primag IP 3 E 3 Ea 32 P33%é .... ... —pU6II-IaelRa2 Untreated .- pUC119 (379-928) ... pUC119 l. l ... .. _ pU6/HaelRaZ GST ... pUC119 No Nuclear ... . pU6/HaelRaZ Extract .. . pUC119 1234 118 or treated with GST-RB or GST. Portions of these reactions were analyzed by RNase protection assays for full-length transcripts driven from the U6 promoter while the remainder was subjected to formaldehyde cross-linking followed by or-RNA polymerase III immunoprecipitation. Secondary immunoprecipitations were then performed using I gG, or-RNA polymerase III, or or-RB antibodies. The precipitates were then analyzed for the presence of the U6 reporter construct (pU6/Hae/RA.2) or a negative control plasmid (pUC119) that was included in the original transcription reactions. Figure 3-6A shows the level of U6 gene transcription from samples containing HeLa cell nuclear extract (lanel), extract plus GST-RB (lane 2), or extract plus GST (lane 3). Transcription from samples lacking any nuclear extract is shown in lane 4. As expected, GST-RB effectively :repressed U6 transcription as compared to the effect of GST. The result of the sequential immunoprecipitations from these transcription reactions is shown in Figure 3-6B. RNA polymerase III was detected at the U6 promoter-containing plasmid in transcription reactions performed with untreated nuclear extract (top panel) but not in reactions to Vvhich no extract was added Gmttom panel). Significantly, RNA polymerase III was also detected at this promoter DNA in transcription reactions that were completely repressed by GST-RB (second panel). In these GST-RB repressed transcription assays, RB also associated with the U6 promoter but not with the irrelevant plasmid. The pattern of factor association with the U6 promoter plasmid in the GST-treated samples (third panel) was Similar to that observed with samples containing the untreated nuclear extract. Together, tlrese observations indicate that under repressed conditions, RB does not displace RNA polymerase 111, but rather RB and RNA polymerase 111 can co-occupy the same repressed promoter. 119 Discussion To better understand how RB acts to prevent tumor progression it is important to understand how RB regulates gene expression. Previous studies illustrate that RB can repress transcription by RNA polymerase I (3), II (31), and III (35) suggesting a role for RB in the regulation of a diverse set of genes. This versatility allows RB to participate in many processes critical to normal cell function including cell cycle progression, apoptosis, DNA replication, and differentiation. RB’s contribution to each of these processes may play a role in the suppression of tumor formation. Additionally, RB may regulate cell growth by the regulation of RNA polymerase III gene expression. The non-translated genes transcribed by RNA polymerase III provide metabolic building blocks that increase the biosynthetic capacity of the cell. An increase in a cell’s biosynthetic capacity elevates the cell’s growth potential, which unchecked, could lead to tumorigenesis. Bypass of RB regulation of non-translated RNAs could represent a significant obstacle to overcome during the progression of tumor formation. RB can repress transcription by RNA polymerase 111 both in vivo (35) and in vitro (15, 35) including transcription of U6 snRNA genes (13). The data herein illustrate that RB can associate with the U6 snRNA promoter, a step that may be important for subsequent repression of transcription. The chromatin immunoprecipitation assays clearly show the RB protein occupying U6 snRNA promoter in vivo suggesting that RB does in fact target this gene for transcriptional regulation. In contrast, RB is not present at the U1 and U2 120 RNA polymerase II transcribed snRNA genes. Interestingly, RB occupancy of snRNA promoters in vivo correlates with the ability to repress transcription as demonstrated by in vitro transcription experiments. Whereas RB can repress U6 snRNA transcription by RNA polymerase 111, it is unable to repress transcription by RNA polymerase II initiated from a U1 snRNA promoter. Indeed, the data presented herein, illustrate that RB Preferentially targets and represses RNA polymerase III transcribed snRNA genes. One potential mechanism for the recruitment of RB to target promoters is through interaction with general factors required for transcription. In the case of the U6 snRNA gene both SNAPc and TFIIIB could be potential recruitment factors for RB. Previous studies demonstrated that RB can associate with SNAPc (13). However, if SNAPc alone were capable of mediating RB repression of U6 snRNA genes, then it should also direct RB repression activities to other genes at which SNAPc is present such as the U1 or U2 snRN A genes. Because RB was not detected at these genes, it is possible that SNAPc is not the sole determining factor for selective RB targeting RNA polymerase III transcribed snRNA genes. One obvious difference between snRNA gene transcribed by RNA polymerase II and III is the presence of a TATA element in core promoter region of RNAP 111 type snRNA genes. One possibility is that proteins or protein complexes recruited to this distinguishing promoter element are candidate selectivity factors directing RB solely to RNAP III transcribed snRNA genes rather than all snRNA genes. 121 Indeed, TFIIIB is one candidate selectivity factor that binds to the TATA elements of U6 snRNA promoters. The results in Figure 3-2 demonstrate that TFIIIB associates with endogenous RB by co-immunoprecipitation and that individual components of TFIIIB (del and TBP) interact with RB in GST-pulldown experiments. Thus, an alternative TFIIIB complex consisting of TBP, Brf2, and deI that associates with U6 snRNA promoters via the TATA element may be important for ensuring efficient RB recruitment to human U6 snRNA genes. In EMSA experiments, TFIIIB, like SNAPc, can recruit RB to U6 promoter DNA. RB recruitment is significantly enhanced in the presence of SNAPc, suggesting cooperative interactions between TFIIIB and SNAPc direct RB to the U6 snRNA promoter. Additionally, all components of SNAPc and TFIIIB are necessary for efficient recruitment of RB. The results of the EMSA experiments indicate that RB can interact with SNAPc and TFIIIB on a U6 snRNA DNA probe rather than simply displacing these factors to repress transcription. This result is also supported by the promoter occupancy studies of the U6 snRNA gene using sequential chromatin immunoprecipitation analysis. Sequential chromatin immunoprecipitation analysis revealed that RB, SNAPc and TFIIIB are present at the same U6 snRNA promoter in vivo again indicating that association of these basal factors is not interupted by RB. The unique surprising result from these sequential ChIP experiments is that RB and RNA polymerase III occupy the same U6 snRNA promoter. Further in vitro analysis also indicates the RB and RNA polymerase 111 can occupy the same repressed U6 snRNA promoter. 122 The results presented herein are inconsistent with the previous model in which RB represses transcription by blocking the binding of RNA polymerase III to cognate promoters. Sutcliffe and co-workers (26) proposed a mechanism of RNA polymerase III transcriptional repression in which RB sequesters required factors. RB was proposed to bind to TFIIIB interrupting important interactions with TFIIIC. Since interaction with TFIIIC is critical for recruitment to tRNA genes, RB effectively sequesters TFIIIB away from the promoter thereby not allowing pre-initiation complex formation to occur. Similarly, RB was proposed to repress some RNA polymerase II transcribed genes by targeting the TFIID-TFIIA complex formation step to prevent PIC formation for RNA polymerase II (21). Since RB and RNA polymerase III occupy the same promoter under repressed conditions, RB is clearly not using a commonly accepted mechanism, disrupting pre-initiation complex formation, to repress transcription at the U6 snRNA promoter. Again, the results described herein disagree with accepted mechanisms of RB repression. Instead, we propose that, at least for human U6 transcription, RB impedes critical steps in RNA polymerase III transcription that occur subsequently to polymerase recruitment to the promoter. For example, RB may create a physical barrier in the chromatin at the start site of transcription that would block the progression of the polymerase. Potentially RB could recruit histone modification activities that would modify local chromatin structure preventing transcription by RNA polymerase 111. However, several studies have indicated that HDAC mediated RB repression is promoter specific and may not occur at all RB regulated promoters (l6). HDAC mediated repression of U6 genes may be 123 unlikely as data from other groups suggest that RB can repress expression of U6 gene in an HDAC independent fashion (26). Another method by with RB could use to create a physical barrier in chromatin to prevent U6 gene expression is through the recruitment of ATP-dependent chromatin remodeling complexes such as SWI-SNF. Previous studies have demonstrated interaction between RB and SWI-SNF complexes and that RB required SWI-SNF to repress transcription initiated from some promoters (28, 37). In vivo, the U6 promoter contains a positioned nucleosome between the DSE and PSE that is vital to proper gene expression (38). Recruitement of chromatin remodeling factors by RB to a U6 promoter could potentially mis-align this nucleosome and thus effectively abolish U6 gene expression. Additionally, RB may hinder steps that allow for promoter escape and subsequent transcription of the U6 snRNA gene. Furthermore, RB could inhibit steps that convert an initiating polymerase to an elongating polymerase such as phosphorylation of the enzyme. Perhaps RB interferes with elongation resulting in a stalled complex in the middle of the U6 snRNA gene. Whichever case may apply, it represents a novel mechanism for RB regulation of gene expression. Acknowledgments We gratefully thank S. Conrad, P. Voss, J. Wang, F. Herrera, S. Triezenberg, and N. Hernandez for supplying reagents as well as C. Hinkley, D. Arnosti, S. Triezenberg, and G. Chen for critical reading of the manuscript. We also thank J. Mann for technical 124 assistance. This work was supported by grants from the National Institutes of Health and American Cancer Society to RWH. References l. 10. ll. Brehm, A., Miska, E. A., McCance, D. J., Reid, J. L., Bannister, A. J., and Kouzarides, T. (1998). Retinoblastoma protein recruits histone deacetylase to repress transcription. Nature 391, 597-601. Cabart, P., and Murphy, S. (2002). Assembly of human snRNA gene-specific TFIIIB complex de novo on and off promoter. J Biol Chem 16, 16. Cavanaugh, A. H., Hempel, W. M., Taylor, L. J., Rogalsky, V., Todorov, G., and Rothblum, L. I. (1995). Activity of RNA polymerase I transcription factor UBF blocked by Rb gene product. Nature 374, 177-80. Chellappan, S. P., Hiebert, S., Mudryj, M., Horowitz, J. M., and Nevins, J. R. (1991). The E2F transcription factor is a cellular target for the RB protein. Cell 65, 1053-61. Dagher, S. F ., Wang, J. L., and Patterson, R. J. (1995). Identification of galectin- 3 as a factor in pre-mRNA splicing. Proc Natl Acad Sci U S A 92, 1213-7. Dynlacht, B. D. (1997). Regulation of transcription by proteins that control the cell cycle. Nature 389, 149-52. Dyson, N. (1998). The regulation of E2F by pRB-family proteins. Genes Dev 12, 2245-62. Harbour, J. W., and Dean, D. C. (2000). The Rb/E2F pathway: expanding roles and emerging paradigms. Genes Dev 14, 2393-409. Henry, R. W., Mittal, V., Ma, B., Kobayashi, R., and Hernandez, N. (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., Sadowski, C. L., Kobayashi, R., and Hernandez, N. (1995). A TBP-TAF complex required for transcription of human snRNA genes by RNA polymerase II and 111. Nature 374, 653-6. Hiebert, S. W., Chellappan, S. P., Horowitz, J. M., and Nevins, J. R. (1992). The interaction of RB with E2F coincides with an inhibition of the transcriptional activity of E2F. Genes Dev 6, 177-85. 125 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. Hinkley, C. 8., Hirsch, H. A., Gu, L., Lamere, B., and Henry, R. W., (2003). The Small Nuclear RNA-activating Protein 190 Myb DNA Binding Domain Stimulates TATA Box-binding Protein-TATA Box Recognition J. Biol. Chem. 278: 18649- 18657. Hirsch, H. A., Gu, L., and Henry, R. W. (2000). The retinoblastoma tumor suppressor protein targets distinct general transcription factors to regulate RNA polymerase III gene expression. Mol Cell Biol 20, 9182-91. Kaelin, W. G., Jr. (1997). Recent insights into the functions of the retinoblastoma susceptibility gene product. Cancer Invest 15, 243-54. Larminie, C. G., Cairns, C. A., Mital, R., Martin, K., Kouzarides, T., Jackson, S. P., and White, R. J. (1997). Mechanistic analysis of RNA polymerase III regulation by the retinoblastoma protein. EMBO J 16, 2061-71. Luo, R. X., Postigo, A. A., and Dean, D. C. (1998). Rb interacts with histone deacetylase to repress transcription. Cell 92, 463-73. Magnaghi-Jaulin, L., Groisman, R., Naguibneva, 1., Robin, P., Lorain, 8., Le Villain, J. P., Troalen, F., Trouche, D., and Harel-Bellan, A. (1998). Retinoblastoma protein represses transcription by recruiting a histone deacetylase. Nature 391, 601-5. Mital, R., Kobayashi, R., and Hernandez, N. (1996). RNA polymerase III transcription from the human U6 and adenovirus type 2 VAI promoters has different requirements for human BRF, a subunit of human TFIIIB. Mol Cell Biol 16, 7031- 42. Murphy, 8., Yoon, J. B., Gerster, T., and Roeder, R. G. (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. Nevins, J. R., Chellappan, S. P., Mudryj, M., Hiebert, S., Devoto, S., Horowitz, J., Hunter, T., and Pines, J. (1991). E2F transcription factor is a target for the RB protein and the cyclin A protein. Cold Spring Harb Symp Quant Biol 56, 157-62. Ross, J. F., Liu, X., and Dynlacht, B. D. (1999). Mechanism of transcriptional repression of E2F by the retinoblastoma tumor suppressor protein. Mol Cell 3, 195- 205. Schramm, L., Pendergrast, P. 8., Sun, Y., and Hernandez, N. (2000). Different human TFIIIB activities direct RNA polymerase III transcription from TATA- containing and TATA-less promoters. Genes Dev 14, 2650-63. 126 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. Scott, P. H., Cairns, C. A., Sutcliffe, J. E., Alzuherri, H. M., McLees, A., Winter, A. G., and White, R. J. (2001). Regulation of RNA polymerase III transcription during cell cycle entry. J Biol Chem 276, 1005-14. Stampfer, M. R., and Bartley, J. C. (1985). Induction of transformation and continuous cell lines from normal human mammary epithelial cells after exposure to benzo[a]pyrene. Proc Natl Acad Sci U 8 A 82, 2394-8. Strobeck, M. W., Knudsen, K. E., Fribourg, A. F., DeCristofaro, M. F., Weissman, B. E., Imbalzano, A. N., and Knudsen, E. S. (2000). BRG-l is required for RB-mediated cell cycle arrest. Proc Natl Acad Sci U S A 97, 7748-53. Sutcliffe, J. E., Brown, T. R., Allison, S. J., Scott, P. H., and White, R. J. (2000). Retinoblastoma protein disrupts interactions required for RNA polymerase III transcription. Mol Cell Biol 20, 9192-202. Teichmann, M., Wang, Z., and Roeder, R. G. (2000). A stable complex of a novel transcription factor IIB- related factor, human TFIIIBSO, and associated proteins mediate selective transcription by RNA polymerase III of genes with upstream promoter elements. Proc Natl Acad Sci U S A 97, 14200-5. Trouche, D., Le Chalony, C., Muchardt, C., Yaniv, M., and Kouzarides, T. (1997). RB and hbrrn cooperate to repress the activation functions of E2F 1. Proc Natl Acad Sci U S A 94, 11268-73. Wang, Z., and Roeder, R. G. (1995). Structure and function of a human transcription factor TFIIIB subunit that is evolutionarily conserved and contains both TFIIB- and high- mobility-group protein 2-related domains. Proc Natl Acad Sci U S A 92, 7026—30. Weinberg, R. A. (1995). The retinoblastoma protein and cell cycle control. Cell 81, 323-30. Weintraub, S. J., Prater, C. A., and Dean, D. C. (1992). Retinoblastoma protein switches the E2F site from positive to negative element. Nature 358, 259-61. White, R. J. (1997). Regulation of RNA polymerases I and III by the retinoblastoma protein: a mechanism for grth control? Trends Biochem Sci 22, 77-80. White, R. J., Gottlieb, T. M., Downes, C. S., and Jackson, S. P. (1995). Cell cycle regulation of RNA polymerase III transcription. Mol Cell Biol 15, 6653-62. White, R. J., Gottlieb, T. M., Downes, C. S., and Jackson, S. P. (1995). Mitotic regulation of a TATA-binding-protein-containing complex. Mol Cell Biol 15, 1983- 92. 127 35. 36. 37. 38. White, R. J., Trouche, D., Martin, K., Jackson, S. P., and Kouzarides, T. (1996). Repression of RNA polymerase III transcription by the retinoblastoma protein. Nature 382, 88-90. Yoon, J. B., Murphy, 8., Bai, L., Wang, Z., and Roeder, R. G. (1995). Proximal sequence element-binding transcription factor (PTF) is a multisubunit complex required for transcription of both RNA polymerase II- and RNA polymerase III- dependent small nuclear RNA genes. Mol Cell Biol 15, 2019-27. Zhang, H. 8., Gavin, M., Dahiya, A., Postigo, A. A., Ma, D., Luo, R. X., Harbour, J. W., and Dean, D. C. (2000). Exit from GI and S phase of the cell cycle is regulated by repressor complexes containing HDAC-Rb-hSWI/SNF and Rb- hSWI/SNF. Cell 101, 79-89. Zhao, X., Pendergrast, P. 8., and Hernandez, N. (2001). A positioned nucleosome on the human U6 promoter allows recruitment of SNAPc by the Oct-l POU domain. Mol Cell 7, 539-49. 128 Chapter 4 The intact Large A/B pocket domain and C-terminus of RB is required for efficient repression of RNA polymerase III transcription. Abstract The Retinoblastoma tumor suppressor protein plays a central role in many cellular processes including cell cycle regulation, apoptosis, differentiation, and growth control. This protein contributes to growth control through its regulation of RNA polymerase III transcription. Here we dissect the region of RB required for repression of transcription and interaction with basal RNA polymerase III transcription factors. The region of RB between amino acids 379-870 is sufficient to repress both U6 snRNA and Ad VAI transcription since the ability to regulate transcription is lost in a RB mutant protein containing only amino acids 379-772. Interestingly this phenomenon correlates with the ability to interact with SNAP43, a component of the SNAP complex required for snRNA expression. Introduction RB is an important regulator of eukaryotic cellular growth. Its ability to perform this function can partially be attributed to regulation of RNA polymerase III activity (8, 18, 19). RB contains 928 amino acids and can be divided into at least three regions: the N- terminal region from amino acids 1-378, the NB region from 393-772, and the C region from 768-869 (16, 17). Most functions ascribed to RB including tumor suppressor 129 activity and interactions with regulatory target proteins require either the NB and/or C regions. When re-introduced into RB -/- mice, the region of RB between amino acids 379-928 was sufficient to rescue the phenotype and allow mice to develop normally (21). This same region of RB also appears to be the minimal region necessary for effective tumor suppression (21). Interestingly, the regions required for tumor suppression and transcription repression are different. Previous data has shown that the region from 379- 792 was sufficient to repress transcription from RNA polymerase II transcribed reporter constructs (3). Additionally, the region of RB from amino acids 379-772 was shown to be sufficient to recruit histone deacetylase activity to repress RNA polymerase II transcription (11). RB interacts with several components of RNA polymerase III basal machinery. Both SNAP43 and SNAPSO of the s_rI_RNA activating protein pomplex (SNAPc) interact strongly with RB (7). RB also interacts with TATA binding protein, TBP, (9 and HAH unpublished data) and B double prime, de1, which are components of all TFIIIB complexes. The TFIIB related factor 1, Brfl, a component of TFIIIB complexes involved in tRNA transcription, also weakly interacts with RB (9 and HAH unpublished data). RB does not appear to interact with RNA polymerase III itself (15 and HAH unpublished results). Additionally, RB has been shown to interact with TFIIIC (4, 15), a basal factor required for tRNA and SS rRNA expression, although this complex will not be studied filrther in this paper. Further characterization of RB interaction with basal factors and further insight into the mechanisms RB uses to regulate gene expression is essential in understanding the function of this critical tumor suppressor protein function. 130 Materials and Methods Tissue culture: Human mammary epithelieal cells (18485) were a gift from Susan Conrad. Cells were maintained in Dulbecco’s Minimum Essential Media (DMEM - Gibco) plus 10% Fetal Bovine serum (Gibco), 200 mM Glutarnine, and penicillin- streptomycin in 37°C incubator with 5% C02. Recombinant protein expression and purification: Mini-SNAPc: Plasmids encoding GST-SNAP43 Ex-XB, pET-GST-SNAPSO, and pET- GST-SNAP190(1-505) were transformed into E. coli BL21 DE3 codon + (Stratagene) and grown at 37°C in ZBM9 media until OD at 600 nm is 0.5. Temperature was lowered to 16°C and protein expression was induced with 1 mM IPTG for 20 hours. Pelleted bacteria were resuspended in HEMGT-150 (20 mM Hepes pH 7.9; 0.5 mM EDTA; 10 mM MgC12; 10% glycerol; 0.1% Tween-20) containing protease inhibitors (0.1 mM PMSF, 1 mM sodium bis-sulfate, 1 mM benzamidine, 1 11M pepstatin A) and 1 mM DTT and sonicated using a Branson sonifier 4 times for 30 pulses output=6 duty 60%. Debris was pelleted at 13,000 RPM at 4°C for 30 minutes. Soluble protein was bound to glutathione sepharose (Pharmacia) overnight at 4°C and washed 4 times in HEMGT-150 plus protease inhibitors and twice in HEMGT-150 without protease inhibitors. Proteins were cleaved from the beads using 20 units thrombin (Sigma) on ice for 1.5 hours vortexing every 15 minutes. SNAPSO and SNAP190(1-505) were collected as fractions in HEMGT-150. Appropriated fractions were pooled and dialyzed against Dignam buffer D80 (5). SNAP43 was collected in step wise salt gradient (150 to 1000 mM KCL in HEMGT buffer) and appropriate fractions pooled. Mini-SNAPc was assembled from 131 approximately 100 ug of each protein at room temperature for 2 hours. The assembly reaction was diluted such that the final salt concentration was less than 80 mM KCl and the final glycerol concentration was less than 5%. The diluted assembly reaction was allowed to equilibrilate on ice for 1 hour. Assembled mini-SNAPc was then loaded onto a 1 ml Mono-Q column. Bound proteins were eluted from the column in 0.5 ml fractions in a 0 to 500 mM salt gradient in Dignam buffer D80 (5% glycerol). Fractions were adjusted to 20% glycerol and assayed for SNAPc activity by SDS-PAGE and EMSA. The peak of SNAPc activity corresponded to fractions 20-24 (~380-420 mM KCl). Recombinant RB proteins were expressed and purified as before (7) with the exception that RB mutants were grown at a temperature of 30°C to an CD. of 0.6 and induced at 16°C for 20 hours. Chromatin immunoprecipitation: Chromatin immunoprecipitations were performed similarly to that described previously (2). Human 184BS cells were grown to 75% confluency and then crosslinked with formaldehyde for 30 minutes. After cell lysis and sonication, chromatin immunoprecipitations were performed (approximately 1 x 107 cells per immunoprecipitation) in Dilution buffer containing 400 mM NaCl using 1 ug of each antibody overnight at 4°C. Cross-links were reversed at 65°C overnight and recovered chromatin was suspended in 50 111 TE buffer. PCR analysis was performed using 5 11L of immunoprecipitated chromatin or input chromatin. The primers used for each gene are: U6 forward — 5’-GTACAAAATACGTGACGTAGAAAG-3’, U6 reverse — 5’-GGTGTTTCGTCCTTTCCAC-3’, U1 forward - 5’—CACGAAGGAGTTCCCGTG-3’, 132 U1 reverse — 5’-CCCTGCCAGGTAAGTATG-3 ’, U2 forward — 5’-AGGGCGTCAATAGCGCTGTGG-3 ’, U2 reverse - 5’-TGCGCTCGCCTTCGCGCCCGCCG-3 ’, GAPDH forward - 5’-AGGTCATCCCTGAGCTGAAC-3’, GAPDH reverse — 5’-GCAATGCCAGCCCCAGCGTC-3’, tRNA-lys forward — 5’-GGTTTCCCTCAAGGAGGGGG-3 ’, tRNA-lys reverse — 5 ’-GCCCGGATAGCTCAGTCGGTAG-3 ’ PCR products were separated by 2% TBE-agarose electrophoresis and visualized using Kodak imaging software. In vitro transcription: In vitro transcription assays were performed essentially as described previously (6). 2 ul of HeLa cell nuclear extract (~15 mg/ml) was used for the transcription reactions using 0.25 11g pU6/Hae/RA.2 or M13-VAI reporter plasmids. Transcription reactions were supplemented with 250 and 1000 ng of GST-RB (3 79-928), GST-RB (379-870), GST-RB (379-772), GST-RB (379-577) or GST as designated in the figure legend. Cross-linked immunoprecipitation of repressed promoters: In vitro transcription reactions were performed as described above with the following exceptions. Three- quarters of this reaction was processed as described above by T1 RNase protection (Figure 4A). The remaining fourth was diluted to 150 u] with ChIP Dilution buffer (as described in chapter 3) and was cross-linked in 1% formaldehyde for 15 minutes at room temperature and quenched in 0.125 M glycine. Ten ul of each cross-linked reaction was 133 used as starting material to perform immunoprecipitations as described in the immunoprecipitation section in chapter 3 using 10 ml of glutathione sepharose beads (Pharmacia). Recovered DNA was analyzed using the following primers: U6 forward — 5’-GTACAAAATACGTGACGTAGAAAG-3’, U6 reverse — 5’-GGTGTTTCGTCCTTTCCAC-3 ’, VAI forward — 5’-TCCGTGGTCTGGTGG-3’, VAI reverse — 5’-CGGGGTTCGAACCCGG-3’. Electrophoretic Mobility Shift Assays: DNA binding reactions were performed in 60 mM KCl, 20 mM HEPES pH 7.9, 5 mM MgC12, 0.2 mM EDTA, 10% glycerol, 0.5 ug poly(dI-dC), and 0.5ug pUC119 plasmid. Approximately 25 ng of SNAPc were incubated with increasing amount of GST-RB (379-928) or GST at room temperature for 20 minutes. 5000 CPM DNA probe (prepared as described previously (6)) containing with a high affinity PSE or mutant PSE was added to each reaction and incubated for 30 minutes at room temperature. The resulting complexes were separated by 5% Tris-Glycine polyacrylamide containing 5% glycerol and visualized by autoradiography. For the EMSA performed in figure 2, 1 ug goat or- RB (Santa Cruz) antibodies or goat IgG was added to the binding reactions at the times indicated. GST-Pulldown: GST-pulldowns were performed as described previously (7). 134 Results Characterization of the RNA polymerase III repression domain in RB In order to determine the minimal region of RB required to regulate RNA polymerase III transcription, C-terrninal truncation mutagenesis was performed on RB separating the protein into its pocket regions (Figure 4-1A) based on the structural domains observed in the RB crystal structure (10). These mutantions were created by Ms. Liping Gu in the Henry lab. Truncated RB proteins were expressed as GST fusion proteins in E. coli and affinity purified using gluthione-sepharose resin. Eluted proteins were desalted and concentrated using spin columns. Each RB protein was purified to near homogeneity (>90%) as shown by SDS-PAGE analysis and subsequent coomassie blue staining (Figure 4-1B). In vitro repression assays were used to test the ability of the RB truncation protein mutants to repress RNA polymerase III transcription. The two representative RNA polymerase III promoters examined, U6 snRNA and Adenovirus VAI, are diagramed in Figure 4-1C. For these experiments, HeLa nuclear extracts were pre-incubated with GST- RB, the RB truncation mutants, or GST as a negative control. Transcription was initiated by the addition of template and nucleotides. Correctly initiated transcripts were visualized by autoradiography. As shown in Figure 4-1D, addition of increasing amounts of GST- RB (379-928) (lanes 2-3) significantly decreases the amount of transcription correctly initiated from the U6 snRNA or VAI promoters as compared to untreated extract (lane 1) 135 Figure 4-1: Amino acids 379-870 of RB contain the minimal region necessary to repress RNA polymerase III transcription. (A) Schematic representation of the RB truncation mutants used in these experiments. (B) Analysis of GST-RB truncation mutants and GST proteins used in transcription reactions. GST-RB (379-928) (lane 2), GST-RB (379—870) (lane 3), GST-RB (379-772) (lane 4), GST-RB (379-577) (lane 5) and GST (lane 6) were expressed in E. coli and purified by affinity chromatography using glutathione agarose beads and competitive elution with glutathione. After dialysis against Dignam buffer D proteins were separated by SDS-PAGE and visualized by staining with Coomassie blue. Lane 1 contains a protein size standard. (C) Schematic representation of the adenovirus VAI and human U6 snRNA promoters. (D) GST-RB (379-928) and GST-RB (379-870) repress adenovirus U6 snRNA and VAI transcription by RNA polymerase III. Approximately 2 uL of HeLa cell nuclear extract (approx. 7.5 ug/uL) was incubated with 250 and 1000 ng of GST-RB (379-928) (lanes 2- 3), GST-RB (379-870) (lanes 4-5), GST-RB (379-772) (lanes 6-7), GST-RB (379-577) (lanes 8-9) or GST protein (lanes 10-11) at 30°C for 30 minutes. Lane 1 shows the level of transcription with the untreated extract. Correctly initiated transcripts from the U6 promoter (labeled U6 5') were detected by RNase T1 protection essentially as described in Chapters 2 and 3 (20). In vitro transcription of the Ad VAI gene (bottom) was then initiated by addition of template, cold rNTPs, [a32P]CTP, and transcription buffer. Sample handling was monitored by a non-specific RNA handling control transcript (bottom panels). 136 l naessrryr primers. 1 mm 118 (u. 111 f. (”til .1. (1 contain tea a rains rpm promoters. {NA Infill lucla' 3131 93311.1“? RB (3 59'? 10115 1116155 from {if I 1" as 1159’?” ml “'1‘ 5"” . .5, iptlon ll“ .41 [ml Ina-:- GST o In I GST-BB (379-928) GST a '9. GST-RB(379-870) l I F GST o lo GST-FIB (379-772) 1 I esr GST-RB (379-577) GST l GST \ \ \ \ eta stat \ \ K \ «eeee “96‘9“ 6.3" «5‘ 65‘ #90 e o o o 7‘ 123456 U6 snRNA W AdVAI 3W GST-FIB GST-FIB GST-RB GST-FIB (379-928) (379-870) (379-772) (379-577) 5 Handling Control Ad VAI DEED-Inum-QIWNW ' Control 137 and extract treated with a molar equivalent of GST (lane 10). Removal of the extreme C- terminus (GST-RB (379-870)) results in lowered, but still significant repression (lanes 4- 5) of U6 snRNA and VAI gene expression. However, deletion of the C-pocket (GST-RB (379-772)) results in abolition of RB’s repressor ability (lanes 6-7) even though A/B pocket integrity should still be maintained in this mutant. In fact, genes transcribed by RNA polymerase II have been shown to be regulated by RB truncation mutants containing only amino acids 379-772 (3, 11). Similarly, the A pocket alone (GST-RB (379-577)) was unable to repress U6 or VAI gene expression. Together, these results suggest that the NB pocket as well as the C region of RB is necessary for repression of RNA polymerase III repression. Interestingly, this corresponds to the region of RB that is required for grth suppression. RB occupancy of RNA polymerase III promoters in vitro and in vivo To ascertain whether the ability to repress transcription correlates with the ability to occupy a target RNA polymerase III transcribed promoter, cross-linked immuno- precipitations were performed from the transcription reactions used in the previous figure. In vitro U6 transcription reactions were performed with HeLa cell nuclear extracts that were either left untreated or treated with GST-RB mutants or GST. Portions of these reactions were analyzed by RNase protection assays for full-length transcripts driven from the U6 promoter (see above) while the remainder was subjected to formaldehyde cross-linking followed by affinity purification, using glutathione sepharose to isolate the RB mutant proteins. The precipitates were then analyzed for the presence of the U6 reporter construct (pU6/Hae/RA.2) or the Ad VAI reporter construct (M13-Ad VAI) 138 Figure 4-2: RB occupancy of RNA polymerase III transcribed promoters in vitro and in viva (A) GST-pull down experiments were performed from the crosslinked U6 5’ transcription reactions in figure 4-1D. Associated plasmid DNA was detected by PCR using primers specific to U6 promoter. (B) GST-pull down experiments were performed from the crosslinked Ad VAI transcription reactions in figure 4-1D. Associated plasmid DNA was detected by PCR using primers specific to M1 3-Ad VAI promoter. (C) Chromatin immunoprecipitation experiment to determine RB occupancy of class 2 and class 3 promoters in viva. Chromatin immunoprecipitation experiments were performed similar to those described in Chapter 3. A ten fold serial dilution of input chromatin from 10% to 0.01% is shown in lanes 1-4. Immunoprecipitation reactions were set up using pre-immune sera (lane 5), or-SNAP43 (lane 6), a-RB (lane 7), or or- TBP (lane 8). Recovered DNA was analyzed using primers specific to U2 and U6 snRNA promoters, tRNA (lys), and GAPDH. Part C of this figure was performed by Gauri Jawdekar. 139 m R m 6 U A .095 oz , 50 535 $58 352.9 9550 65.29 magma 88-2.8 9.68 33.5 Input lpUG-Hae-RAZ 12345678910 AdVAI seam. oz, .60 £3-29 magma $.35 mmemo 83.29 938 88-2.8 magma Baas: Input J M13 -Ad VAI 12345678910 mate am...-e 932sz 09-5 Input E-- —- -— - -]U6snRNA - - - fluzsnRNA h--~ - ~—ltRNAIys J GAPDH 2345678 1 140 using primers specific to each of these plasmids. As shown in Figure 4-2A, GST-RB (3 79— 928) exhibits significant amounts of U6 promoter occupancy compared to untreated, GST treated or reactions containing no nuclear extract. This result correlates to its ability to completely repress transcription. Additionally, GST-RB (3 79-870) also associates with the U6 reporter construct, although to a lesser extent, suggesting that the lowered ability to repress transcription may be due to a lowered affinity for the U6 promoter. Again this result suggests an important role for the extreme C-terminus of RB in the regulation of U6 snRNA gene expression. Interestingly, GST-RB (379-772) can also occupy the U6 reporter construct promoter to some extent, even though it does not repress transcription, indicating that recruitment to the promoter may not be enough for repression and that there may be additional co-factors required to repress transcription. These potential co- repressors may require the intact C pocket region of RB to be recruited to the U6 promo ter. The RB mutant containing amino acids 379-577 did not appear to support Significant promoter occupancy similar to its inability to repress transcription. Surprisingly, none of the RB mutants occupied the Ad VAI reporter constructs even though they were capable of repressing transcription to varying extents (Figure 4-2B). This t‘esult implies that RB may have fundamentally different mechanisms for regulating the e"EIDIession of class 2 versus class 3 RNA polymerase III transcribed genes. T0 fl-ll‘ther define the mechanisms used for repression of class 2 and class 3, the in vivo RB 0Cczupancy of a tRNA(lys) and a U6 snRNA promoter were analyzed by chromatin imuno131*ecipitation. Normal human mammary epithelial cells (184B5) were treated with formal(idehyde to covalently cross-link proteins to DNA and other neighboring proteins. 141 Subsequently, lysates were prepared and sonicated to fragment genomic DNA into segments approximately 500-800 bp in size such that there should not be more than one promoter per segment. Lysates were then subjected to immunoprecipitation using antibodies directed against SNAP43, RB or TBP, and the resultant protein-DNA complexes were affinity purified using protein-G agarose beads. The recovered DNA segments were analyzed by PCR using primers specific to human tRNA, U2 and U6 snRNA promoters as well as primers specific to GAPDH exon 2 as a negative control. A ten fold serial dilution of input chromatin was also analyzed for each gene of interest to serve as a standard curve, allowing estimation of relative levels of promoter DNA precipitated and to illustrate that PCR conditions for each set of primers was in the dynamic range. As shown in Figure 4-2C, immunoprecipitations performed with on- SNAP43 antibodies were significantly enriched for U6 snRNA and U2 snRNA promoter DNA but not tRNA(lys) or GAPDH as compared to lgG precipitations as expected (23). Similarly, precipitations performed with a—TBP antibodies were enriched for U6 snRNA, U2 snRNA, and tRNA(lys) promoter DNA as has been shown previously (23). As observed previously, immunoprecipitations performed using Ot-RB antibodies were enriched for U6 snRNA promoter DNA but not U2 or GAPDH promoter DNA. Interestingly, the oc-RB immunprecipitations were not enriched for tRNA promoter DNA, correlating to the results seen in the in vitro assays shown above. Together, these results suggest that RB may regulate RNA polymerase III gene expression by two different mechanisms. 142 Characterization of the region of RB interacts with SNAPc bound to the PSE In order for RB to efficiently regulate U6 expression, RB may be directed to U6 promoters either by direct binding to specific DNA control elements or through recruitment by other trans-acting factors. Interaction between RB and SNAPc has been shown previously, and therefore, it is possible that RB could be recruited to human U6 snRNA promoters through interaction with SNAPc. To determine whether RB can interact with SNAPc bound to DNA, electrophoretic mobility shift assays (EMSA) were performed. It should be noted here that these assays were performed using a much different gel system as compared to chapter 3. This tris-glycine gel system is optimized for the best possible SNAPc-RB interaction, all other gel conditions did not support SNAPc and RB interaction. The reason that this type of gel system was not used in chapter 3 is that the TFIIIB complexes will not form. For these experiments, a mini- SNAPc (12) was assembled fiom recombinant proteins (SNAP43, SNAPSO, and SNAP190 (1-505)) and was incubated with a radio-labeled probe containing either a high affinity or mutant PSE in the presence and absence of GST-RB (379-928). Since RB interacts efficiently with both SNAP43 and SNAPSO (7), a complex containing these factors could suffice to recruit RB to the U6 promoter. As shown in Figure 4-3, DNA binding reactions performed with increasing amounts of GST-RB (379-928) result in a protein-DNA complex with slower mobility than that observed for reactions containing only mini-SNAPc (compare lanes 4-6 to lane 3), suggesting that GST-RB (379-928) is forming a complex with mini-SNAPc on DNA. In contrast, addition of a GST control protein had no effect on DNA binding by mini-SNAPc (lanes 8 -lO). In all cases, DNA binding is dependent upon the PSE because no binding of any type is observed for 143 Figure 4-3: SN APc can recruit RB to promoter DNA. Left panel: Electrophoretic mobility shift assays were performed with purified recombinant mini-SNAPc alone (lane 3) or with mini-SNAPc plus 300 ng, 1000 ng and 3000 ng GST-RB (379-928) (lanes 4 -7) or GST (lanes 8-1 I). Radio-labeled DNA probe containing either wild type or mutant PSE was then incubated with each reaction. The resulting protein-DNA complexes were resolved by 5% acrylamide gel electrophoresis and were visualized by autoradiography. Addition of RB to reactions containing SNAPc results in formation of a lower mobility complex (lanes 4-6) that is PSE specific (lane 7) whereas GST does not affect SNAPc/DNA binding. Addition of GST-RB also results in increased SNAPc binding activity whereas GST alone does not. As shown in lanes 12 and 13, RB alone does not bind to DNA. GST also does not bind DNA (lanes 14 and 15). Right Panel: A cross titration of GST-RB (379-928) and GST was performed to illustrate that the increased DNA binding activity observed in the left panel is a specific effect of GST-RB and not a non-specific protein effect. 144 vaFmFNFSOF a o x. o n v m N_. n m m V n N? . 099m moi l—I onzzw I 1...... ‘3 1.... Hmm+oa>=z§=zi§=2 1332.2, mwm Ben. 0n_90% homogeneity whereas SNAPSO contained at least three major contaminating proteins. These purified proteins were assembled into a mini-SNAPc complex and fiirther purified by anion exchange chromatography to remove contaminating proteins and unincorporated subunits. The resulting fractions were analyzed for SNAPc activity by EMSA and analyzed for purity by SDS-PAGE analysis. Figure A-IA illustrates the chromatogram associated with SNAPc purification over a mono-Q column. Protein concentration was monitored by UV absorbance at both 280 and 220 nm wavelengths. Figure A-IB shows the SDS-PAGE analysis of the fractions illustrating the removal of contaminating bands in fractions 20 to 22 as compared to the starting material. As shown in Figure A-lC, SNAPc activity, as monitored by DNA binding, peaks in fractions 20 to 22. 180 q? Figure A-l: SNAPc assembly and binding to U6 snRNA promoter DNA in vitro: (A) Chromatograph of protein fractions obtained during column chromatographic purification of SNAPc. (B) SDS-PAGE of SNAPc starting material and eluted column 1 fractions. (C) EMSA analysis for SNAPc activity. 181 combat 383888888828:88 o o e «.5 .. 1; 41:3 ::1 E; _ 1: 2.. 2.21 _ .1 Q1,“ Il.lmw-._M 11 .o W m _ 2 gr 8.. 1.8“ 8.. m :8” av .. 200'” W on: comm. W 2 com“ 8 .. :02» 2... , w . :8» 8 -. . M . $8 . coo. 8:11.111: 8: o2... “ u . u a . . a . 82 ...: 88.8 80.5 08.8 8 8.8. 8.8 .38 .88 «8.3 «8.2 32.02.80' :8 >31.- 8« 3|— A co__._mo£5n_ Cocos. od