nannies. r : .nw. 3:2.” thfirush.nuahr . arm at! ; L « . 4,... . « r. 4.,au...uoé. mung an 1 E u , . (Finn-uni £3". LIBRARY Michigau State University This is to certify that the dissertation entitled Regulation of human small nuclear RNA gene transcription by the tumor suppressor protein p53 presented by Anastasia Alekseevna Gridasova has been accepted towards fulfillment of the requirements for the Ph.D. degree in Biochemistry and Molecular Biology 46mm Major Proffi/sor’s Signature /ML/05/ Date MSU is an Affirmative Action/Equal Opportunity Institution --—._.-t-.—- ----c-a-u-o---o--n-o-.-------- _-- _.-.-.-.-o-.—.--._-+_-t.—~— PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 2/05 p:/CIRC/DateDue.indd-p.1 REGULATION OF HUMAN SMALL NUCLEAR RNA GENE TRANSCRIPTION BY THE TUMOR SUPPRESSOR PROTEIN P53 By Anastasia Alekseevna Gridasova A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Biochemistry and Molecular Biology 2005 ABSTRACT REGULATION OF HUMAN SMALL NUCLEAR RNA GENE TRANSCRIPTION BY THE TUMOR SUPPRESSOR PROTEIN P53 By Anastasia Alekseevna Gridasova Activation of the tumor suppressor protein p53 or loss of the Cockayne syndrome complementation group B (CSB) protein induces fragile site formation at RNA polymerase II-transcribed U1 and U2 snRNA gene loci and at RNA polymerase III- transcribed SS rRNA gene loci. Yu et a1. (2000) hypothesized that p53 interferes with transcription elongation functions of CSB, resulting in accumulation of stalled RNA polymerase and impaired chromatin condensation at these gene loci. However, a role for p53 and CSB in transcription of these genes has not been investigated. In this study I show that both p53 and CSB are involved in human U1 snRNA and SS rRNA gene transcription. I found that p53 represses U1 snRNA and SS rRNA gene transcription by RNA polymerases II and III, respectively. p53 also represses U6 snRNA gene transcription by RNA polymerase 111. Both DNA binding competent and defective forms of p53 revealed similar levels of snRNA promoter occupancy during transcription repression, suggesting that sequence-specific DNA binding by p53 is not essential for repression of snRNA gene transcription. I further demonstrated that CSB plays a positive role in snRNA gene transcription by both polymerases II and III and a negative role in transcription of those other classes of RNA polymerase III-transcribed genes that contain an intragenic arrangement of promoter elements. The functional interplay between p53 and CSB in snRNA gene transcription was also investigated. Firstly, removing CSB from cell extracts modulates p53 transcription activity in vitro. CSB immunodepletion potentiates the inhibitory effect of p53 on U1 snRNA gene transcription, but does not affect p53-mediated repression of U6 snRNA gene transcription. Interestingly, at low amounts p53 activates and at higher amounts represses SS rRNA gene transcription when transcription is performed with CSB depleted extracts. Secondly, CSB association with snRNA gene promoters was diminished after UV light treatment concomitant with increased p53 promoter association. As CSB was described as an elongation factor for RNA polymerase 11, p53 may affect elongation by interfering with CSB promoter association. Thirdly, transient transfection of p53 results in snRNA gene transcription repression concomitant with accumulation of covalently modified forms of RNA polymerase 111. These forms of RNA polymerase III are more enriched in CSB cells, suggesting that p53 and CSB have opposing roles in post- translational modifications of RNA polymerase III. I speculate that p53 represses elongation by RNA polymerase III by facilitating post-translational modifications of the polymerase. Together, these results suggest that p53 may modulate CSB transcriptional activity and support the hypothesis that fragile sites at U1 and U2 snRNA and SS rRNA gene loci may be caused by the inhibitory effect of p53 on CSB-mediated elongation by RNA polymerases. ACKNOWLEDGEMENTS I would like to express my sincere gratitude to my mentor R. William Henry, whose knowledge and guidance helped me during the years of my dissertation research. I would also like to thank the members of my thesis committee, Dr. David Amosti, Dr. Zachary Burton, Dr. John LaPres and Dr. Richard Schwartz for their guidance during these years. I am also grateful to Dr. David Amosti and Dr. Zachary Burton and also Dr. James Geiger and Dr. Min-Hao Kuo for helping me to secure my post-doctoral position. Additionally, I would like to thank Dr. Min-Hao Kuo and Dr. Asha Acharya for an interesting and productive collaboration. I would like to thank the current and former members of the Henry lab: Craig Hinkley, Zakir Ullah, Heather Hirsch, Liping Gu, Xianzhou Song, Melissa Bosma, and especially Gauri J awdekar and Tharakeswari Selvakumar for all their help and friendship. Also, I would like to thank Dr. Zeikus and the members of his laboratory, especially Harini Krishnamurty for her friendship and Jake McKinlay for his support and for being a very special person in my life. Additionally, I would like to thank Dr. Scott Hill and his family (wife Peggy and daughter Heather) for encouraging me to come to study in the US, welcoming me to Michigan, and helping me over these years. Finally, I would like to thank my mom for her love and support. iv TABLE OF CONTENTS LIST OF FIGURES ........................................................................................................ vii KEY TO SYMBOLS AND ABBREVIATIONS ........................................................... ix CHAPTER 1: Introduction ....................................................................................................................... l 1. Human small nuclear RNA ............................................................................................ 1 1.1. Diverse functions in cells ............................................................................... .l 1.2. Promoter organization of human snRNA genes ................................... 2 1.3. The General Transcriptional Machinery ............................................ 5 1.4. Regulation of human snRNA gene transcription .................................. 8 2. Tumor suppressor protein p53 .................................................................. 8 2.1. Discovery of p53 .............................................................................................. 9 2.2. Cellular functions of the p53 tumor suppressor protein ................................... 9 2.2.1. Cell cycle control ......................................................... 10 2.2.2. Apoptosis ........................................................................................ 10 2.2.3. DNA repair ...................................................................................... 11 2.2.4. Cellular response to p53 and p53 protein partners .......................... 12 2.3. p53 as a transcriptional factor ........................................................................ 13 2.3.1. Role of p53 in RNA polymerase I, II and III transcription ............. 13 2.3.2. Possible mechanisms of the transcriptional control by p53 ............ 14 2.4. p53 stability, cellular stress and post-translational modifications ................. 16 2.5. Structure of the p53 protein ........................................................................... 19 2.5.1. Crystal structure of the p53 domains .............................................. 19 2.5.2. DNA binding activity ...................................................................... 22 2.6. The p53 family ............................................................................................... 23 2.6.1. p53/p63/p73 ............................................................... 23 2.6.2. alternative forms of p53 .................................................. 24 3. Cockayne Syndrome factor B protein ......................................................... 25 3.1. Functional interplay with p53 ...................................................... 25 3.2. Functions of CSB ..................................................................... 26 3.3.1. Role in DNA repair ....................................................... 26 3.3.2. Role in transcription regulation ......................................... 27 3.3. Structure of CSB ..................................................................... 28 3.4. Mutant CSB protein and Cockayne syndrome .................................... 31 CHAPTER 2: The p53 tumor suppressor protein represses human snRNA gene transcription by RNA polymerases II and [11 independently of sequence-spesific DNA binding ...................................................................................... 47 Abstract ................................................................................................................. 47 Introduction ........................................................................................................... 48 Materials and Methods .......................................................................................... 51 Results ................................................................................................................... 57 Discussion ............................................................................................................. 91 References ............................................................................................................ 95 CHAPTER 3: The Cockayne syndrome complementation group B protein modulates transcription regulatory functions of the tumor suppressor protein p53 ................ 102 Abstract ............................................................................................................... 102 Introduction ......................................................................................................... 103 Materials and Methods ........................................................................................ 106 Results ................................................................................................................. 1 10 Discussion ........................................................................................................... 134 References ........................................................................................................... 140 CHAPTER 4: Summary ........................................................................................................................ 144 Summary ............................................................................................................. 144 References ........................................................................................................... 149 APPENDIX: Possible role of HDACs in p53-mediated snRNA gene transcription repression....151 Results and Discussion ....................................................................................... 151 Materials and Methods ........................................................................................ 162 References ........................................................................................................... 166 vi Figure l-l. Figure 1-2. Figure 1-3. Figure 1-4. 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. LIST OF FIGURES Schematic representation of RNA polymerase II- and III-transcribed promoters ............................................................................ 4 Schematic representation of the general transcription machinery for RNA polymerase II- and III- transcribed genes 7 Schematic representation of the structure of p5 3 and ANp53 proteins and the consensus DNA binding sites .......................................... 21 Predicted motifs of CSB ......................................................... 30 p53 represses human snRNA gene transcription by both RNA polymerases II and III in vitro ................................................... 59 UV light inhibits snRNA gene transcription and stimulates p53 binding to human snRNA gene promoters .............................................. 63 Human Ul snRNA gene core promoters contain a high-affinity p53 binding site .................................................................... 72 The high affinity p53 element in the U1 promoter is not essential for p53 repression in vitro ........................................................ 77 The p53 C terminus is sufficient for transcriptional repression and promoter association .............................................................. 83 Endogenous p53 associates with the general transcription factor SNAPc ....................................................................... 88 Endogenous CSB associates with RNA polymerase II-transcribed snRNA gene promoters and RNA polymerase III-transcribed promoters ......................................................................... 112 Endogenous CSB associates with RNA polymerase III ................... 115 CSB regulates snRNA gene transcription .................................... 119 UV light exposure affects p53 and CSB occupancy on snRNA gene promoters in vivo ................................................................. 123 p53 affects CSB and RNA polymerase III occupancy on U6 snRNA gene promoters in vitro. 125 vii Figure 3-6. Figure 3-7. Figure 3-8. Figure A-l. Figure A-2. Figure A-3. Figure A-4. Loss of CSB accentuates the RNA polymerase II and III transcriptional response to p53 ................................................. 128 CSB modulates p53-dependent modifications of RNA polymerase IH ............................................................. 131 Model: Roles of p53 and CSB in RNA polymerase III elongation and stability ....................................................................... 138 Endogenous HDACl and HDAC2 associate with human snRNA gene promoters .......................................... 153 Sumoylated protein population is enriched on U1 snRNA gene promoters afier UV light exposure ....................... 156 p53 sumoylation contributes to p53-mediated snRNA gene transcription repression in vitro ................................ 159 Sumoylated p53 is enriched in acetylated population of p53 in vivo ...................................................................... 161 viii ASSP ATM Brfl -TFIIIB Brfl-TFIIIB CDK CHI-I ChIPs CKII CO-IP CS CSB CTD C-terminal Da DBD ds DSE ELL EMSA GAPDH HAT KEY TO SYMBOLS AND ABBREVIATIONS apoptotic-stimulating proteins of p53 ataxia telangiectasia mutated kinase TFIIIB complex composed of TBP, Brfl and del TFIIIB complex composed of TBP, 8er and del cyclin-dependent kinase cartilage-hair hypoplasia chromatin immunoprecipitations casein kinase II co-immunoprecipitation Cockayne syndrome the Cockayne syndrome complementation group B protein carboxy terminal domain of RNA polymerase II largest subunit carboxy-terminal Dalton DNA binding domain double stranded distal sequence element the Eleven Lysine-rich Leukemia protein electrophoretic mobility shift assay glyceraldehyde-3-phosphate dehydrogenase histone acetyl transferase ix HDAC Hdm2 IE JMY Mdm2 mRNA NLS NTB N-terminal PSE P-TEFb PTF RB RD RNAP RPA rRNA RT-PCR SNAPC snRNAs histone deacetylase the human double minute protein intermediate element immunoprecipitation junction mediating and regulatory protein the murine double minute 2 protein messenger RNA nucleotide excision repair nuclear export signal nuclear localization signal nucleotide binding domain amino-terminal proximal sequence element positive transcription elongation factor-b proximal sequence element transcription factor the retinoblastoma tumor suppressor protein the regulatory domain RNA polymerase the replication protein A ribosomal RNA reverse transcriptase polymerase chain reaction the snRN A activating protein complex small nuclear RNAs snRNPs SS SUMO SV40 TAD TAF TBP TCR TD tRN A small nuclear ribonucleoprotein particles single stranded the small ubiquitin—like modifier Simian virus 40 large T antigen the transactivation domain TBP-associated factor TATA binding protein transcription coupled repair the tetramerization domain transfer RNA ultraviolet light xi CHAPTER 1 INTRODUCTION 1. Human small nuclear RNA 1.]. Diverse functions in cells Small nuclear (sn) RNAs are short, stable, nontranslated RNAs that are evolutionarily well conserved and found in the nuclei of all eukaryotic cells. These RNAs are not polyadenylated and contain unusual 5’ cap structures. They exist in the cell packaged with proteins as small nuclear ribonucleoprotein particles (snRNPs). Newly synthesized snRNAs are immediately exported to the cytoplasm where they undergo maturation and assembly into snRNPs prior to their reentry into the nucleus (55). SnRNA perform essential functions in cells. As part of snRNPs, uridine rich snRNAs are involved in messenger (m) RNA splicing (e.g. U1, U2, U4, U5 and U6) and ribosomal (r) RNA processing (e.g. U3) (55). It is estimated that up to 15% of all point mutations causing human genetic disease result in an mRNA splicing defect (70, 138). In addition to their roles in RNA metabolism, some snRNAs also participate in regulation of transcription initiation (e.g. U1 snRNA and BZ RNA) or transcription elongation (e.g. 7 SK and components of the splicing apparatus) through association with transcription factors (36, 39, 76, 108, 153). U1 snRNA associates with TFIII-I and stimulates transcription initiation in vitro (76). In contrast, B2 RNA represses transcription initiation when it binds to RNA polymerase 11 upon heat shock of mouse cells (36). Repression of transcription elongation by 7SK RNA occurs through the inhibition of the kinase activity of the positive transcription elongation factor (P-TEFb) (108, 153). Recent evidence suggests additional unexpected snRNA cellular functions in innate immunity (58), human hereditary diseases (e.g. cartilage-hair hypoplasia (CHH) disease) (116) and cancer (70, 138). I . 2. Promoter organization of human snRNA genes Human snRNA genes are transcribed by either RNA polymerase II (e.g. U1 snRNA gene) or by RNA polymerase III (e. g. U6 snRNA gene) and yet have very similar promoter architecture (Figure 1—1). Both RNA polymerase II- and III- transcribed snRNA genes contain the proximal sequence element (PSE) within the core promoter region. This essential element is located at approximately —45 bp upstream of the start site of transcription. In addition, the RNA polymerase III-transcribed snRNA genes contain a TATA box located in the core promoter adjacent to the PSE. In humans, the TATA box acts as dominant element for determining the specificity of RNA polymerase III- transcribed snRNA gene promoters (56, 75, 88). Both the RNA polymerase II and III snRNA gene promoters contain the distal sequence element (DSE), located in the regulatory region around —220 (105). The DSE functions as an enhancer and is required for maximum promoter activity. As shown in Figure 1-1, human U6 snRNA genes are distinct from other RNA polymerase III-transcribed genes. U6 snRNA genes belong to class 3 promoters, which are defined by their extragenic RNA polymerase III promoters. In contrast, the class 1 and 2 RNA polymerase III-specific genes contain intragenic promoter elements and are Figure 1-1. Schematic representation of RNA polymerase II- and III- transcribed promoters. Class 1 and 2 RNA polymerase III-transcribed promoters are intragenic and are exemplified by the SS rRNA and tRNA promoters, respectively. Class 1 genes contain A and C boxes that are separated by an intermediate element (IE). Class 2 promoters consist of A and B boxes. Class 3 promoters are defined as extragenic RNA polymerase III promoters and are exemplified by the human U6 snRNA promoter. The architecture of the class 3 RNA polymerase III-transcribed snRNA gene promoters is similar to other snRNA gene promoters that are transcribed by RNA polymerase 11 (e. g. U1 snRNA gene). Both RNA polymerase II- and III— transcribed snRNA gene promoters contain the proximal sequence element (PSE) and the distal sequence element (DSE). In addition, class 3 RNA polymerase III-transcribed promoters have a TATA box. =m>OmmE 30mm: 9.1 A”. l I «32350 >>>o>> Somme 3.32 553 Bommm 35%! :5 3a 21 L3 and R273 within the C-terminal or-helix) contact DNA directly. The other four (R175, G245, R249 and R282) stabilize the surrounding protein structure (19, 149). The TD is represented by a B-strand linked to an or-helix. A TD monomer has a V-shape. Two monomers form a dimer through interactions between the B-strands and the or-helices arranged in an anti-parallel fashion. The interactions that hold two monomers together as dimers are mainly provided through hydrogen bonds in the B-strands and hydrophobic interactions from both the B-strands and the or-helices. Two dimers are held together as a tetrarner by a large hydrophobic surface of each or-helix (25). 2. 5. 2. DNA binding activity As shown in figure 1-3B, the consensus p53 DNA binding site consists of 2 half sites, each comprised of two copies of the sequence 5’-Pu-Pu-Pu-C-AfT-3’ (Pu is purine), arranged head-to-head (for p53 acting as a transcriptional activator) or head-to-tail (for p53 acting as a transcriptional repressor), and separated by 0 to 14 nucleotides (33, 64). Some variations within consensus PuPuPuC(A/T) sequence are also permissible (115). These specific cognate sites can bind tightly to the DBD of p53. It was suggested that one p53 DBD dimer binds first to one half of the consensus DNA-binding site, increasing the probability for the binding of the second p53 dimer to the adjacent half of the site (95, 96). In addition to binding specifically to DNA at p53 consensus sites, p53 also binds non-specifically to ssDNA, nicked DNA, damaged DNA with ds breaks, and DNA with Holliday junctions (87). These DNA structures represent the intermediates of DNA damage and repair. Binding to non-specific DNA was primarily mapped to the C- terminal domain of p53 (112, 159). It was generally accepted that the C-terminus of p53 22 is a negative regulator of p53 sequence-specific DNA binding. Several groups have demonstrated that various alterations of the C-terminal domain (deletion, post- translational modifications and interaction with antibodies directed at a C-terminal epitope) result in an increase of p53 DNA binding (46, 62, 120). However, more recent studies have shown that p53 binds its target sites in vitro and in vivo in the absence of DNA damage or extensive modifications of the C-terminus (6, 35) and the C-terminal- deleted p53 is substantially less efficient at binding and transactivation of its targets in vivo (94). It suggests that the C-terminus does not maintain p53 in a state that is inactive for DNA binding, but rather that it is required for efficient binding of p53 to its target promoters. 2. 6. T he p53 family 2.6.1. p53/p63/p73 Since the discovery of p53 in 1979, two more members have been added to the p53 superfarnily, p63 and p73. The p53 family members share very significant homology both at the genomic and at the protein levels (152). Each contains a TAD, a DBD, and TD. In addition, p63 and p73 contain long C-termini. As a result of the alternative splicing of their C-termini, three p63 isoforms (or to y) and seven p73 isofonns (or to n) were identified. Additional complexity is also achieved because these isoforrns (called the TA and AN isoforrns, respectively) are transcribed from the upstream promoter as well as from a cryptic promoter within intron 3 (152). The highest level of homology between the p53 family of proteins is reached in the DBD, which suggests that the three proteins can bind to the same DNA sequence and regulate the same genes. Indeed, p63 23 and p73 bind to p53 consensus DNA elements and trans-activate certain p53 target genes (38). p63 and p73 may also inhibit the transcriptional activity of p53 by competing for the same binding sites on DNA (38). High conservation of the TD sequence between p53 family members results in formation of hetero-oligomers as well as homo-oligomers. ANp63 and ANp73 forms lack TAD and may inhibit p53 in a dominant-negative fashion through forming hetero-complexes with p53 (101). Unlike p53, the genes encoding p63 and p73 are rarely mutated in human cancer. The phenotype of the p63- and p73- deficient mice suggests that the primary biological function of these proteins is to regulate development, which is in contrast to p53-null mice, which are highly tumor prone but lack a developmental phenotype (101). 2. 6. 2. Alternative forms of p53 The human ANp53 and its murine counterpart p44 are naturally occun'ing isoforrns of p53 (26, 91, 127). ANpS3 is encoded by the p53 locus, but uses an alternative translation start site located in exon 4 at codon 40 in human RNA. The resultant 44 kDa protein lacks the corresponding N—terminal amino acids (Figure 1-3A). Choice of start site depends on an interaction between p53 and its cognate RNA, which requires the N- terrninal domain of p53 and its DNA binding domain. The complex of the p53 protein with newly synthesized RNA prevents the ribosome at ATG (codon 1) from being activated and translating full-length p53. Instead, translation of ANp53 by the ribosome at ATG (codon 41) occurs. The RNA — p53 protein complex is destabilized by Mdm2, which competes with RNA for binding to N-terminus of p53 (127). Since the ANpS3 24 lacks the Mdm2 binding site, it is not a subject to Mdm2-mediated degradation. This results in a prolonged half-life of ANpS 3. ANp53 is severely compromised in ability to activate target genes due to complete absence of N-terminal TAD that binds to the basal transcription machinery. Since ANpS3 is able to hetero-tetramerize with full length p53, the ratio of fiill-length to ANp53 can determine cellular functions of p53. At low levels, ANp53 would be exclusively found in tetramers with full—length. At high levels, however, excess short form would also be present as homo-tetramers or even as non-tetrameric forms, such as monomers or dimer, which could have several different effects (127). Homo- and hetero- tetramers of ANp53 would be severely compromised in their ability to activate target genes due to complete absence of N-terminal TAD in ANpSB (97). Also, non-tetrarneric forms of ANp53 could replace p53 in transcription-independent activities, such as mitochondrial cytochrome c release, altering the ability of p53 to mediate apoptosis (97). 3. Cockayne Syndrome factor B protein 3.]. Functional interplay with p53 Several studies demonstrated that infection of human cells by Adenovirus type 12 induces specifically four fragile sites including efficiently induced fi'agility of the U1 and U2 snRNA gene loci and U1 snRNA pseudogenes in several different cell lines and weakly induced fragility of the SS rRNA gene in primary human embryonic kidney cells (30, 41, 84, 125). Formation of the same fragile sites were also observed after treatment of cells with actinomycin D or cytosine arabinoside C (83, 90). All these factors are known to activate p53 and do not induce fragility in cells lacking firnctional p53 (83). In 25 addition, these fragile sites can be induced by over-expression of the full length wild type p53 or just the C-terminal domain of p53 alone (155). Together, these observations suggest that p53 plays a direct role in fragile site formation at U1 and U2 snRNA and 5S rRNA genes, and the C-terminus of p53 is required for p53-mediated induction of fragile sites. Interestingly, loss of functional CSB in cells also results in fragile site formation at the U1 snRNA, U2 snRNA and SS rRNA gene loci (155). The requirement of p53 activation or loss of CSB activity for fragile site formation at these loci suggests a possible role for these factors in controlling transcription of these genes, even though U1 and U2 snRNA genes are transcribed by RNA polymerase II and SS rRNA genes are transcribed by RNA polymerase III. Studies by Selby and Sancar (1997) suggested a role of CSB for elongation of gene, encoding highly structured RNAs (such as U1 and U2 snRNAs) (128). Interestingly, CSB has been shown to interact with p53 within the C- terrninal region (147, 155). It was suggested that p53 binding to CSB may interfere with CSB activity; thus, mimicking loss of CSB (155). Yu et al. (2000) proposed that p53 may repress RNA polymerase II transcribed snRNA genes by interfering with Cockayne Syndrome group B (CSB)-mediated elongation, causing RNA polymerase II stalling at U1 and U2 snRNA genes. Stalled RNA polymerase protein complexes may interfere with local chromatin condensation, causing locus-specific chromosome fragility (155). 3.2. Functions of CSB 3.2.]. Role in DNA repair The CSB protein was originally identified as a DNA repair protein and it was cloned on the basis of its ability to complement the transcription coupled repair (TCR) 26 defect in CS cells (142, 145). It has been proposed that CSB recruits the NER apparatus to sites of stalled RNA polymerase II to permit rapid repair (145). CSB is also critical for the repair of nucleotide base damage induced by reactive oxygen species when such lesions are located on the transcribed strand of active genes (80). CSB chromatin remodeling activity may be required to open chromatin around lesions, thereby stimulating repair. In addition, the CSB protein may also play a role in clearing the stalled RNA polymerase II molecule from the lesion site so that repair can occur and transcription resume (49). The observed interaction of CSB with RNA polymerases I and 11 both in vivo and in vitro might specifically target CSB to sites of blocked transcription (11). 3.2.2. Role in transcription regulation Various in vitro and in vivo experiments point to a possible role for CSB in transcription. CSB was shown to associate with RNA polymerase I and 11 protein complexes (11, 144). Gel mobility shift assays revealed that CSB interacts with a ternary complex of DNA, RNA polymerase II, and nascent RNA (139) and in vitro transcription experiments showed that CSB stimulates elongation by RNA polymerase II (128). It is suggested that CSB functions as an elongation factor to promote transcription of genes encoding RNA products with significant secondary structure, such as human U1 and U2 snRNA genes (155). Additionally, CSB is also implicated in chromatin remodeling during transcription. It was demonstrated that CSB remodels nucleosomes at the expense of ATP hydrolysis and interacts with core histones in vitro (24). Also, CSB can alter DNA double helix 27 conformation upon binding by wrapping DNA using energy from ATP hydrolysis (7). One possibility is that by changing DNA conformation CSB may disrupt the histone- DNA interactions, as well as the interaction of stalled RNA polymerase with damaged DNA. 3.3. Structure of CSB The CSB gene encodes a 168 kDa protein, containing several conserved motifs (142). As illustrated in figure 1-4, the central part of CSB is represented by the seven consecutive ATPase motifs I, la, II, III, IV, V, and VI fi'om amino acids 527-950. These motifs form a nucleotide-binding fold and are conserved among three superfamilies of RNA and DNA helicases. The region of CSB encompassing these motifs is highly homologous to proteins of the SNF2-like family (32). The N-terminus of CSB has an acidic (A) domain (amino acids 356-394) (142). Between the acidic and SNF2 domains is a glycine-rich (G) stretch and a highly hydrophilic region (H). C-terminal to the ATPase motifs is a putative nucleotide-binding (NT B) domain. The protein also contains a bipartite nuclear-localization signal (NLS). Studies by Christiansen et al. (2005) demonstrated that CSB protein functions as a dimer. The homodimerization occurs via the central ATPase domain of the CSB protein and is essential for ATP hydrolysis by CSB (21). Of the seven ATPase motifs, motifs I and 11 contain consensus NTP-binding sequences and are involved in ATP binding by CSB (48). Mutant CSB protein with point mutations in ATPase motifs I and II of CSB cannot complement UV sensitivity, 28 Figure 1-4. Predicted motifs of CSB: A = Acidic domain, G = glycine-rich stretch, NLS = Nuclear localization sequence, NTB = Nucleotide binding motif. 29 £on 8mm? 83 omm mm vmm 8m — mmU .. . . 5:5: _> >>_ 5 __ <_ _ o _|_ [ _ _ _llL Fill. mhz m._z 59:8 859 Sam. Ezm 28892: 222 30 suggesting that ATP hydrolysis by CSB protein is required for TCR of DNA damage (20, 23). Studies on CSB ATPase motif Ia and III mutants revealed only partial complementation of the sensitivity of CS cells towards UV and recovery of RNA synthesis after UV irradiation (104). Studies of homologous proteins suggest the involvement of motifs Ia and III in energy transduction between the ATPase site and the nucleic acid binding site (48). The ATPase motif V and VI CSB mutants exhibit a similar inhibition of ATPase activity and reduced ATP binding in vitro (20). These mutants also have comparable defects in recovery of RNA synthesis after UV irradiation (143). So far, there have been no reported structure-function experiments involving the putative NLS, the glycine-rich stretch, the hydrophobic segment, or the ATPase motif IV. Also, no functions have thus far been assigned to the acidic domain and putative NTP box (13, 137). 3.4. Mutant CSB protein and Cockayne syndrome Transcription-coupled repair (TCR) is a cellular pathway for the removal of the many DNA lesions that block and arrest transcription. TCR is responsible for the rapid and preferential repair of damage in the transcribed strand of active genes (10). It occurs in both prokaryotic and eukaryotic organisms. In humans, the absence of TCR is associated with Cockayne Syndrome (CS). The majority of CS cases are caused by defects in the Cockayne Syndrome complementation group B (CSB) protein (85). CS is a premature aging syndrome with complex symptoms, including developmental abnormalities, neurologic disfunction and a short average life span. Cellular characteristics include hypersensitivity to UV light, and failure of RNA synthesis 31 to recover to normal rates following UV irradiation (93, 107, 145). The photosensitivity of CS patients can be attributed to the TCR defect. It is assumed that accumulation of the DNA damage causes growth arrest and apoptosis, which may explain the progressive course of the disease. Interestingly, CS patients, despite their DNA repair deficiency, do not have a predisposition to cancer. However, CSB deficiency itself has antineoplastic effect in cancer predisposed mice (89). It has been suggested that precancerous cells in CS patients are more efficiently eliminated by apoptosis than they are in healthy person (50, 89). In contrast, neurological and developmental features of this disease can be a result of the problems with transcription (50). Could misregulation of U1 snRNA gene transcription contribute to development of the CS phenotypes? It was shown that in addition to mutations in CSB and CSA genes the CS phenotype can result from mutations in XPB, XPD, and XPG genes (133). The products of these genes were shown to be a part or directly associate with the TFIIH protein complex involved in both basal and activated transcription and NER (85), suggesting that misfunctioning of TFIIH may be a common step in developing the CS phenotypes. Interestingly, U1 snRNA was also shown to associate with TFIIH and stimulate transcription initiation by TFIIH (76). Thus, misregulation of U1 snRNA in CSB deficient cells may result in misfunctioning of TFIH-I similarly to the XBP, ED and HG gene mutations and may contribute to development of the CS disease. Another interesting possibility is that CS patients may additionally experience misregulation of other non- coding RNAs, which may contribute to the pleotropic phenotype of the CS disease. 32 References 10. ll. Abramova, N. A., J. Russell, M. Botchan, and R. Li. 1997. Interaction between replication protein A and p53 is disrupted after UV damage in a DNA repair- dependent manner. Proc Natl Acad Sci U S A 94:7186-91. Akoulitchev, S., T. P. Makela, R. A. Weinberg, and D. Reinberg. 1995. Requirement for TFIIH kinase activity in transcription by RNA polymerase 11. Nature 377:557-60. An, W., J. Kim, and R. G. Roeder. 2004. Ordered cooperative functions of PRMTl, p300, and CARMl in transcriptional activation by p53. Cell 117:735-48. Ard, P. G., C. Chatterjee, S. Kunjibettu, L. R. Adside, L. E. Gralinski, and S. B. McMahon. 2002. Transcriptional regulation of the mdm2 oncogene by p53 requires TRPJXP acetyltransferase complexes. Mol Cell Biol 22:5650-61. Attardi, L. D., E. E. Reczek, C. Cosmas, E. G. Demic‘co, M. E. McCurrach, S. W. Lowe, and T. Jacks. 2000. PERP, an apoptosis-associated target of p53, is a novel member of the PMP-22/gas3 family. Genes Dev 14:704-18. Barlev, N. A., L. Liu, N. H. Chehab, K. Mansfield, K. G. Harris, T. D. Halazonetis, and S. L. Berger. 2001. Acetylation of p53 activates transcription through recruitment of coactivators/histone acetyltransferases. Mol Cell 8:1243- 54. Beerens, N., J. H. Hoeijmakers, R. Kanaar, W. Vermeulen, and C. Wyman. 2005. The CSB protein actively wraps DNA. J Biol Chem 280:4722-9. Bell, 8., C. Klein, L. Muller, S. Hansen, and J. Buchner. 2002. p53 contains large unstructured regions in its native state. J Mol Biol 322:917-27. Blau, J., H. Xiao, S. McCracken, P. O'Hare, J. Greenblatt, and D. Bentley. 1996. Three functional classes of transcriptional activation domain. Mol Cell Biol 16:2044-55. Bohr, V. A., C. A. Smith, D. S. Okumoto, and P. C. Hanawalt. 1985. DNA repair in an active gene: removal of pyrimidine dimers from the DHFR gene of CHO cells is much more efficient than in the genome overall. Cell 40:359-69. Bradsher, J., J. Auriol, L. Proietti de Santis, S. Iben, J. L. Vonesch, I. Grummt, and J. M. Egly. 2002. CSB is a component of RNA pol I transcription. Mol Cell 10:819-29. 33 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. Brooks, C. L., and W. Gu. 2003. Ubiquitination, phosphorylation and acetylation: the molecular basis for p53 regulation. Curr Opin Cell Biol 15: 164-71. Brosh, R. M., Jr., A. S. Balajee, R. R. Selzer, M. Sunesen, L. Proietti De Santis, and V. A. Bohr. 1999. The ATPase domain but not the acidic region of Cockayne syndrome group B gene product is essential for DNA repair. Mol Biol Cell 10:3583-94. Calms, C. A., and R. J. White. 1998. p53 is a general repressor of RNA polymerase III transcription. Embo J 17 :31 12-23. Chehab, N. H., A. Malikzay, M. Appel, and T. D. Halazonetis. 2000. Chk2/thsl fiinctions as a DNA damage checkpoint in G(1) by stabilizing p53. Genes Dev 14:278-88. Chen, X., L. J. Ko, L. Jayaraman, and C. Prives. 1996. p53 levels, functional domains, and DNA damage determine the extent of the apoptotic response of tumor cells. Genes Dev 10:2438-51. Chesnokov, 1., W. M. Chu, M. R. Botchan, and C. W. Schmid. 1996. p53 inhibits RNA polymerase III-directed transcription in a promoter-dependent manner. Mol Cell Biol 16:7084-8. Chipuk, J. E., and D. R. Green. 2004. Cytoplasmic p53: bax and forward. Cell Cycle 3:429-31. Cho, Y., S. Gorina, P. D. Jeffrey, and N. P. Pavletich. 1994. Crystal structure of a p53 tumor suppressor-DNA complex: understanding tumorigenic mutations. Science 265:346-55. Christiansen, M., T. Stevnsner, C. Modin, P. M. Martensen, R. M. Brosh, Jr., and V. A. Bohr. 2003. Functional consequences of mutations in the conserved SF2 motifs and post-translational phosphorylation of the CSB protein. Nucleic Acids Res 31 :963-73. Christiansen, M., T. Thorslund, B. Jochimsen, V. A. Bohr, and T. Stevnsner. 2005. The Cockayne syndrome group B protein is a functional dimer. Febs J 272:4306-14. Chuikov, S., J. K. Kurash, J. R. Wilson, B. Xiao, N. Justin, G. S. Ivanov, K. McKinney, P. Tempst, C. Prives, S. J. Gamblin, N. A. Barlev, and D. Reinberg. 2004. Regulation of p53 activity through lysine methylation. Nature 432:353-60. Citterio, E., S. Rademakers, G. T. van der Horst, A. J. van Gool, J. H. Hoeijmakers, and W. Vermeulen. 1998. Biochemical and biological 34 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. characterization of wild-type and ATPase-deficient Cockayne syndrome B repair protein. J Biol Chem 273:11844-51. Citterio, E., V. Van Den Boom, G. Schnitzler, R. Kanaar, E. Bonte, R. E. Kingston, J. H. Hoeijmakers, and W. Vermeulen. 2000. ATP-dependent chromatin remodeling by the Cockayne syndrome B DNA repair-transcription- coupling factor. Mol Cell Biol 20:7643-53. Clore, G. M., J. Ernst, R. Clubb, J. G. Omichinski, W. M. Kennedy, K. Sakaguchi, E. Appella, and A. M. Gronenborn. 1995. Refined solution structure of the oligomerization domain of the tumour suppressor p53. Nat Struct Biol 2:321-33. Courtois, S., G. Verhaegh, S. North, M. G. Luciani, P. Lassus, U. Hlbner, M. Oren, and P. Hainaut. 2002. DeltaN-p53, a natural isoform of p53 lacking the first transactivation domain, counteracts growth suppression by wild-type p53. Oncogene 21 :6722-8. Crighton, D., A. Woiwode, C. Zhang, N. Mandavia, J. P. Morton, L. J. Warnock, J. Milner, R. J. White, and D. L. Johnson. 2003. p53 represses RNA polymerase III transcription by targeting TBP and inhibiting promoter occupancy by TFIIIB. Embo J 22:2810-20. Dawson, R., L. Muller, A. Dehner, C. Klein, H. Kessler, and J. Buchner. 2003. The N-terminal domain of p53 is natively unfolded. J Mol Biol 332:1131-41. Dornan, D., I. Wertz, H. Shimizu, D. Arnott, G. D. Frantz, P. Dowd, K. O'Rourke, H. Koeppen, and V. M. Dixit. 2004. The ubiquitin ligase COPl is a critical negative regulator of p53. Nature 429:86-92. Durnam, D. M., J. C. Menninger, S. H. Chandler, P. P. Smith, and J. K. McDougall. 1988. A fragile site in the human U2 small nuclear RNA gene cluster is revealed by adenovirus type 12 infection. Mol Cell Biol 8:1863-7. Eichhorn, K., and S. P. Jackson. 2001. A role for TAF3B2 in the repression of human RNA polymerase III transcription in nonproliferating cells. J Biol Chem 276:21158-65. Eisen, J. A., K. S. Sweder, and P. C. Hanawalt. 1995. Evolution of the SNF2 family of proteins: subfamilies with distinct sequences and functions. Nucleic Acids Res 23:2715-23. el-Deiry, W. S., T. Tokino, V. E. Velculescu, D. B. Levy, R. Parsons, J. M. Trent, D. Lin, W. E. Mercer, K. W. Kinzler, and B. Vogelstein. 1993. WAFl, a potential mediator of p53 tumor suppression. Cell 75:817-25. 35 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. Eliceiri, G. L., and J. H. Smith. 1983. Sensitivity to UV radiation of small nuclear RNA synthesis in mammalian cells. Mol Cell Biol 3:2151-5. Espinosa, J. M., and B. M. Emerson. 2001. Transcriptional regulation by p53 through intrinsic DNA/chromatin binding and site-directed cofactor recruitment. Mol Cell 8:57-69. Espinoza, C. A., T. A. Allen, A. R. Hieb, J. F. Kugel, and J. A. Goodrich. 2004. 82 RNA binds directly to RNA polymerase II to repress transcript synthesis. Nat Struct Mol Biol 11:822-9. Farmer, G., J. Colgan, Y. Nakatani, J. L. Manley, and C. Prives. 1996. Functional interaction between p53, the TATA-binding protein (TBP), andTBP- associated factors in vivo. Mol Cell Biol 16:4295-304. Flores, E. R., K. Y. Tsai, D. Crowley, S. Sengupta, A. Yang, F. McKeon, and T. Jacks. 2002. p63 and p73 are required for p53-dependent apoptosis in response to DNA damage. Nature 416:560-4. Fong, Y. W., and Q. Zhou. 2001. Stirnulatory effect of splicing factors on transcriptional elongation. Nature 414: 929- 33. 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. Gargano, S., P. Wang, E. Rusanganwa, and S. Bacchetti. 1995. The transcriptionally competent U2 gene is necessary and sufficient for adenovirus type 12 induction of the fragile site at 17q21-22. Mol Cell Biol 15:6256-61. Gostissa, M., A. Hengstermann, V. Fogal, P. Sandy, S. E. Schwarz, M. Scheffner, and G. Del Sal. 1999. Activation of p53 by conjugation to the ubiquitin-like protein SUMO-1. Embo J 18:6462-71. Gridasova, A. A., and R. W. Henry. 2005. The p53 tumor suppressor protein represses human snRNA gene transcription by RNA polymerases II and III independently of sequence-specific DNA binding. Mol Cell Biol 25:3247-60. Grossman, S. R., M. E. Deato, C. Brignone, H. M. Chan, A. L. Kung, H. Tagami, Y. Nakatani, and D. M. Livingston. 2003. Polyubiquitination of p53 by a ubiquitin ligase activity of p300. Science 300:342-4. Gu, L., W. J. Esselman, and R. W. Henry. 2005. Cooperation between small nuclear RNA-activating protein complex (SNAPC) and TATA-box-binding protein antagonizes protein kinase CK2 inhibition of DNA binding by SNAPC. J Biol Chem 280:27697-704. 36 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. Cu, W., and R. G. Roeder. 1997. Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell 90:595-606. Gu, W., X. L. Shi, and R. G. Roeder. 1997. Synergistic activation of transcription by CBP and p53. Nature 387:819-23. Hall, M. C., and S. W. Matson. 1999. Helicase motifs: the engine that powers DNA unwinding. Mol Microbiol 34:867-77. Hanawalt, P. C. 2001. Controlling the efficiency of excision repair. Mutat Res 485:3-13. Hanawalt, P. C., D. J. Crowley, J. M. Ford, A. K. Ganesan, D. R. Lloyd, T. Nouspikel, C. A. Smith, G. Spivak, and S. Tornaletti. 2000. Regulation of nucleotide excision repair in bacteria and mammalian cells. Cold Spring Harb Syrnp Quant Biol 65:183-91. Haupt, S., M. Berger, Z. Goldberg, and Y. Haupt. 2003. Apoptosis - the p53 network. J Cell Sci 116:4077-85. Haupt, Y., R. Maya, A. Kazaz, and M. Oren. 1997. Mdm2 promotes the rapid degradation of p53. Nature 387 :296-9. Henry, R. W., V. Mittal, B. Ma, R. Kobayashi, and N. Hernandez. 1998. SNAP19 mediates the assembly of a functional core promoter complex (SNAPC) shared by RNA polymerases II and III. Genes Dev 12:2664-72. Henry, R. W., C. L. Sadowski, R. Kobayashi, and N. Hernandez. 1995. A TBP-TAF complex required for transcription of human snRNA genes by RNA polymerase II and 111. Nature 374:653-6. Hernandez, N. 1992. Transcription of vertebrate snRNA genes and related genes, p. 281-313. In S. McKnight and K. Yamamoto (ed.), Transcriptional Regulation. Cold Spring Harbor Laboratory Press, Cold Spring Harbor. Hernandez, N., and R. Lucito. 1988. Elements required for transcription initiation of the human U2 snRNA gene coincide with elements required for snRNA 3' end formation. Embo J 7 :3125-34. 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. Hoffman, R. W., T. Gazitt, M. F. Foecking, R. A. Ortmann, M. Misfeldt, R. Jorgenson, S. L. Young, and E. L. Greidinger. 2004. U1 RNA induces innate immunity signaling. Arthritis Rheum 50:2891-6. 37 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. Hollstein, M., D. Sidransky, B. Vogelstein, and C. C. Harris. 1991. p53 mutations in human cancers. Science 253:49-53. Hu, P., K. Samudre, 8. Wu, Y. Sun, and N. Hernandez. 2004. CK2 phosphorylation of del executes cell cycle-specific RNA polymerase III transcription repression. Mol Cell 16:81-92. Hu, P., S. Wu, and N. Hernandez. 2003. A minimal RNA polymerase III transcription system from human cells reveals positive and negative regulatory roles for CK2. Mol Cell 12:699-709. Hupp, T. R., D. W. Meek, C. A. Midgley, and D. P. Lane. 1992. Regulation of the specific DNA binding function of p53. Cell 71 :875-86. Ito, A., C. H. Lai, X. Zhao, S. Saito, M. H. Hamilton, E. Appella, and T. P. Yao. 2001. p300/CBP-mediated p53 acetylation is commonly induced by p53- activating agents and inhibited by MDM2. Embo J 20:1331-40. Johnson, R. A., T. A. Ince, and K. W. Scotto. 2001. Transcriptional repression by p53 through direct binding to a novel DNA element. J Biol Chem 276:27716- 20. Juan, L. J., W. J. Shia, M. H. Chen, W. M. Yang, E. Seto, Y. S. Lin, and C. W. Wu. 2000. Histone deacetylases specifically down-regulate p53-dependent gene activation. J Biol Chem 275:20436-43. Kaeser, M. D., and R. D. Iggo. 2002. Chromatin immunoprecipitation analysis fails to support the latency model for regulation of p53 DNA binding activity in vivo. Proc Natl Acad Sci U S A 99:95-100. Kapoor, M., and G. Lozano. 1998. Functional activation of p53 via phosphorylation following DNA damage by UV but not gamma radiation. Proc Natl Acad Sci U S A 95:2834-7. Klein, C., E. Planker, T. Diercks, H. Kessler, K. P. Kunkele, K. Lang, S. Hansen, and M. Schwaiger. 2001. NMR spectroscopy reveals the solution dimerization interface of p53 core domains bound to their consensus DNA. J Biol Chem 276:49020-7. Ko, L. J., and C. Prives. 1996. p53: puzzle and paradigm. Genes Dev 10:1054- 72. Krawczak, M., J. Reiss, and D. N. Cooper. 1992. The mutational spectrum of single base-pair substitutions in mRNA splice junctions of human genes: causes and consequences. Hum Genet 90:41-54. 38 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. Kubhutat, M. H., S. N. Jones, and K. H. Vousden. 1997. Regulation of p53 stability by Mdm2. Nature 387:299-303. Kubhutat, M. H., and K. H. Vousden. 1998. Keeping an old fi'iend under control: regulation of p53 stability. Mol Med Today 4:250-6. Kubicka, S., F. Kuhnel, L. Zender, K. L. Rudolph, J. Plumpe, M. Manns, and C. Trautwein. 1999. p53 represses CAAT enhancer-binding protein (C/EBP)- dependent transcription of the albumin gene. A molecular mechanism involved in viral liver infection with implications for hepatocarcinogenesis. J Biol Chem 274:32137-44. Kuhlman, T. C., H. Cho, D. Reinberg, and N. Hernandez. 1999. The general transcription factors IIA, IIB, IIF, and IIE are required for RNA polymerase II transcription from the hmnan U1 small nuclear RNA promoter. Mol Cell Biol 19:2130-41. Kunkel, G. R., and T. Pederson. 1989. Transcription of a human U6 small nuclear RNA gene in vivo withstands deletion of intragenic sequences but not of an upstream TAT ATA box. Nucleic Acids Res 17:7371-9. Kwek, K. Y., S. Murphy, A. Furger, B. Thomas, W. O'Gorman, H. Kimura, N. J. Proudfoot, and A. Akoulitchev. 2002. U1 snRNA associates with TFIIH and regulates transcriptional initiation. Nat Struct Biol 9:800-5. Kwek, S. S., J. Derry, A. L. Tyner, Z. Shen, and A. V. Gudkov. 2001. Functional analysis and intracellular localization of p53 modified by SUMO-1. Oncogene 20:2587-99. Lai, L, K. V. Ferry, M. A. Diamond, K. E. Wee, Y. B. Kim, J. Ma, T. Yang, P. A. Benfield, R. A. Copeland, and K. R. Auger. 2001. Human mdm2 mediates multiple mono-ubiquitination of p53 by a mechanism requiring enzyme isomerization. J Biol Chem 276:31357-67. Lane, D. P., and S. Benchimol. 1990. p53: oncogene or anti-oncogene? Genes Dev 4: 1-8. Leadon, S. A., and P. K. Cooper. 1993. Preferential repair of ionizing radiation- induced damage in the transcribed strand of an active human gene is defective in Cockayne syndrome. Proc Natl Acad Sci U S A 90:10499-503. Leng, R. P., Y. Lin, W. Ma, H. Wu, B. Lemmers, S. Chung, J. M. Parant, G. Lozano, R. Hakem, and S. Benchimol. 2003. Pirh2, a p53-induced ubiquitin- protein ligase, promotes p53 degradation. Cell 112:779-91. 39 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. Li, M., D. Chen, A. Shiloh, J. Luo, A. Y. Nikolaev, J. Qin, and W. Gu. 2002. Deubiquitination of p53 by HAUSP is an important pathway for p53 stabilization. Nature 416:648-53. Li, Z., A. Yu, and A. M. Weiner. 1998. Adenovirus type 12-induced fragility of the human RNU2 locus requires p53 function. J Virol 72:4183-91. Liao, X. C., J. Tang, and M. Rosbash. 1993. An enhancer screen identifies a gene that encodes the yeast U1 snRNP A protein: implications for snRNP protein function in pre-mRNA splicing. Genes Dev 7:419-28. Licht, C. L., T. Stevnsner, and V. A. Bohr. 2003. Cockayne syndrome group B cellular and biochemical functions. Am J Hum Genet 73:1217-39. Linzer, D. 1., and A. J. Levine. 1979. Characterization of a 54K dalton cellular SV40 tumor antigen present in SV40-transformed cells and uninfected embryonal carcinoma cells. Cell 17:43-52. Liu, Y, and M. Kulesz-Martin. 2001. p53 protein at the hub of cellular DNA damage response pathways through sequence-specific and non-sequence-specific DNA binding. Carcinogenesis 22: 851 6.0 Lobo, S. M., and N. Hernandez. 1989. A 7 bp mutation converts a human RNA polymerase II snRNA promoter into an RNA polymerase III promoter. Cell 58:55-67. Lu, Y., H. Lian, P. Sharma, N. Schreiber-Agus, R. G. Russell, L. Chin, G. T. van der Horst, and D. B. Bregman. 2001. Disruption of the Cockayne syndrome B gene impairs spontaneous tumorigenesis in cancer-predisposed Ink4a/ARF knockout mice. Mol Cell Biol 21:1810-8. MacArthur, H. L., M. L. Agarwal, and S. Bacchetti. 1997. Induction of fragility at the human RNU2 locus by cytosine arabinoside is dependent upon a transcriptionally competent U2 small nuclear RNA gene and the expression of p53. Somat Cell Mol Genet 23:379-89. Maier, B., W. Gluba, B. Bernier, T. Turner, K. Mohammad, T. Guise, A. Sutherland, M. Thorner, and H. Scrable. 2004. Modulation of mammalian life span by the short isoform of p53. Genes Dev 18:306-19. Malkin, D. 1994. Germline p53 mutations and heritable cancer. Annu Rev Genet 28:443-65. Mayne, L. V., A. R. Lehmann, and R. Waters. 1982. Excision repair in Cockayne syndrome. Mutat Res 106:179-89. 40 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. McKinney, K., M. Mattia, V. Gottifredi, and C. Prives. 2004. p53 linear diffirsion along DNA requires its C terminus. Mol Cell 16:413-24. McLure, K. G., and P. W. Lee. 1998. How p53 binds DNA as a tetrarner. Embo J 17:3342-50. McLure, K. G., and P. W. Lee. 1999. p53 DNA binding can be modulated by factors that alter the conformational equilibrium. Embo J 18:763-70. Mihara, M., S. Erster, A. Zaika, O. Petrenko, T. Chittenden, P. Pancoska, and U. M. Moll. 2003. p53 has a direct apoptogenic role at the mitochondria. Mol Cell 11:577-90. Miller, J., L. Fairall, and D. Rhodes. 1989. A novel method for the purification of the Xenopus transcription factor IIIA. Nucleic Acids Res 17:9185-92. Mital, R., R. Kobayashi, and N. Hernandez. 1996. RNA polymerase III transcription from the human U6 and adenovirus type 2 VA] promoters has different requirements for human BRF, a subunit of human TFIIIB. Mol Cell Biol 16:7031-42. Mittal, V., M. A. Cleary, W. Herr, and N. Hernandez. 1996. The Oct-1 POU- specific domain can stimulate small nuclear RNA gene transcription by stabilizing the basal transcription complex SNAPC. Mol Cell Biol 16:1955-65. Moll, U. M., and N. Slade. 2004. p63 and p73: roles in development and tumor formation. Mol Cancer Res 2:371-86. Momand, J., G. P. Zambetti, D. C. Olson, D. George, and A. J. Levine. 1992. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell 69: 1237-45. Morra, D. S., S. H. Lawler, B. P. Eliceiri, and G. L. Ellceiri. 1986. Inhibition of small nuclear RNA synthesis by ultraviolet radiation. J Biol Chem 261:3142-6. Muftuoglu, M., R. Selzer, J. Tuo, R. M. Brosh, Jr., and V. A. Bohr. 2002. Phenotypic consequences of mutations in the conserved motifs of the putative helicase domain of the human Cockayne syndrome group B gene. Gene 283:27- 40. Murphy, S., J. B. Yoon, T. Gerster, and R. G. Roeder. 1992. Oct-l and Oct-2 potentiate functional interactions of a transcription factor with the proximal sequence element of small nuclear RNA genes. Mol Cell Biol 12:3247-61. Nagata, S., and P. Golstein. 1995. The F as death factor. Science 267 :1449-56. 41 107. 108. 109. 110. 111. 112.' 113. 114. 115. 116. 117. 118. Nance, M. A., and S. A. Berry. 1992. Cockayne syndrome: review of 140 cases. Am J Med Genet 42:68-84. Nguyen, V. T., T. Kiss, A. A. Michels, and O. Bensaude. 2001. 7SK small nuclear RNA binds to and inhibits the activity of CDK9/cyclin T complexes. Nature 414:322-5. Nicholson, D. W., and N. A. Thornberry. 2003. Apoptosis. Life and death decisions. Science 299:214-5. O'Reilly, D. R. 1986. p53 and transformation by SV40. Biol Cell 57:187-96. Oda, K., H. Arakawa, T. Tanaka, K. Matsuda, C. Tanikawa, T. Mori, H. Nishimori, K. Tamai, T. Tokino, Y. N akamura, and Y. Taya. 2000. p53AIP1, a potential mediator of p53-dependent apoptosis, and its regulation by Ser-46- phosphorylated p53. Cell 102:849-62. Pavletich, N. P., K. A. Chambers, and C. 0. Paho. 1993. The DNA-binding domain of p53 contains the four conserved regions and the major mutation hot spots. Genes Dev 7:2556-64. Pieler, T., J. Hamm, 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. Pines, J. 1995. Cyclins and cyclin-dependent kinases: a biochemical view. Biochem J 308 ( Pt 3):697-711. Qian, H., T. Wang, L. Naumovski, C. D. Lopez, and R. K. Brachmann. 2002. Groups of p53 target genes involved in specific p53 downstream effects cluster into different classes of DNA binding sites. Oncogene 21 :7901-1 1. Ridanpaa, M., H. van Eenennaam, K. Pelin, R. Chadwick, C. Johnson, B. Yuan, W. vanVenrooij, G. Pruijn, R. Salmela, S. Rockas, O. Makitie, I. Kaltila, and A. de la Chapelle. 2001. Mutations in the RNA component of RNase MRP cause a pleiotropic human disease, cartilage-hair hypoplasia. Cell 104:195-203. Rodriguez, M. S., J. M. Desterro, S. Lain, C. A. Midgley, D. P. Lane, and R. T. Hay. 1999. SUMO-1 modification activates the transcriptional response of p53. Embo J 18:6455-61. Rubbi, C. P., and J. Milner. 2003. p53 is a chromatin accessibility factor for nucleotide excision repair of DNA damage. Embo J 22:975-86. 42 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. 129. 130. 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. Sakaguchi, K., J. E. Herrera, S. Saito, T. Miki, M. Bustin, A. Vassilev, C. W. Anderson, and E. Appella. 1998. DNA damage activates p53 through a phosphorylation-acetylation cascade. Genes Dev 12:2831-41. Sakamuro, D., P. Sabbatini, E. White, and G. C. Prendergast. 1997. The polyproline region of p53 is required to activate apoptosis but not growth arrest. Oncogene 15:887-98. Samuels-Lev, Y., D. J. O'Connor, D. Bergamaschi, G. Trigiante, J. K. Hsieh, S. Zhong, I. Campargue, L. Naumovski, T. Crook, and X. Lu. 2001. ASPP proteins specifically stimulate the apoptotic function of p53. Mol Cell 8:781-94. Schmidt, D., and S. Muller. 2002. Members of the PIAS family act as SUMO ligases for c-Jun and p53 and repress p53 activity. Proc Natl Acad Sci U S A 99:2872-7. Schneider, E., M. Montenarh, and P. Wagner. 1998. Regulation of CAK kinase activity by p53. Oncogene 17:2733-41. Schramayr, S., D. Caporossi, I. Mak, T. Jelinek, and S. Bacchetti. 1990. Chromosomal damage induced by human adenovirus type 12 requires expression of the E13 55-kilodalton viral protein. J Virol 64:2090-5. Schramm, L., P. S. Pendergrast, Y. Sun, and N. Hernandez. 2000. Different human TFIIIB activities direct RNA polymerase III transcription from TATA- containing and TATA-less promoters. Genes Dev 14:2650-63. Scrable, H., T. Sasaki, and B. Maier. 2005. DeltaNp53 or p44: priming the p53 pump. Int J Biochem Cell Biol 37:913-9. Selby, C. P., and A. Sancar. 1997. Cockayne syndrome group B protein enhances elongation by RNA polymerase II. Proc Natl Acad Sci U S A 94:11205- 9. Seoane, J., H. V. Le, and J. Massague. 2002. Myc suppression of the p21(Cipl) Cdk inhibitor influences the outcome of the p53 response to DNA damage. Nature 419:729-34. Seto, E., A. Usheva, G. P. Zambetti, J. Momand, N. Horikoshi, R. Weinmann, A. J. Levine, and T. Shenk. 1992. Wild-type p53 binds to the TATA-binding protein and represses transcription. Proc Natl Acad Sci U S A 89: 12028-32. 43 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. Shikama, N., C. W. Lee, S. France, L. Delavaine, J. Lyon, M. Krstic- Demonacos, and N. B. La Thangue. 1999. A novel cofactor for p300 that regulates the p53 response. Mol Cell 4:365-76. Shinobu, N., T. Maeda, T. Aso, T. Ito, T. Kondo, K. Koike, and M. Hatakeyama. 1999. Physical interaction and functional antagonism between the RNA polymerase II elongation factor ELL and p53. J Biol Chem 274:17003-10. Spivak, G. 2004. The many faces of Cockayne syndrome. Proc Natl Acad Sci U S A 101:15273-4. Stein, T., D. Crighton, J. M. Boyle, J. M. Varley, and R. J. White. 2002. RNA polymerase III transcription can be derepressed by oncogenes or mutations that compromise p53 function in tumours and Li-Fraumeni syndrome. Oncogene 21:2961-70. Stein, T., D. Crighton, L. J. Warnock, J. Milner, and R. J. White. 2002. Several regions of p53 are involved in repression of RNA polymerase III transcription. Oncogene 21 :5540-7. Sturm, R. A., and W. Herr. 1988. The POU domain is a bipartite DNA-binding structure. Nature 336:601-4. Sunesen, M., R. R. Selzer, R. M. Brosh, Jr., A. S. Balajee, T. Stevnsner, and V. A. Bohr. 2000. Molecular characterization of an acidic region deletion mutant of Cockayne syndrome group B protein. Nucleic Acids Res 28:3151-9. Susani, L., A. Pangrazio, C. Sobacchi, A. Taranta, G. Mortier, R. Savarirayan, A. Villa, P. Orchard, P. Vezzoni, A. Albertini, A. Frattini, and F. Pagani. 2004. TCIRGl-dependent recessive osteopetrosis: mutation analysis, functional identification of the splicing defects, and in vitro rescue by U1 snRNA. Hum Mutat 24:225-35. Tantin, D., A. Kansal, and M. Carey. 1997. Recruitment of the putative transcription-repair coupling factor CSB/ERCC6 to RNA polymerase II elongation complexes. Mol Cell Biol 17:6803-14. Taylor, W. R., and G. R. Stark. 2001. Regulation of the G2/M transition by p53. Oncogene 20:1803-15. Thornborrow, E. C., S. Patel, A. E. Mastropietro, E. M. Schwartzfarb, and J. J. Manfredi. 2002. A conserved intronic response element mediates direct p53- dependent transcriptional activation of both the human and murine bax genes. Oncogene 21 :990-9. 44 142. 143. 144. 145. 146. 147. 148. 149. 150. 151. Troelstra, C., R. M. Landsvater, J. Wiegant, M. van der Ploeg, G. Viel, C. H. Buys, and J. H. Hoeijmakers. 1992. Localization of the nucleotide excision repair gene ERCC6 to human chromosome 10q11-q21. Genomics 12:745-9. Tuo, J., M. Muftuoglu, C. Chen, P. Jaruga, R. R. Selzer, R. M. Brosh, Jr., H. Rodriguez, M. Dizdaroglu, and V. A. Bohr. 2001. The Cockayne Syndrome group B gene product is involved in general genome base excision repair of 8- hydroxyguanine in DNA. J Biol Chem 276:45772-9. van Gool, A. J., E. Citterio, S. Rademakers, R. van Os, W. Vermeulen, A. Constantinou, J. M. Egly, D. Bootsma, and J. H. Hoeijmakers. 1997. The Cockayne syndrome B protein, involved in transcription-coupled DNA repair, resides in an RNA polymerase II-containing complex. Embo J 16:5955-65. Venema, J., L. H. Mullenders, A. T. Natarajan, A. A. van Zeeland, and L. V. Mayne. 1990. The genetic defect in Cockayne syndrome is associated with a defect in repair of UV-induced DNA damage in transcriptionally active DNA. Proc Natl Acad Sci U S A 87 :4707-1 1. Venot, C., M. Maratrat, C. Dureuil, E. Conseiller, L. Bracco, and L. Debussche. 1998. The requirement for the p53 proline-rich functional domain for mediation of apoptosis is correlated with specific PIG3 gene transactivation and with transcriptional repression. Embo J 17:4668-79. Wang, X. W., H. Yeh, L. Schaeffer, R. Roy, V. Moncollin, J. M. Egly, Z. Wang, E. C. Freidberg, M. K. Evans, B. G. Taffe, and et al. 1995. p53 modulation of T FIIH-associated nucleotide excision repair activity. Nat Genet 10:188-95. Waterman, M. J., E. S. Stavridi, J. L. Waterman, and T. D. Halazonetis. 1998. ATM-dependent activation of p53 involves dephosphorylation and association with 14-3-3 proteins. Nat Genet 19:175-8. Wong, K. B., B. S. DeDecker, S. M. Freund, M. R. Proctor, M. Bycroft, and A. R. Fersht. 1999. Hot-spot mutants of p53 core domain evince characteristic local structural changes. Proc Natl Acad Sci U S A 96:8438-42. Wu, G. S., T. F. Burns, E. R. McDonald, 3rd, W. Jiang, R. Meng, I. D. Krantz, G. Kao, D. D. Gan, J. Y. Zhou, R. Muschel, S. R. Hamilton, N. B. Spinner, S. Markowitz, G. Wu, and W. S. el-Deiry. 1997. KILLER/DRS is a DNA damage-inducible p53-regulated death receptor gene. Nat Genet 17:141-3. Xirodimas, D. P., M. K. Saville, J. C. Bourdon, R. T. Hay, and D. P. Lane. 2004. Mdm2-mediated NEDD8 conjugation of p53 inhibits its transcriptional activity. Cell 118:83-97. 45 152. 153. 1 54. 155. 156. 157. 158. 159. Yang, A., and F. McKeon. 2000. P63 and P73: P53 mimics, menaces and more. Nat Rev Mol Cell Biol 1:199-207. Yang, Z., Q. Zhu, K. Luo, and Q. Zhou. 2001. The 7SK small nuclear RNA inhibits the CDK9/cyclin T1 kinase to control transcription. Nature 414:317-22. Yoon, J. B., S. Murphy, L. Bai, Z. Wang, and R. G. Roeder. 1995. Proximal sequence element-binding transcription factor (PTF) is a multisubunit complex required for transcription of both RNA polymerase II- and RNA polymerase III- dependent small nuclear RNA genes. Mol Cell Biol 15:2019-27. Yu, A., H. Y. Fan, D. Liao, A. D. Bailey, and A. M. Weiner. 2000. Activation of p53 or loss of the Cockayne syndrome group B repair protein causes metaphase fragility of human U1, U2, and SS genes. Mol Cell 5:801-10. Yu, J., and L. Zhang. 2003. No PUMA, no death: implications for p53- dependent apoptosis. Cancer Cell 4:248-9. Zhai, W., and L. Comai. 2000. Repression of RNA polymerase I transcription by the tumor suppressor p53. Mol Cell Biol 20:5930-8. Zieve, G., and S. Penman. 1976. Small RNA species of the HeLa cell: metabolism and subcellular localization. Cell 8: 19-3 1. Zotchev, S. B., M. Protopopova, and G. Selivanova. 2000. p53 C-terrninal interaction with DNA ends and gaps has opposing effect on specific DNA binding by the core. Nucleic Acids Res 28:4005-12. 46 CHAPTER TWO THE P53 TUMOR SUPPRESSOR PROTEIN REPRESSES HUMAN SNRNA GENE TRANSCRIPTION BY RNA POLYMERASES II AND 111 INDEPENDENTLY OF SEQUENCE-SPECIFIC DNA BINDINGi Abstract Human U1 and U6 snRNA genes are transcribed by RNA polymerases H and 111, respectively. While the p53 tumor suppressor protein is a general repressor of RNA polymerase III transcription, whether p53 regulates snRNA gene transcription by RNA polymerase II is uncertain. The data presented herein indicate that p53 is an effective repressor of snRNA gene transcription by both polymerases. Both U1 and U6 transcription in vitro is repressed by recombinant p53, and endogenous p53 occupancy at these promoters is stimulated by UV light. In response to UV light, U1 and U6 transcription is strongly repressed. Human U1 genes, but not U6 genes, contain a high- affinity p53 response element located within the core promoter region. Nonetheless, this element is not required for p53 repression and mutant p53 molecules that do not bind DNA can maintain repression, suggesting a reliance on protein interactions for p53 promoter recruitment. Recruitment may be mediated by the general transcription factors TAT A-box binding protein and snRNA-activating protein complex, which interact well with p53 and function for both RNA polymerase II and III transcription. This work was published as the following manuscript: Anastasia A. Gridasova and R. William Henry (2005) The p53 tumor suppressor protein represses human snRNA gene transcription by RNA polymerases II and III independently of sequence-specific DNA binding. Molecular and Cellular Biology, Apr. 2005, pp. 3247-3260. 47 Introduction The p53 tumor suppressor protein plays a critical role in preventing unwarranted cellular proliferation by activating transcription of key target genes that influence cell grth and apoptosis (reviewed in references 28, 31, 35, and 64). Though p53 can enable both pathways, the switch controlling which cellular outcome is enacted is uncertain (reviewed in references 65 and 66), but both the p53 level and the nature of the DNA damage can influence apoptotic response (8). Altogether, p53 activity serves to prevent passage of mutations to daughter cells afier DNA damage. Recent evidence suggests that p53 regulates transcription of genes that are not obviously involved in controlling cell cycle arrest or apoptosis. Indeed, p53 can repress RNA polymerase I (3, 72) and III (S, 9) transcription of genes encoding a variety of nontranslated RNAs that play critical roles at numerous points during global gene expression. RNA polymerase III activity is elevated in p53-/- knockout fibroblasts (5) and in a variety of cancer-derived cell lines that lack p53 function (57). However, the mechanism for p53 regulation of RNA polymerase III transcription is controversial. A kinetic analysis of RNA polymerase III repression using p53 expressed from a stably integrated inducible p53 gene suggested that RNA polymerase HI repression is mediated indirectly through p53-dependent degradation of TFIIIB (11). In contrast, recombinant p53 can repress in vitro transcription from a variety of RNA polymerase III-specific promoters and can interact with components of the general transcription machinery required for RNA polymerase III transcription (5, 9, 10, 58), indicating that p53 might directly repress transcription by RNA polymerase III. 48 Within the group of genes transcribed by RNA polymerase IH, the human snRNA gene family is intriguing because these genes contain similar sets of promoter elements, and yet only some genes are transcribed by RNA polymerase III while others are transcribed by RNA polymerase 11 (see references 19, 2,3, 24, and 42 for review). Regardless of polymerase specificity, human snRNA genes contain a distal sequence element in the upstream promoter region that serves as the recognition element for activator proteins, including Oct-1, STAF, and Spl (33, 54). These factors activate transcription from the core promoters that commonly contain a proximal sequence element (PSE). The PSE is directly recognized by a general transcription factor called the snRNA activating protein complex (SNAPC) (52), which is also known as the PSE transcription factor (49). SNAPC is involved in human snRNA gene transcription by both RNA polymerases II and 111 (20—22, 51, 69). RNA polymerase III-transcribed snRNA genes also contain a TATA box that serves to recruit the TATA-box binding protein (TBP) as part of an snRNA-specific TFIIIB complex (45, 55, 60). The conservation of important promoter elements among human snRNA genes suggests that transcription of these genes by RNA polymerases II and 111 may be coordinately regulated. However, it is not known whether p53 can regulate human snRNA gene transcription by RNA polymerase II. A role for p53 in this process is suggested from two sources. Firstly, in response to UV light treatment, human U1 and U2 snRNA genes exhibit a delayed and prolonged reduction in transcription by RNA polymerase II (14, 47, 48). In part, this reduction may be attributable to increased hyperphosphorylation of the carboxy-terminal domain of the RNA polymerase 11 largest subunit in response to UV light (27). However, in normal human diploid fibroblasts, the 49 balance of hyper- and hypophosphorylated RNA polymerase II is restored by 6 h after UV light treatment (46), suggesting additional cellular mechanisms that enable snRNA gene repression after UV light exposure. Potentially, p53 activation by DNA damage might play a direct role in the prolonged repression of these genes. Secondly, infection of human cells by adenovirus serotype 12 causes metaphase fragility at four chromosomal sites, including the U1 snRNA (RNUl) and U2 snRNA (RNU2) loci (1, 36), in a process that requires p53 (38, 39). It was postulated that fragile site formation occurs during viral infection, because RNA polymerase II stalls at these genes and interferes with chromosome condensation during metaphase (37). Interestingly, p53 that harbors mutations in the DNA binding domain supports fragile site formation (39), and overexpression of the C-terrninal domain of p53 alone, which lacks the DNA binding domain, induces fragility during transient transfection (71). Together, these data indicate that p53 is important for generation of fragile sites at the U1 and U2 snRNA gene loci and may play a role in regulation of these genes in a fashion that does not require sequence-specific binding of p53 to DNA. In this study, the role of p53 in governing human snRNA gene transcription by RNA polymerase II and III was examined. We show that recombinant p53 represses both U1 and U6 transcription by RNA polymerase II and 111, respectively. Repression is supported by the C-terminal region of p53 alone, indicating that sequence-specific DNA binding by p53 is not critical for repression. Both the full-length and C terminus of p53 alone can associate with the U1 and U6 promoters during repression, and promoter recruitment may be assisted through interactions with the general transcription factors SNAPC and TBP, which are commonly required for transcription of both U1 and U6 50 snRNA genes. In vivo, p53 can bind to both U1 and U6 snRNA genes in untreated human MCF-7 cells, and promoter occupancy is stimulated after UV light treatment. These results firrther indicate that p53 contributes to snRNA gene regulation in response to DNA damage. Materials and Methods Cell culture and UV irradiation Human MCF-7 and HeLa cells were maintained in Dulbecco’s modified Eagle’s medium supplemented with penicillinstreptomycin and 10% (MCF-7) or 5% (HeLa) fetal bovine serum. Cells grown to 70 to 80% confluence were washed with phosphate- buffered saline and irradiated with 50 J of UV light (254-nm peak)/m2 by using a UV Stratalinker (Stratagene). After irradiation, growth medium was added and cells were incubated at 37°C under 5% C02 for the indicated times. Additionally, HeLa cells were grown to 50% confluence in ISO-mm plates and were then transiently transfected with 2.5 ug of the pRc/RSV or pRc/RSV-p53-Flag.wt plasmids by using Lipofectin reagent (Invitrogen) for 6 h. Subsequently, the medium was replaced and cells were incubated for 48 h for further analysis in nuclear run-on assays. Nuclear run-on assays Nuclear run-on assays were performed as described elsewhere (6) in the presence of [or-32P]UTP using approximately 107 nuclei that were isolated from MCF-7 or HeLa cells before or 8 h after exposure to UV light. Additional assays were performed using HeLa cells transiently transfected with pRc/RSV or pRc/RSV-p53-Flag.wt as described. 51 Labeled RNA was recovered and hybridized to a nitrocellulose membrane containing approximately 7 ug of U1, U6, and 5S rRNA target gene DNAs or 10 ug of pUC119 plasmid, as a negative control. Target gene DNAs corresponding to the coding regions of the indicated genes were generated by PCR and were immobilized on a nylon membrane at levels calculated to be in excess relative to the corresponding snRNA population in the nuclei. Hybridizations were performed for 16 h at 42°C in hybridization buffer containing 50% forrnamide. Membranes were then washed extensively in 2X SSC (1X SSC is 0.15 M NaCl plus 0.015 M sodium citrate) and 0.1% sodium dodecyl sulfate (SDS) at 50°C. Hybridized RNA transcripts were visualized by autoradiography for 7 days. Similar results were observed when signals were normalized to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) expression by varying the exposure time (data not shown). RNA isolation and RT -PCR RNA was isolated using the TRIzol reagent as recommended by the manufacturer (Gibco-BRL). RNA preparations were quantified by UV spectrometry and examined for integrity by agarose formaldehyde morpholinepropanesulfonic acid gel electrophoresis. Reverse transcription-PCR (RT-PCR) was performed by a two-step procedure using U1-, U6-, and GAPDH-specific primers. The primers used for amplification of each gene were the following: U1 forward, 5’-ATACTTACCTGGCAGGGGAG-3’; U1 reverse, 5’- CAGGGGGAAAGCGCGAACGCA-3’; U6 forward, 5’-GGAATCTAGAACATATACT AAAATTGGAAC-3’; U6 reverse, 5’-GGAACTCGAGTTTGCGTGTCATCCTTGCGC- 3’; GAPDH forward, 5’-AGGTCATCCCTGAGCTGAAC-3’; and GAPDH reverse, 5’- GCAATGCCAGCCCCAGCGTC-3’. 52 Expression and purification of recombinant proteins Glutathione S-transferase (GST), GST-tagged full-length human p53, and a GST- tagged C terminus of human p53 (amino acids 301 to 393) [p53 (301-393)] were expressed in Escherichia coli BL21(DE3) codon+ cells (Stratagene) and were affinity purified by binding to glutathione agarose beads (Sigma). GST and GST-p53 were then eluted from beads in HEMGT-lSO buffer containing 50 mM glutathione for 4 h at 4°C or, alternatively, untagged p53 was obtained by digestion with thrombin. Proteins were further purified by chromatography using a Mono-Q (HRS/5) column (Pharrnacia) and were concentrated by centrifugation using a Centricon YM-30 spin column (Millipore) in HEMGT-80 buffer (20 mM HEPES [pH 7.9], 0.1 mM EDTA, 10 mM MgC12, 10% glycerol [vol/vol], 0.1% Tween 20, 80 mM KCl) containing protease inhibitors and 1 mM dithiothreitol. In vitro transcription assays In vitro transcription assays were performed as described previously (21, 43) using 18, 2, 2, and 10 uL of HeLa cell nuclear extract for the U1 snRNA, U6 snRNA, 5S rRNA, and adenovirus major late promoter (AdML) transcription reactions, respectively. The pU1-4.0 (1 pg), pU6/Hae/RA.2 (250 ng), pHSSsa (250 ng), and M13-AdML (250 11g) templates were used for the U1 snRNA, U6 snRNA, SS rRNA, and AdML transcription reactions, respectively. Purified p53 or GST-tagged p53 proteins were added in the amounts indicated in the figure legends. Transcription reactions were performed for 1 h at 30°C. Transcripts were separated by denaturing 6% polyacrylamide gel electrophoresis (PAGE) and visualized by autoradiography. 53 Chromatin immunoprecipitation assay Chromatin immunoprecipitation assays were performed as described previously (25). Human MCF-7 cells were grown to 60 to 80% confluence and were then cross- linked with 1% formaldehyde for 30 min at room temperature. After cell lysis and sonication, immunoprecipitation reactions were performed overnight at 4°C using chromatin from approximately 107 cells per reaction mixture and 1 ug of each antibody. The anti-p53 antibodies used were the following: anti-p53 (21-25) (Ab-6; Oncogene), anti-p53 (371-380) (Ab-1; Oncogene), anti-p53 (213-217) (Ab240; Pharmingen), anti- acetyl-p53 (373-382) (Upstate), and anti-acetyl-p53 (320) (Upstate). Recovered chromatin was suspended in 50 11L of H20, and PCR analysis was performed using 5 11L of immunoprecipitated chromatin or input chromatin. The primers used for amplification of each gene were the following: U1 forward, 5’-CACGAAGGAGTTCCCGTG-3’; U1 reverse, 5’-CCCTGCCAGGTAAGTATG-3’; U2 forward, 5’-AGGGCGTCMTAGCGC TGTGG-3’; U2 reverse, 5’-TGCGCTCGCCTTCGCGCCCGCCG-3’; U6 forward, 5’- GTACAAAATACGTGACGTAGAAAG-3’; U6 reverse, 5’-GGTGTTTCGTCCTTTCC AC-3’; GAPDH forward, 5’AGGTCATCCCTGAGCTGAAC-3’; GAPDH reverse, 5’- GCAATGCCAGCCCCAGCGTC-3’; U1 upstream forward, S’-GAACTTACTGGGATC TGG-3’; U1 upstream reverse, 5’-GAGACAACTGAGCCACTTG-3’; p21 upstream forward, 5’-CCGCTCGAGCCCTGTCGCAAGGATCC-3’; p21 upstream reverse, 5’- GGGAGGAAGGGGATGGTAG-3’. PCR products were separated by 2% agarose electrophoresis in Tris-borate-EDTA buffer and were stained with ethidium bromide. 54 Immunoprecipitations from in vitro transcription reactions In vitro transcription assay mixtures containing U1 or U6 promoter plasmids and equal molar amounts of pUCll9 were performed as described previously (25) in the absence or presence of full-length GST-p53 (wild-type or R175H), GST-p53 (301-393), or GST. Five microliters of each transcription reaction mixture was diluted to 500 1.1L and was cross-linked in 1% formaldehyde for 10 min at room temperature, quenched with 125 mM glycine for 10 min at room temperature, and immunoprecipitated with immunoglobulin G (IgG), anti-SNAP43 (CS48), or anti-p53 (Ab-1; Oncogene) antibodies. Recovered plasmid DNA was analyzed by PCR using primers specific to the U1 and U6 promoter regions or to pUC119 as a negative control. DNase I footprinting Footprinting assays were generally performed as described elsewhere (4). Linear DNA encompassing the human U1 promoter from -151 to +13 was generated by PCR using primers that were end labeled with [y-32P]ATP by using T4 polynucleotide kinase (New England BioLabs). U1 promoter probes were incubated with increasing amounts of recombinant GST-p53 for 40 min at room temperature and were then digested with 0.04 U of DNase I (Roche) for 2 min at room temperature. The resultant fragments were purified and separated by 8% denaturing PAGE. Footprints were visualized by autoradiography and were mapped relative to sequencing ladders generated from the same labeled primers used to generate the U1 promoter probes. 55 EMSA Electrophoretic mobility shift assays (EMSA) were performed as described elsewhere (7). The amounts of p53 used are indicated in the figure legends. Reaction mixtures were incubated at room temperature for 20 min prior to addition of radiolabeled probes. Unless otherwise noted, the U1 probe encompasses -312 to +13 and the U6 probe encompasses -267 to +1. DNA binding reactions were carried out at room temperature for 20 min, and resulting DNA-protein complexes were separated on a 4% polyacrylamide gel in 0.5X Tris-borate-EDTA running buffer at 150 V. Complexes were visualized by autoradiography. Coimmunoprecipitation and GS T pull-down experiments GST pull-down assays were performed as previously described (25). Coimmunoprecipitation assays were performed using 5 mg of total protein contained in MCF-7 nuclear extracts from untreated or UV-treated cells and 2 pg of rabbit anti- SNAP43 antibodies (CS48) (22). Western blot analyses of recovered proteins were performed using anti-SNAP43 (CS48), anti-p53 (Ab-6; Oncogene), and anti-galectin-3 (Mac2) antibodies. The reciprocal immunoprecipitation reactions were performed using 2 pg of anti-p53 antibody (Ab-6) with approximately 1.6 or 5 mg of MCF-7 extract, followed by anti-SNAP43 Western blot analysis. 56 Results p53 represses human snRNA gene transcription by both RNA polymerases II and 111. To determine whether p53 can repress U1 transcription by RNA polymerase II, as has been previously shown for U6 transcription by RNA polymerase III (5, 9), the effect of recombinant p53 on human U1 in vitro transcription was tested. The recombinant full- length wild-type p53 and the GST proteins used for these experiments are shown in Fig. 1A. As shown in Fig. 1B, p53 effectively repressed correctly initiated U1 transcription (labeled U1 5’) by RNA polymerase II, and this repressive effect was specific, because concomitant RNA polymerase II transcription of an mRNA read-through transcript derived from the same plasmid was unaffected in these reactions. As a positive control for p53 activity, human U6 snRNA gene transcription was tested. Indeed, the same amounts of p53 effectively repressed U6 snRNA transcription by RNA polymerase III, while RNA polymerase II transcription from the AdML was unaffected. Therefore, p53 can repress human snRN A gene transcription by both RNA polymerases II and 111. As a first step towards understanding the mechanism for p53 repression of U1 and U6 transcription, a time course for p53 repression was performed (Fig. 1C). As a positive control, p53 repression of SS rRNA gene transcription was also examined. To ensure maximal repression, an excess of p53 was used, because 5S rRNA gene transcription appears less sensitive to p53 repression (9) (data not shown). As was demonstrated previously (5), p53 can repress SS rRNA transcription when added to reactions concomitantly with nuclear extract (lane 2) or nuclear extract plus template DNA (lane 3) prior to initiation of transcription by nucleotide addition. However, p53 did not repress 57 Figure 2-1. p53 represses human snRNA gene transcription by both RNA polymerases II and III in vitro. (A) Recombinant full-length wild-type p53 and GST proteins were separated by SDS—12.5% PAGE and were stained with Coomassie blue. (B) In vitro transcription fi'om U1, U6, and AdML promoter constructs was tested using HeLa nuclear extracts containing 0, 50, 200, and 800 11g of p53 (lanes 1 to 4) or 800 ng of GST (lane 5). Fifty nanograms of p53 represents an approximate 2:1 molar ratio of monomeric p53 to U1 promoter template DNA and an approximate 8:1 molar ratio to the U6 and AdML promoter plasmids. p53 effectively repressed correctly initiated U1 transcription (U l 5 ) and U6 transcription (U6 5 ), but it didn’t affect read-through (RT) transcription from the U1 reporter plasmid or transcription from the AdML promoter. (C) U1, U6, and SS rRNA in vitro transcription reaction mixtures were supplemented with 800 ng of active or heat-inactivated p53 (lanes 2 to 4 and lanes 5 to 7, respectively) at different times, as indicated. Transcription was allowed to proceed for an additional 60 min. Recombinant p53 repressed U6 gene transcription both prior to and after preinitiation complex assembly but did not repress U1 and 5S rRNA gene transcription after the formation of a preinitiation complex. 58 97.4— ‘9 66- w 45— one. “"""' _p53 31 '- quip 14.4- ‘- 1 2 3 Coomassie - -- —u15' fit « fi—ues' .---- -AdM'- 1 2 3 4 5 In vitro transcription 59 0' 15' 30' +Extract +DNA +NTPs — 0' 15' 30' o' 15' 30' Tlme _ _ _ _ + + 4. HI p53 — + + + — — ._ P53 - RT — U1 5' — U6 5' j— 58 5' In vitro transcription 60 5S rRNA transcription when the nuclear extract was preincubated with template DNA prior to p53 addition (lane 4). Presumably, p53 cannot repress SS rRNA gene transcription after the formation of a preinitiation complex. For all time points, repression was specific for functional p53, because repression was disabled by heat inactivation of p53 (lanes 5 to 7). In contrast, p53 could effectively repress U6 transcription by RNA polymerase III even after the nuclear extract had been preincubated with the template DNA. This result suggests that the U6 preinitiation complex is not recalcitrant to p53 repression. Surprisingly, the pattern for p53 repression of U1 transcription by RNA polymerase H was similar to that of SS rRNA rather than the U6 repression pattern. This observation suggests that formation of a preinitiation complex could render U1 snRNA genes refractory to p53 repression. This result also suggests that p53 represses U1 and U6 snRNA gene transcription by different mechanisms. As UV light exposure activates p53, this treatment was used here to determine whether p53 is involved in human snRNA transcriptional regulation in vivo. The majority of in vivo studies presented herein were performed with human MCF-7 breast adenocarcinoma cells, because these cells exhibit a robust increase in p53 levels in response to UV light treatment (Fig. 2A, lanes 4 to 6) compared to human HeLa cervical carcinoma cells, wherein p53 levels are low and remain unchanged after UV light exposure (lanes 1 to 3). To determine the effect of UV light on snRNA gene transcription, nuclear run-on experiments were performed using nuclei harvested from HeLa and MCF-7 cells before and 8 h after UV light exposure. As shown in Fig. 2B, UV light treatment of HeLa cells did not substantially affect U1 transcription by RNA polymerase 11 compared to cells that 61 HeLa MCF7 'O 'O .03. 9 8 + UV 3 + uv *3 I ' e I ' f: 4hr 8hr : 4hr 8hr 1 2 3 anti-p53 Western -UV +UV -UV +UV 0 hr 8 hr 0 hr 8 hr ,i — pUC119 _ __ .. ~— — U1 snRNA “ u .. ~---- — U6 snRNA 1 2 3 4 Nuclear run-on Nuclear run-on HeLa MCF-7 63 Figure 2-2. UV light inhibits snRNA gene transcription and stimulates p53 binding to human snRNA gene promoters. (A) Whole-cell extracts from untreated and UV light-treated HeLa (lanes 1 to 3) and MCF-7 (lanes 4 to 6) cells were analyzed by SDS— 12.5% PAGE and Western blot analysis of endogenous p53 and actin. MCF-7 cells exhibited robust accumulation of endogenous p53 in response to UV light treatment, whereas no change was observed in HeLa cells. (B) UV light represses transcription of endogenous human U1 and U6 snRNA genes in MCF-7 cells, but not in HeLa cells. Nuclear run-on assays measuring polymerase density at U1 snRNA, U6 snRNA, and SS rRNA genes in nuclei from untreated HeLa cells (lane 1) or MCF-7 cells (lane 3) were compared to results with nuclei harvested 8 h after UV light treatment (lanes 2 and 4). After hybridization, membranes were exposed to film for 7 days. Similar trends were also obtained when exposure times were varied to normalize to GAPDH gene transcription, which was unaffected by UV light treatment in these assays (data not shown). (C) Transiently transfected p53 represses U1 snRNA gene transcription in HeLa cells. Nuclear run-on assays were performed on HeLa cells (lane 1) or HeLa cells transiently transfected with either the empty vector pRC/RSV (lane 2) or pRC-RSV expressing wild- type full-length Flag-tagged p53 (pRc/RSV-p53-Flag) (lane 3). Levels of p53 expression were determined by Western blotting (bottom panel). (D) Endogenous p53 associates with human snRNA gene promoters. Chromatin immunoprecipitation experiments were performed using chromatin harvested from MCF-7 cells prior to or 8 h after UV light treatment and using antibodies directed against SNAP43 (lane 3), various epitopes within p53 (lanes 4 to 7), and nonspecific IgG (lane 8) as a negative control. Enrichment of U1 and U6 promoter regions was measured by PCR and was compared to the p21 promoter (-1.4 kb site), as a positive control, and the U1 upstream region and GAPDH exon 2, as negative controls. (E) Endogenous p53 was not detected at human snRNA gene promoters in untreated or UV light-treated HeLa cells. Chromatin immunoprecipitation experiments were performed using HeLa cell chromatin with the indicated antibodies. (F) UV light causes a decrease in steady-state U1 snRNA levels. (Top panel) Total RNA was isolated fiom untreated MCF-7 cells and was titrated (O, 0.1, 0.3, 1, 3, and 10 ng [lanes 2 through 7, respectively]) into RT-PCRs performed using U1 gene-specific primers. The amount of U1 cDNA amplification is proportional to the amount of total RNA used for RT-PCR. (Bottom panel) Steady-state levels of U1, U6, and GAPDH RNA were measured by RT-PCR using 2 ng, 10 ng, and 1 ug of total RNA, respectively, harvested before (lane 1) or after (lanes 2 to 7) UV light treatment. 62 NT E.V. wtp53 Chromatin IP 64 - pUC119 ""'" "" m - U1 m "r - -- U6 ..... «- -- - 58 rRNA 1 2 3 Nuclear run-on HeLa =3: “* “ =1." J—p53 1 2 3 Western MCF-7 r N co :0 + --aa 8 E‘— m m C? a! co m s a .2 $2 ‘3 “9’- ’5 D- o. <0 N — -' o. E < V V 3‘ 3‘ E 0 2 co m 8 8 °\ to 1.0 ,\° .- "2 9 9 ‘F ‘1: 0 ,... o' :5 5 C5 :5 t5 2’ an... «In-- a-n-a-‘r-I-I- - W -.'.'.."... 2...... .. ‘33,“) U1 snRNA .— ‘----*—‘—' -UVWUZ ’-._ ‘--—--—- +UV snRNA ‘o- —- "LJVml ._ _.,.. __ __ +UVus snRNA 1 .: j: 31’, U1 Upstream 3;: .;'L’,‘\’,lGAPDH .— .— - 1 -.-. ---—--—- 43)/I p21 1 2 - 3 4 5 6 7 8 lTl 1 234 RT-PCR 65 6 7 HeLa MCF-7 [_ A lfiAl "ol 8 .2 5' ‘3 J— 3- a .8 a $- 9‘- 5 °\0 2 i?) 3 ,\° - “P 0 9 9 .- o‘ 8 2’ :5 e — —- "’" 'UV -- ]U1 snRNA .. ._, - +UV - -- "UV i - -- U1 upstream an. -- +UV. a -UV 3'": -- i p21 al- - +UVJ 3'" 'le PDH - M - +UV GA 1 2 3 4 5 i 6 Chromatin IP 6 + RNA .35 V l m — A E IV- . m —~ —U‘lSt‘lRNA 1 2 3 4 5 6 7 -UV UV treated F I O 0 1 2 4 8 24 hr hunch-u-» —U18nRNA did not receive UV light exposure. Similarly, U6 snRNA and SS rRNA transcription by RNA polymerase III was unaffected, suggesting that the transcription of these genes is insensitive to UV light. These results for U1 transcription are in contrast with that previously described wherein U1 transcription in nuclear run-on assays was markedly reduced 2 h after UV light treatment of HeLa cells (48). In the present study, the 8-h posttreatrnent time point was selected because RNA polymerase II is ubiquitylated and degraded in response to UV light but normal levels are restored by 6 h after UV light treatment (46). Additionally, a longer recovery period after UV light treatment was desirable to allow sufficient time for DNA damage repair. In contrast with HeLa cells, MCF-7 cells exhibited a marked reduction in U1 transcription 8 h after UV light treatment. Additional studies revealed that repression was already established by 4 h posttreatrnent (data not shown). Interestingly, UV light elicited different effects on RNA polymerase III-transcribed genes, causing reduced U6 transcription while stimulating SS rRNA transcription. In all cases, the signals detected for these transcripts are specific, because no hybridization to pUC119 was detected in any of these experiments. Therefore, U1 and U6 transcription exhibits cell type-specific responses to UV light treatment, with UV light invoking a prolonged repressive effect in MCF-7 cells but not in HeLa cells. In two independent replicates of this experiment, U1 and U6 transcription levels were reduced to 42 and 39%, respectively, of the untreated sample levels, whereas SS RNA transcription rates were increased to 170%. Stimulated SS rRNA gene transcription under these conditions was unexpected but does indicate that the SS rRNA transcriptional response to DNA damage depends upon cellular p53 status. 66 To determine whether p53 contributes to regulation of endogenous snRNA genes, p53 was overexpressed in HeLa cells and the effect on endogenous U1, U6, and SS rRNA gene transcription was again measured by nuclear run-on assays. As shown in Fig. 2C, increased p53 expression was correlated with diminished U1 transcription. In contrast, U6 transcription was unaffected, whereas SS rRNA transcription was stimulated. The reason for the unresponsiveness of U6 transcription to p53 expression is unknown, but p53 may require additional UV light-stimulated modification for activity at this gene. Interestingly, the expression pattern for U1 snRNA and SS rRNA transcription in response to p53 expression is similar to that observed with UV light treatment of MCF-7 cells, consistent with the idea that p53 regulates these genes in response to DNA damage. Endogenous p53 associates with human snRNA gene promoters. Chromatin immunoprecipitation experiments were then performed to determine whether p53 is directly involved in the regulation of endogenous snRNA genes in response to UV light. As shown in Fig. 2D, substantial enrichment of U1 and U2 snRNA promoter DNA was observed in anti-p53-immunoprecipitated samples (lanes 4 and 5) with chromatin harvested from MCF-7 cells prior to UV light treatment, and these levels were markedly enriched by using chromatin harvested from cells 8 h after UV light treatment. In contrast, only low levels of U6 promoter DNA were enriched in the anti- p53-immunoprecipitated samples prior to UV light treatment, but promoter recovery was noticeably enhanced after treatment. Previously, it was shown that p53 is acetylated within its C terminus in response to DNA damage, which may stimulate DNA binding by p53 (18, 44) and increase 67 recruitment of coregulatory proteins (2). Furthermore, acetylation but not phosphorylation of p53 within the C-terrninal domain contributes to fragile site formation at the U2 snRNA gene loci, indicating that p53 acetylation may be important for p53 function at snRNA genes (71). Therefore, immunoprecipitation reactions were also performed with antibodies that specifically recognize p53 acetylated at K320 or at K372 and K382 (Fig. 2D). Interestingly, significant levels of U1 and U2 promoter enrichment were observed with antibodies that recognize acetylated p53 (lanes 5 and 6), but UV light treatment either did not affect promoter enrichment or caused a modest reduction. In contrast, no significant recovery of U6 promoter DNA was obtained with antibodies that recognize acetylated p53. Enrichment of the p21 promoter (-1.4 kb site) in the anti-p53- immunoprecipitated samples was low prior to UV light treatment, but recovery increased significantly after treatment, as has been previously demonstrated (30). UV light treatment also resulted in increased p21 promoter enrichment for those reactions performed using anti-acetylated p53 antibodies. Together, these data indicate that p53 associates with the endogenous U1 and U2 snRNA gene promoters prior to genotypic stress and UV light stimulates p53 association with these promoters, although the apparent proportion of p53 that is acetylated decreases. Second, low levels of p53 associate with the U6 promoter prior to stress and UV light stimulates p53 promoter association, but this p53 is not acetylated to a significant degree. These observations are in contrast to those seen with the p21 promoter, where p53 association is low but the total p53 level and proportion that is acetylated increase in response to UV light treatment. The data in Fig. 2B show that neither U1 nor p21 promoter association by p53 was observed in HeLa cells (lane 5) using an anti-p53 antibody that efficiently recovered 68 these DNA segments from MCF-7 cells (lane 6). As previously observed for MCF-7 cells, UV light also did not affect SNAPC occupancy at the U1 promoter in HeLa cells (lane 3). Human snRNA molecules are abundant and very stable ( 13, 17, 68) and, thus, it is not clear what effect diminished U1 transcription would. have on overall U1 snRNA levels. Therefore, RT-PCR assays were employed to determine the effect of UV light on steady-state U1 and U6 snRNA levels. As shown in Fig. 2F (top panel), addition of increasing amounts of total cellular RNA harvested from untreated MCF-7 cells resulted in a linear amplification of U1 sequences (lanes 3 to 7), thus demonstrating that this assay is suitable for measuring changes in steady-state U1 snRNA levels. Similar preliminary experiments were performed to determine the range for linear amplification of both U6 snRNA and GAPDH mRNA (data not shown). Under these conditions, U1 steady-state levels were noticeably reduced 8 h after UV light treatment, whereas U6 snRNA and GAPDH mRNA levels remained relatively stable before and after treatment. In three independent replicates of this experiment, the steady-state level of U1 snRNA at 8 h posttreatrnent was 43% of levels in untreated cells (data not shown). The reduction in U1 steady-state levels in response to UV light could be attributable to increased degradation of this RNA, decreased transcription from U1 snRNA genes, or a combination of both factors. U1 snRNA is traditionally viewed as extremely stable, with a half-life greater than 24 h and, thus, little change was expected in steady-state U1 levels by 8 h, even if transcription were completely repressed. The kinetics of the decrease in U1 steady-state levels and the results shown in Fig. 2B suggest that both a reduction in U1 transcription and an increase in U1 snRNA degradation contribute to reduced U1 snRNA levels after UV light treatment. Together, these results indicate that UV light initiates a complicated 69 network of control governing expression of human snRNA genes. Human U1 snRNA gene core promoters contain a high affinity p53 binding site. As a first step towards understanding the mechanism for p53 repression, EMSA were performed to determine whether p53 could bind directly to human U1 snRNA gene promoters. Indeed, recombinant p53 bound extremely well to a U1 probe encompassing the region from -422 to +13 of the promoter (Fig. 3A, lanes 2 to 4). Competition experiments suggest that p53 affinity for the U1 promoter is comparable to the p53 binding element contained within the GADD4S promoter (data not shown). Interestingly, two different complexes formed on the U1 promoter probe, suggesting that the U1 promoter may contain two p53 binding elements. The protein-DNA complexes formed on these probes are due to p53, because inclusion of anti-p53 antibodies in the DNA binding reactions retarded the migration of these complexes (data not shown). To determine the location of the hi gh-affinity p53 binding element, EMSA was also performed with probes containing different regions of the promoter. Strong binding of p53 to DNA was observed in reactions with equivalently labeled probes containing the -312 to +13 and -150 to +13 regions of the promoter. Consistently, low-affinity binding was observed in probes containing the -422 to +152 and -312 to +152 promoter regions. Together, these data indicate that the high-affinity p53 binding element is contained between -150 and +13 of the U1 core promoter. DNase I footprinting experiments were then performed to further map the location of the high-affinity p53 binding element. As shown in Fig. 38, those reactions in which 70 Figure 2-3. Human U1 snRNA gene core promoters contain a high-affinity p53 binding site. (A) EMSA with increasing amounts of recombinant p53 (0, 80, 160, and 320 11g) were performed with double-stranded DNA probes encompassing various regions of the core U1 promoter, as indicated. At higher p53 concentrations, two p53- dependent complexes were formed on those probes that exhibited high-affinity p53 binding. (B) DNase I footprinting reactions were performed using labeled template stand (lanes 1 to 5) or nontemplate stand (lanes 6 to 10) U1 promoter probes encompassing -150 to +13 with increasing amounts of GST-p53 (0, 100, 200, and 400 ng). Digestion of the probe DNA without added p53 is shown in lanes 1, S, 6, and 10. The relative positions of the PSE and transcription start site are indicated. Two protected regions within the template strand (labeled F1 and F2) overlap with the three protected regions (labeled Fl’, F2’, and F 3’) from the nontemplate strand. 71 53 53 53 53 53 -1‘ -4p‘ .1‘ p4 -J‘ c . p53-DNA complexes Free probe 11231.@678H9mngmmummpnmgg A B C D E EMSA -422 +13 A 457 4% B -312 -152 C -312 +13 D 4% H35 72 GST-p53 GST-p53 . -. 9‘ weave-alumna «a» :J—w'. M ' m. ~J~>n - set. '-'~ '.,.’»;) f .g.‘ 1“ near-14.1. 1 2 3 4 5 6 7 8 9 10 DNase lfootprinting 73 the template strand was end labeled exhibited two sites of protection (labeled F1 and F 2) in the presence of GST-p53 (left panel), whereas three regions (F1’, F2’, and F3’) were protected in reactions with the labeled nontemplate strand (right panel). The F1 ’ and F3’ regions correspond to the same region as the F l footprint (hereafter referred to as p53 footprint 1), whereas the F2 and F 2’ regions map to the same region (hereafter referred to as p53 footprint 2). Interestingly, the regions protected by p53 flank the PSE, which is required for DNA binding by SNAPC and is an essential promoter element for high level expression of human snRNA genes. The juxtaposition of p53 footprintsl and 2 with the PSE raises the possibility that p53 represses U1 transcription by occluding SNAPC promoter binding. However, to date we have not observed any effect of p53 on promoter recognition by SNAPc (data not shown). p53 repression and sequence-specific DNA binding are separable activities. As a transcription factor, p53 can either activate or repress transcription, depending upon the structure of the target gene promoter. Notably, p53 can activate transcription from gene promoters that contain consensus p53 binding elements (12). A few examples have been described where p53 represses target genes that contain consensus p53 binding elements (reviewed in reference 26), but it is generally believed that p53 can repress transcription of other target genes whose promoters lack specific p53 recognition elements or contain a noncanonical p53 binding element (29). A comparison of p53 binding elements from target genes that are activated by p53 revealed that a consensus p53 binding site contains two half sites separated by O to 13 bp (50). Each half site contains two quarter sites containing the sequence PuPuPuC(A/T) arranged in a head- 74 to-head orientation. Interestingly, the p53 footprint 1 region contains a sequence that is similar to the consensus p53 binding element usually associated with transcriptional activation by p53 (Fi g. 4A). To investigate the contribution of the high-affinity p53 binding element for U1 promoter recognition and transcriptional repression, a scanning mutagenesis of the U1 core promoter was performed. The sequence of the U1 core promoter region and the location of the p53 footprints are shown in Fig. 4A. This figure also shows the location and identity of the mutations introduced into the U1 core promoter. The ability of recombinant p53 to bind DNA probes harboring these mutations was tested by EMSA (Fig. 4B). The majority of mutations throughout the U1 core promoter, including mutations within the region corresponding to p53 footprint 2 (U 1-4.5 and Ul-4.6 probes), had no significant effect on DNA binding by p53. A slight reduction for p53-DNA complex 2 formation for the U1-4.5 and U1-4.6 probes seen in this figure was not observed in replicates of this experiment. In contrast, mutations within the p53 footprint 1 region caused a marked reduction in p53 binding to the U1 promoter. The strongest effect was seen with the Ul-4.9, U1-4.10, and U1-4.11 probes, whereas the U1-4.8 probe exhibited only a modest reduction in p53 binding. Formation of both p53-DNA complex 1 and p53-DNA complex 2 was affected by these mutations. These data indicate that the region adjacent to the U1 transcriptional start site contains the high-affinity p53 binding element. Next, the requirement of the high-affinity p53 binding element for p53 repression was tested. As shown in Fig. 4C, recombinant p53 effectively repressed transcription from all U1 reporter constructs tested. Neither single nor double sets of mutations within 75 Figure 2-4. The high-affinity p53 element in the U1 promoter is not essential for p53 repression in vitro. (A) Primary sequence of the U1 core promoter region and the location of the p53 footprints. The plasmid pUl-4.0 contains a wild-type promoter sequence from -151 to +13. Scanning mutagenesis across this region was performed, and the introduced mutations and plasmid identity are indicated. Dots represent positions of identity to this wild-type sequence. (B) Mutations in the p53 footprint 1 region disrupt p53 binding. EMSA was performed using the various U1 promoter probes and either 200 or 400 ng of p53, as indicated (lanes 2 to 37). Lane 1 contains the wild-type U1 promoter probe and no added p53. Mutations within the p53 footprint 1 region caused a marked reduction in p53 binding to the U1 promoter (probes 4.8, 4.9, 4.10, and 4.11). (C) Mutations within the p53 footprint 1 region do not affect p53 repression in vitro. Selected mutations were incorporated into a U1 G-less reporter plasmid for in vitro U1 transcription assays (lanes 1 to 20) in the absence or presence of p53 (800 11g). Lane 21 shows transcription from the wild-type Ul reporter in the presence of GST (800 11g). The read-through (RT) and correctly initiated transcripts (U l S ) are indicated. (D) p53 represses transcription of the wild-type and mutant U1 reporter constructs to similar extents. (Left panel) An extensive titration of p53 (0, 20, 40, 80, and 160 ng) into U1 transcription assay mixtures was performed using the wild-type (U1 4.0; lanes 1 to 4) and mutant (U 1 4.9 4.10; lanes 6 to 9) U1 reporter. Lanes 5 and 10 show transcription from the wild-type U1 plasmid in the presence of 160 ng of GST, as a negative control. (Right panel) Transcription levels for two independent experiments were normalized to the signals from transcription reactions containing no added p53, and the average dose- response curves are shown. 76 0.? #9904858084044000ggswwwwu809088§84888408040§80804<80Ugfiwgwguuwugugokbgwghuwg A'— it A v n w l P areas Rd N .éeoe Ba mmn. < 77 2932638» 95> 5 FN om mF 9 RF oF mF 3F mF NF FF 9 m w n o m 3 m N F .m F31 all all i t... . . I! III in? II... I So + - + - + - + - + - + - + - + - + - + - . : : L. ._ _. : iZIil... L. : . oF.v+m.v m.v+m.3 9.3 m3 Q3 m3 F3 0.3 o o.v oF.F~+w.v mm...» 3+ 2:4 >3- :3 .cm >3+ .54 >3- 22o csm >3+ 2:4 >2- :.o a 53525 or m m m m v m _ .|I_ _ _ rum. a mu. . h '_ _i T. 11 _ ...., 1 T. 3L T .21 T in T. -. _ 11 _ I 1L T IL T... II T ll T 11 was m was meI Rio—2 Emwbma: 5 58an o: .90an 5 :. Q<2m __ n_939) O Cl 20 46 so as 100 120 140 {$0 p53 added, ng 128 snRNA (RNAP ll) Class 1 (RNAP Ill) Class 3 (RNAP lll) rRNA gene transcription in the absence of CSB as compared to the extent of p53- mediated SS rRNA gene transcription repression when CSB was present. Together, these data suggest a modulatory role of CSB on p53 transcription repression functions on these promoters. CSB modulates p53-dependent modifications of RNA polymerase III. It has been reported that RNA polymerase 11 large subunit (LS) becomes ubiquitylated and proteolytically degraded following UV light treatment in normal but not in CSB deficient fibroblasts. The degradation of elongation incompetent RNA polymerase at sites of DNA damage may be important for the efficient recovery of transcription as cells recover from stress (3). As with RNA polymerase II, CSB may also affect RNA polymerase III stability. To understand the role of p53 - CSB functional interplay for RNA polymerase III stability, I compared RNA polymerase HI levels in cells with and without functional CSB or p53 under normal conditions and in response to UV light treatment (Figure 3-7A). Western blot analysis of RNA polymerase III levels in CSB deficient cells revealed multiple slower migrating forms of the polymerase III, which may represent post-translational covalently modified forms of RNA polymerase. These modified forms of RNA polymerase III were less pronounced in MCF-7 cells containing functional CSB and were undetectable in HeLa cells. The proportion of the slower migrating forms of RNA polymerase III to the putative unmodified RNA P01ymerase III was the highest in CSB deficient cells (Figure 3-7B), suggesting a negative role of CSB in formation of these forms of RNA polymerase III. Interestingly, 129 Figure 3-7. CSB modulates p53-dependent modifications of RNA polymerase III. (A) The Cockayne syndrome complementation group B (CSB) protein-defective human fibroblast cells (CSB), HeLa and MCF7 cells were treated with UV light and harvested at indicated times afier UV exposure. Western blot analysis revealed slower migrating forms of RNA polymerase III, which were more prominent in untreated CSB cells as compared to MCF-7 cells containing functional CSB. These RNA polymerase III forms were underrepresented in HeLa cells with defective p53. UV light triggered p53 accumulation in CSB and MCF-7 cells and concomitant reduction of modified forms of RNA polymerase III in these cells. (B) Western blot analysis of whole cell extracts from MCF-7 (lanes 1 through 3) and CSB (lanes 4 through 6) cells revealed the enrichment of modified forms of RNA polymerase III in CSB deficient cells as compared to MCF-7 cells. (C) Transient transfection of wild type or acetylation-incompetent KRSA mutant p53 expression plasmids into H1299 (p53-/-) cells causes formation of the higher molecular weight form of RNA polymerase 111. After exposure of H1299 cells to UV light, transient transfection of wild type p53, but not acetylation-incompetent KRSA p53, resulted in formation of the slower migrating form of RNA polymerase III. 130 ncrase 111.11) active human d han'estedat .‘er migrating C 58 cells as 356 111 forms iggered p53 fled forms of extracts from nrichment of rd lO Mil-i RSA mutant 1 the his‘ii’ cells 10 ii" : 101531151 CSB fibroblasts M +UV 0 1 2 4 8 16 hrs ‘ 3 RNA pol Ill ’ - — aCtin HeLa M +UV 0 1 2 4 3 16 hrs .t. --.-. I’jnNApollll Fri" J—p53 -__,_..--—- — actin 123456 MCF7 fl +UV 5.31m... ..... ---- jRNApOIIH nu—fi "'" Western 131 \ Western H1299 (p53—/-) -UV +UV wt p53 KRSA p53 wt p53 KRSA p53 r->. +—>Z ZuJ ZuJ .- J. jRNApoIm F .1!!- ,. __2-_-’_l —p53 F----“d —actln 12345678 Western 132 HeLa cells with non-functional p53 had the smallest fraction of modified forms of RNA polymerase III, suggesting a positive role of p53 in the formation of modified forms of RNA polymerase III. Intriguingly, UV light treatment of cells resulted in reduction of modified forms of RNA polymerase III in CSB and MCF-7 cells, suggesting that these RNA polymerase III modifications may serve as a ‘code’ for subsequent modification in response to stress stimuli (e.g. UV light), which may result in intermediate forms of RNA polymerase III, that are more vulnerable for degradation. To determine whether p53 contributes to RNA polymerase III post-translational modifications, wild type p53 or acetylation-incompetent mutant p53 (KRSA p53) were over-expressed in human osteosarcoma H1299 (p53-/-) cells and levels of RNA polymerase III were evaluated by Western blot analysis (Figure 3-7C). The KRSA p53 was chosen because in contrast to the wild type p53, p53 with mutations that prevent its acetylation was not being able to cause fragile sites induction on U1 and U2 snRNA and SS rRNA gene loci (37), suggesting that p53 acetylation status may be an important determinant for p53 in this process. Upon transient transfection of wild type or KRSA mutant p53 expression plasmids in H1299 cells a slower migrating form of RNA polymerase III was observed. This form of RNA polymerase III was not present in cells transfected with a negative control empty vector. Interestingly, in UV light treated cells over-expression of wild-type p53, but not acetylation-incompetent KRSA p53, resulted in RNA polymerase III modification, which suggests that the acetylation of p53 may be important for RNA polymerase III modifications in UV light treated cells. 133 Discussion A role of CSB in U1 snRNA gene transcription by RNA polymerase II and SS rRNA gene transcription by RNA polymerase III was suggested based on observation that loss of functional CSB in cells causes metaphase fragility of these genes (37). The same study revealed that fragility in the same genes can be induced by over-expression of p53 (24, 37). The data presented herein directly demonstrate a role of CSB in regulation of RNA polymerase II- and III transcription. In contrast to the positive role of CSB in RNA polymerase II-transcribed snRNA genes, CSB has a negative role in RNA polymerase III-transcribed class 1 and 2 genes. In contrast to other RNA polymerase III- transcribed genes, class 3 (U6 snRNA) was not repressed by CSB, but was modestly activated by CSB similarly to RNA polymerase II-transcribed snRNA genes. Class 3 RNA polymerase III promoters are structurally similar to RNA polymerase II-transcribed snRNA gene promoters (18). Furthermore, class 3 RNA polymerase 111 genes share some of the general transcription factors with snRNA genes transcribed by RNA polymerase II (18). Thus, CSB may function as a transcriptional activator or repressor depending on promoter architecture and transcription machinery. A positive role of CSB in RNA polymerase II transcription is suggested from the observation that CSB can modestly stimulate the rate of transcription by RNA polymerase II and thus, may function as an elongation factor (30). The ability of CSB to remodel nucleosomes may also contribute to its functions as a transcriptional activator (8). An interesting feature of the CSB protein is its ability to bind and wrap DNA (1). This could affect the distance between regulatory and core promoter elements and either activate or repress gene transcription. Indeed, snRNA gene transcription activation 134 requires the cooperative binding of Oct-1 and SNAPC (13, 38). This cooperativity is mediated by a positioned nucleosome that resides between the DSE and PSE and brings these two snRNA gene promoter elements in proximity (3 8). The ability of CSB to bind and wrap DNA could also modulate distance between DSE and PSE, thus mimicking the effect of positioned nucleosome on snRNA genes. Also, a negative role of CSB in RNA polymerase III transcription may be the result of CSB—mediated degradation of RNA polymerase III. It has been reported that RNA polymerase 11 large subunit (LS) becomes ubiquitylated and proteolytically degraded following UV light treatment in normal but not in CSB deficient fibroblasts, suggesting that CSB play role in RNA polymerase II degradation. Following UV-irradiation, the degradation of elongation incompetent RNA polymerase at sites of DNA damage may be important for the efficient recovery of transcription as cells recover from stress (3). As with RNA polymerase II, CSB may also affect RNA polymerase III stability. When levels of RNA polymerase III were analyzed in different human cell with different status of CSB and p53 proteins, we observed that CSB deficient cells have lower levels of unmodified RNA polymerase (Figure 3-7B). We also detected slower migrating and likely covalently modified forms of RNA polymerase III in cells (Figure 3-7A). Interestingly, a larger portion and variety of these modified RNA polymerase 111 forms was observed in CSB defective fibroblasts (Figure 3-7A and 3-7B), suggesting that presence of functional CSB in HeLa and MCF-7 cells interferes with the accumulation of modified RNA polymerase III in cells. We speculate that these modified RNA polymerase 111 forms may represent elongation incompetent forms of RNA polymerase III. The absence of CSB-triggered release or degradation of elongation 13S incompetent RNA polymerase 111 may result in the accumulation of these forms of RNA polymerase in CSB deficient cells. Based on the present study I suggest a model for the roles of p53 and CSB in RNA polymerase III modifications and degradation (Figure 3-8). In this model I hypothesize that slower migrating forms of RNA polymerase III represent post- translationally modified elongation incompetent forms of RNA polymerase III. p53 acts as a general repressor of RNA polymerase III transcription by affecting RNA polymerase III transcription elongation or re-initiation, because accumulation of elongation incompetent forms of RNA polymerase III directly correlates with presence of p53 in cells. In contrast to p53, the presence of functional CSB in cells antagonizes accumulation of elongation incompetent forms of RNA polymerase III, suggesting that CSB either helps RNA polymerase III to resume elongation or plays a role in removing elongation incompetent RNA polymerase from the gene, so it can undergo degradation. Cellular stress (e.g. UV light) triggers activation of multiple cellular signaling cascades, which may cause additional modifications of RNA polymerase III, affecting RNA polymerase III gene association and stability. In the future it will be important to determine what types of RNA polymerase III post-translational modifications exist and are induced by p53 in cells. In summary, 1 found that in addition to the role of CSB in RNA polymerase I and II transcription, CSB is also involved in RNA polymerase III transcription. Interestingly, CSB can function both as activator or repressor of RNA polymerase III-transcribed genes depending on promoter architecture. I also discovered a novel role of CSB as a modulator of p53 fimctions. The functional antagonism between p53 and CSB most likely occurs at 136 Figure 3-8. Model: Roles of p53 and CSB in RNA polymerase III elongation and stability. p53 plays a role in RNA polymerase III modifications, which associates with elongation incompetent RNA polymerase. In contrast, CSB helps RNA polymerase IH to resume elongation. In response to UV light exposure elongation incompetent RNA polymerase 111 could be a substrate for additional modifications triggered by UV light activated signaling cascades. This results in dissociation of RNA polymerase III from gene and its subsequent degradation. 137 Ecow Econ. @ cozmpfimwo ESwnESE cozmmcsm 283885 :ozmmcofi poEpoE €5.98 mepoE 22298 Eco“ octmmcom Reigngw 1“ >=+ ENE rgatrorr Ind ociata with nerase llllo petent RNA by W light as: HI from the level of RNA polymerase III stability. As p53 triggers RNA polymerase to an elongation incompetent mode, CSB works antagonistically by helping RNA polymerase III to resume elongation or targeting the elongation incompetent RNA polymerase III for destruction. Cockayne syndrome (CS) is a rare autosomal disease that caused by mutations in CSB. Interestingly, mutations in CSB cause at least four different diseases: Cockayne syndrome (CS), UV-sensitive syndrome (UVSS), DeSanctis-Cacchione syndrome (DS-C) and cerebro—oculo-facio-skeletal syndrome (COFS) (25, 31), suggesting that CSB may be involved in different cellular functions possibly via its general role in transcription by all three RNA polymerases. The CSB role in transcription of non-coding RNAs by both RNA polymerase II and III is intriguing. Non-coding RNAs have been shown to be involved in controlling a variety of cellular functions via their role in transcription and RNA metabolisms (l 1, 12, 22, 26, 36) and misregulations of some non-coding RNAs were recently linked to different human diseases such as human hereditary cartilage-hair hyp0plasia disease (28) and cancer (21, 32). Multiple transcription targets for CSB helps to explain multiple phenotypes of CS patients. 139 References 10. Beerens, N., J. H. Hoeijmakers, R. Kanaar, W. Vermeulen, and C. Wyman. 2005. The CSB protein actively wraps DNA. J Biol Chem 280:4722-9. Bradsher, J., J. Auriol, L. Proietti de Santis, S. Iben, J. L. Vonesch, I. Grummt, and J. M. Egly. 2002. CSB is a component of RNA pol I transcription. Mol Cell 10:819-29. Bregman, D. B., R. Halaban, A. J. van Gool, K. A. Henning, E. C. Friedberg, and S. L. Warren. 1996. UV-induced ubiquitination of RNA polymerase II: a novel modification deficient in Cockayne syndrome cells. Proc Natl Acad Sci U S A 93:11586-90. Cairns, C. A., and R. J. White. 1998. p53 is a general repressor of RNA polymerase III transcription. Embo J 17 :31 12-23. Carey, M. a. S. T. S. 2000. Transcriptional regulation in eukaryotes. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY. Chesnokov, 1., W. M. Chu, M. R. Botchan, and C. W. Schmid. 1996. p53 inhibits RNA polymerase III-directed transcription in a promoter-dependent manner. Mol Cell Biol 16:7084-8. Christiansen, M., T. Stevnsner, C. Modin, P. M. Martensen, R. M. Brosh, Jr., and V. A. Bohr. 2003. Functional consequences of mutations in the conserved SF2 motifs and post-translational phosphorylation of the CSB protein. Nucleic Acids Res 31 :963-73. Citterio, E., V. Van Den Boom, G. Schnitzler, R. Kanaar, E. Bonte, R. E. Kingston, J. H. Hoeijmakers, and W. Vermeulen. 2000. ATP-dependent chromatin remodeling by the Cockayne syndrome B DNA repair-transcription- coupling factor. Mol Cell Biol 20:7643-53. Durnam, D. M., J. C. Menninger, S. H. Chandler, P. P. Smith, and J. K. McDougall. 1988. A fragile site in the human U2 small nuclear RNA gene cluster is revealed by adenovirus type 12 infection. Mol Cell Biol 8: 1 863-7. Eisen, J. A., K. S. Sweder, and P. C. Hanawalt. 1995. Evolution of the SNF2 family of proteins: subfamilies with distinct sequences and functions. Nucleic Acids Res 23:2715-23. 140 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. Espinoza, C. A., T. A. Allen, A. R. Hieb, J. F. Kugel, and J. A. Goodrich. 2004. B2 RNA binds directly to RNA polymerase II to repress transcript synthesis. Nat Struct Mol Biol 11:822-9. Fong, Y. W., and Q. Zhou. 2001. Stimulatory effect of splicing factors on transcriptional elongation. Nature 414:929-33. 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-l coactivator OBF-l. Genes Dev 12:3528-40. Gargano, S., P. Wang, E. Rusanganwa, and S. Bacchetti. 1995. The transcriptionally competent U2 gene is necessary and sufficient for adenovirus type 12 induction of the fragile site at 17q21—22. Mol Cell Biol 15:6256-61. Gridasova, A. A., and R. W. Henry. 2005. The p53 tumor suppressor protein represses human snRNA gene transcription by RNA polymerases II and III independently of sequence-specific DNA binding. Mol Cell Biol 25:3247-60. Hanawalt, P. C. 2001. Controlling the efficiency of excision repair. Mutat Res 485:3-13. Henry, R. W., V. Mittal, B. Ma, R. Kobayashi, and N. Hernandez. 1998. SNAP19 mediates the assembly of a functional core promoter complex (SNAPc) shared by RNA polymerases II and III. Genes Dev 12:2664-72. Hernandez, N. 1992. Transcription of vertebrate snRNA genes and related genes, p. 281- 313. In S. McKnight and K. Yamamoto (ed.,) Transcriptional Regulation. Cold Spring Harbor Laboratory Press, Cold Spring Harbor. Hirsch, H. A., G. W. Jawdekar, K. A. Lee, L. Gu, and R. W. Henry. 2004. Distinct mechanisms for repression of RNA polymerase III transcription by the retinoblastoma tumor suppressor protein. Mol Cell Biol 24:5989-99. Hoffman, R. W., T. Gazitt, M. F. Foecking, R. A. Ortmann, M. Misfeldt, R. Jorgenson, S. L. Young, and E. L. Greidinger. 2004. U1 RNA induces innate immunity signaling. Arthritis Rheum 50:2891-6. Krawczak, M., J. Reiss, and D. N. Cooper. 1992. The mutational spectrum of single base-pair substitutions in mRNA splice junctions of human genes: causes and consequences. Hum Genet 90:41-54. Kwek, K. Y., S. Murphy, A. Furger, B. Thomas, W. O'Gorman, H. Kimura, N. J. Proudfoot, and A. Akoulitchev. 2002. U1 snRNA associates with TFIIH and regulates transcriptional initiation. Nat Struct Biol 9:800-5. 141 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. Leadon, S. A., and P. K. Cooper. 1993. Preferential repair of ionizing radiation- induced damage in the transcribed strand of an active human gene is defective in Cockayne syndrome. Proc Natl Acad Sci U S A 90:10499-503. Li, Z., A. Yu, and A. M. Weiner. 1998. Adenovirus type 12-induced fragility of the human RNU2 locus requires p53 function. J Virol 72:4183-91. Licht, C. L., T. Stevnsner, and V. A. Bohr. 2003. Cockayne syndrome group B cellular and biochemical functions. Am J Hum Genet 73: 1217-39. Nguyen, V. T., T. Kiss, A. A. Michels, and O. Bensaude. 2001. 7SK small nuclear RNA binds to and inhibits the activity of CDK9/cyclin T complexes. Nature 414:322-5. Richards, R. I. 2001. Fragile and unstable chromosomes in cancer: causes and consequences. Trends Genet 17 :339-45. Ridanpaa, M., H. van Eenennaam, K. Pelin, R. Chadwick, C. Johnson, B. Yuan, W. vanVenrooij, G. Pruijn, R. Salmela, S. Rockas, O. Makitie, I. Kaitila, and A. de la Chapelle. 2001. Mutations in the RNA component of RNase MRP cause a pleiotropic human disease, cartilage-hair hypoplasia. Cell 104:195-203. Schramayr, S., D. Caporossi, I. Mak, T. Jelinek, and S. Bacchetti. 1990. Chromosomal damage induced by human adenovirus type 12 requires expression of the ElB SS-kilodalton viral protein. J Virol 64:2090-5. Selby, C. P., and A. Sancar. 1997. Cockayne syndrome group B protein enhances elongation by RNA polymerase II. Proc Natl Acad Sci U S A 94:11205- 9. Spivak, G. 2004. The many faces of Cockayne syndrome. Proc Natl Acad Sci U S A 101:15273-4. Susani, M., A. Haitel, and M. Marberger. 2004. Prognostic factors in Bilharzial-related bladder cancer. Hum Pathol 35:1 167-8; author reply 1168. Troelstra, C., R. M. Landsvater, J. Wiegant, M. van der Ploeg, G. Viel, C. H. Buys, and J. H. Hoeijmakers. 1992. Localization of the nucleotide excision repair gene ERCC6 to human chromosome qu1 1-q21. Genomics 12:745-9. van Cool, A. J., E. Citterio, S. Rademakers, R. van Os, W. Vermeulen, A. Constantinou, J. M. Egly, D. Bootsma, and J. H. Hoeijmakers. 1997. The Cockayne syndrome B protein, involved in transcription-coupled DNA repair, resides in an RNA polymerase II-containing complex. Embo J 16:5955-65. 142 35. 36. 37. 38. Venema, J., L. H. Mullenders, A. T. Natarajan, A. A. van Zeeland, and L. V. Mayne. 1990. The genetic defect in Cockayne syndrome is associated with a defect in repair of UV-induced DNA damage in transcriptionally active DNA. Proc Natl Acad Sci U S A 87 :4707-11. Yang, Z., Q. Zhu, K. Luo, and Q. Zhou. 2001. The 7SK small nuclear RNA inhibits the CDK9/cyclin Tl kinase to control transcription. Nature 414:317-22. Yu, A., H. Y. Fan, D. Liao, A. D. Bailey, and A. M. Weiner. 2000. Activation of p53 or loss of the Cockayne syndrome group B repair protein causes metaphase fragility of human U1, U2, and 5S genes. Mol Cell 5:801-10. Zhao, X., P. S. Pendergrast, and N. Hernandez. 2001. A positioned nucleosome on the human U6 promoter allows recruitment of SNAPC by the Oct- 1 POU domain. Mol Cell 7:539-49. 143 CHAPTER FOUR SUMMARY Non-translated snRNAs perform essential cellular functions by controlling RNA synthesis and RNA metabolism (8). The involvement of snRNAs in controlling diverse cellular functions suggests that these genes should be tightly regulated under normal conditions and in response to cellular stress. Indeed, misregulation of some snRNAs have been recently linked to several human diseases, such as cartilage-hair hypoplasia (CHH) disease (15) and cancer (13, 17). In addition, both tumor suppressor proteins (e. g. p53 (3, 4) and RB (6)) and oncoproteins (e. g. Myc (6) and CKII (7,10, 11)) have been linked to snRNA gene transcription, suggesting that regulated snRNA production plays a role in controlling cellular homeostasis. Depending on the promoter architecture, snRNA genes are transcribed by either RNA polymerase II or by RNA polymerase III (8). Regardless of RNA polymerase specificity, snRNA genes have similar promoter structure and share some of the general transcription factors (reviewed in 8), suggesting that these genes may be similarly regulated in response to cellular stress stimuli. The overall goal of my research project was to understand the role of the tumor suppressor protein p53 in human snRNA gene transcription. The tumor suppressor protein p53 is a multifunctional transcription factor known to control cellular proliferation and genomic stability by activating or repressing transcription of target genes by all three classes of RNA polymerases (3-5, 16, 21). It was previously 144 demonstrated that p53 represses snRNA gene transcription by RNA polymerase III (3, 4); however, the importance of p53 for transcription of RNA polymerase II-transcribed snRNA genes was not investigated. My studies revealed that in addition to the role of p53 in snRNA gene transcription by RNA polymerase III, p53 also represses snRNA gene transcription by RNA polymerase 11. Thus, p53 is a general repressor of snRNA gene transcription by both polymerases. I found that p53 associates with human U1 and U6 snRNA genes during transcription repression and represses endogenous snRNA gene transcription upon transient transfection or in response to UV-light, suggesting a role of p53 in snRNA gene transcription regulation in response to DNA damage. It has been shown that p53 can repress transcriptiOn of its target genes by a variety of mechanisms that regulate pre—initiation complex formation, elongation by RNA polymerase and at the level of chromatin (reviewed in 9). It was proposed that p53- mediated repression of RNA polymerase III transcription occurs via TBP-mediated p53 interaction with TFIIIB, which interferes with TFIIIB binding and subsequent recruitment of RNA polymerase III to RNA polymerase III promoters (3, 5). In contrast, similar levels of TBP at snRNA gene promoters before and during p53-mediated transcription repression were observed, suggesting that p53 interferes with snRNA gene transcription at steps occurring afier TBP or TFIIIB recruitment for RNA polymerase II- and III-transcribed genes respectively. Indeed, RNA polymerase III was not detected at U6 snRNA gene promoters during repression by p53, suggesting that p53 interferes with RNA polymerase III recruitment to snRNA gene promoters. Whether p53 also affects the recruitment of RNA polymerase II to snRN A gene promoters remains to be investigated. 145 Interestingly, upon p53 accumulation on snRNA gene promoters the concomitant enrichment of HDACs was observed, suggesting that p53 repression of snRNA gene transcription may in part occur by recruitment of the chromatin remodeling activities of HDACs. The HDAC-mediated p53 transcription repression has been described for some genes (12, 14) and the role of HDACs in deacetylation of core histones as well as p53 itself was suggested (12). What are the determinants for p53 decision to recruit HDAC for transcription repression? Recently, SUMO modification of several transcription factors has been linked to HDACs recruitment and transcription repression (19). Interestingly, I observed accumulation of sumoylated forms of p53 in cells after UV light treatment. When the proportion of sumoylated protein population was analyzed on snRNA gene promoters, the direct correlation between levels of total p53, sumoylated proteins and accumulation of HDACs on snRNA gene promoters was detected, suggesting that promoter-bound sumoylated p53 may recruit HDACs during transcription repression of snRNA genes. As part of our investigation of the mechanism of p53-mediated snRNA gene transcription repression, I tested a hypothesis that p53 represses RNA polymerase II- transcribed snRNA genes by interfering with CSB-mediated elongation causing stalling of RNA polymerase II (20). Our studies reveal a direct role of CSB not only in snRNA gene transcription by RNA polymerase II, but also in transcription by RNA polymerase III, including class 3 RNA polymerase III-transcribed snRNA genes. Thus, CSB may be targeted by p53 during snRNA gene transcription repression by both polymerases. Indeed, our data suggests that CSB modulates p53 transcription repression functions and, in the absence of CSB, p53 exhibits stronger repression of U1 snRNA and SS rRNA gene 146 transcription. In addition to the role of CSB as an elongation factor for RNA polymerase II-transcribed genes, CSB has also been reported to play a role in ubiquitylation and degradation of RNA polymerase 11 large subunit (LS) following UV light treatment (2); however, whether CSB also plays a role in RNA polymerase III stability was uncertain. Degradation of elongation incompetent forms of RNA polymerases at sites of DNA damage may be important for the efficient recovery of transcription as cells recover from stress. Interestingly, when we compared RNA polymerase III levels in human cells with different status of CSB, we observed that levels of RNA polymerase III are lower in CSB deficient cells, suggesting that similarly to RNA polymerase II, CSB may also affect RNA polymerase III stability. However, we found that CSB defective fibroblasts have a larger portion of slower migrating and possibly covalently modified forms of RNA polymerase 111. As CSB was described as an elongation factor for RNA polymerase II, we speculate that these modified forms of RNA polymerase III in CSB deficient fibroblasts represent elongation incompetent forms of RNA polymerase III. Interestingly, as p53 accumulation in cells correlates with RNA polymerase III transcription repression, it also results in enrichment of slower migrating forms of RNA polymerase III in cells. Thus, I hypothesized that p53 represses elongation by RNA polymerase through its post- translational modifications. At this point it is not clear what kinds of covalent modifications of RNA polymerase III exist in cells and which modifications serve as ‘markers’ for elongation incompetent RNA polymerase 111. As of now, these modifications may represent either phosphorylated, ubiquitylated, sumoylated or neddylated forms of RNA polymerase 111. So far, only ubiquitin-modification of proteins are considered to be a marker for protein degradation by the 26S proteasome (18). 147 Though both mono- and poly—ubiquitylation of proteins were described, poly- ubiquitylated proteins are more likely be targeted to proteasomal degradation (18). The function for protein mono-ubiquitylation has not been yet well characterized. It was proposed that mono-ubiquitylation is a prerequisite for poly-ubiquitylation and several ubiquitin-ligases are only capable to mono-ubiquitylate their substrates (1). We speculate that p53 plays a role in mono-ubiquitylation of RNA polymerase III, which results in elongation incompetent forms of RNA polymerase III. What is the mechanism for p53- mediated RNA polymerase III modification? We do not yet have an answer to this question. It is possible that p53 itself serves as an E3 ubiquitin ligase for RNA polymerase III modification; however, to-date neither p53 interactions with polymerases nor p53 activity as an E3 ubiquitin ligase were reported. An other possibility is that p53 may recruit ubiquitin li gases (e. g. Mdm2 and/or others) during transcription repression. Together, our data suggests that p53-mediated snRNA gene transcription repression is complex multilevel process, which may be important to assure snRNA gene repression by p53 in response to cellular stress. 148 References 10. Alarcon-Vargas, D., and Z. Ronai. 2002. p5 3-Mdm2--the affair that never ends. Carcinogenesis 23:541-7. Bregman, D. B., R. Halaban, A. J. van Gool, K. A. Henning, E. C. Friedberg, and S. L. Warren. 1996. UV-induced ubiquitination of RNA polymerase II: a novel modification deficient in Cockayne syndrome cells. Proc Natl Acad Sci U S A 93: 1 1586-90. Cairns, C. A., and R. J. White. 1998. p53 is a general repressor of RNA polymerase III transcription. EmboJ 17:3112-23. Chesnokov, I., W. M. Chu, M. R. Botchan, and C. W. Schmid. 1996. p53 inhibits RNA polymerase III-directed transcription in a promoter-dependent manner. Mol Cell Biol 16:7084-8. Crighton, 1)., A. Woiwode, c. Zhang, N. Mandavia, .r. P. Morton, L. J. Warnock, J. Milner, R. J. White, and D. L. Johnson. 2003. p53 represses RNA polymerase III transcription by targeting TBP and inhibiting promoter occupancy by TFIIIB. Embo J 22:2810-20. Felton-Edkins, Z. A., N. S. Kenneth, T. R. Brown, N. L. Daly, N. Gomez- Roman, C. Grandori, R. N. Eisenman, and R. J. White. 2003. Direct regulation of RNA polymerase III transcription by RB, p53 and c-Myc. Cell Cycle 2:181-4. Gu, L., W. J. Esselman, and R. W. Henry. 2005. Cooperation between small nuclear RNA-activating protein complex (SNAPC) and TATA-box-binding protein antagonizes protein kinase CK2 inhibition of DNA binding by SNAPC. J Biol Chem 280:27697-704. Hernandez, N. 1992. Transcription of vertebrate snRNA genes and related genes, p. 281-313. In S. McKnight and K. Yamamoto (ed.), Transcriptional Regulation. Cold Spring Harbor Laboratory Press, Cold Spring Harbor. He, J., and S. Benchimol. 2003. Transcriptional repression mediated by the p53 tumour suppressor. Cell Death Differ 10:404-8. Hu, P., K. Samudre, S. Wu, Y. Sun, and N. Hernandez. 2004. CK2 phosphorylation of del executes cell cycle-specific RNA polymerase III transcription repression. Mol Cell 16:81-92. 149 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. Hu, P., S. Wu, and N. Hernandez. 2003. A minimal RNA polymerase III transcription system from human cells reveals positive and negative regulatory roles for CK2. Mol Cell 12:699—709. Juan, L. J., W. J. Shia, M. H. Chen, W. M. Yang, E. Seto, Y. S. Lin, and C. W. Wu. 2000. Histone deacetylases specifically down-regulate p53-dependent gene activation. J Biol Chem 275:20436-43. Krawczak, M., J. Reiss, and D. N. Cooper. 1992. The mutational spectrum of single base-pair substitutions in mRNA splice junctions of human genes: causes and consequences. Hum Genet 90:41-54. Murphy, M., J. Ahn, K. K. Walker, W. H. Hoffman, R. M. Evans, A. J. Levine, and D. L. George. 1999. Transcriptional repression by wild-type p53 utilizes histone deacetylases, mediated by interaction with mSin3a. Genes Dev 13:2490-501. Ridanpaa, M., H. van Eenennaam, K. Pelin, R. Chadwick, C. Johnson, B. Yuan, W. vanVenrooij, G. Pruijn, R. Salmela, S. Rockas, O. Makitie, I. Kaitila, and A. de la Chapelle. 2001. Mutations in the RNA component of RNase MRP cause a pleiotropic human disease, cartilage-hair hypoplasia. Cell 104:195-203. Stein, T., D. Crighton, L. J. Warnock, J. Milner, and R. J. White. 2002. Several regions of p53 are involved in repression of RNA polymerase III transcription. Oncogene 21 :5540-7. Susani, L., A. Pangrazio, C. Sobacchi, A. Taranta, G. Mortier, R. Savarirayan, A. Villa, P. Orchard, 1’. Vezzoni, A. Albertini, A. Frattini, and F. Pagani. 2004. TCIRGl-dependent recessive osteopetrosis: mutation analysis, functional identification of the splicing defects, and in vitro rescue by U1 snRNA. Hum Mutat 24:225-35. Welchman, R. L., C. Gordon, and R. J. Mayer. 2005. Ubiquitin and ubiquitin- like proteins as multifunctional signals. Nat Rev Mol Cell Biol 6:599-609. Yang, S. H., and A. D. Sharrocks. 2004. SUMO promotes HDAC-mediated transcriptional repression. Mol Cell 13:611-7. Yu, A., H. Y. Fan, D. Liao, A. D. Bailey, and A. M. Weiner. 2000. Activation of p53 or loss of the Cockayne syndrome group B repair protein causes metaphase fragility of human U1, U2, and SS genes. Mol Cell 5:801-10. Zhai, W., and L. Comai. 2000. Repression of RNA polymerase I transcription by the tumor suppressor p53. Mol Cell Biol 20:5930-8. 150 APPENDIX POSSIBLE ROLE OF HDACs IN P53-MEDIATED snRNA GENE TRANSCRIPTION REPRESSION p53 has been shown to repress transcription of its target genes by mechanisms that regulate pre-initiation complex formation and elongation by RNA polymerase. In addition, transcriptional repression by p53 can be also achieved through p53-mediated alteration of chromatin (reviewed in 6). The ability of p53 to repress transcription at the level of chromatin was linked to recruitment of histone deacetylases (HDACs) to target genes (7, 8). p53 was shown to associate with HDACs via direct interaction of p53 with Sin3a protein of the Sin3/HDAC co-repressor complex (8). Recruitment of HDACs by p53 is believed to result in p53- mediated repression by at least two different mechanisms: core histone deacetylation and p53 deacetylation (7). HDAC-mediated histone deacetylation results in chromatin condensation and reduced promoter accessibility to the transcriptional machinery and/or co-activators (l 1). Consistent with the positive role of p53 acetylation for transactivation activity of p53 (1), p53 deacetylation by HDACs results in p53-mediated transrepression of target gene transcription (7, 8). To test whether HDACs are involved in snRNA gene transcription repression, snRNA gene promoter occupancy by HDACs was analyzed using immunoprecipitation experiments (Figure A-l). As shown in figure A-lA, p53 and HDACl and 2 occupancy on the endogenous U1 snRNA gene promoter was concomitantly increased in response to 151 Figure A-l: Endogenous HDAC] and HDAC2 associate with human snRNA gene promoters. (A) Chromatin from untreated or UV light treated MCF—7 cells was immunoprecipitated with SNAP43 antibodies, antibodies against total p53 (21-25) or K320 acetylated p53, and HDACl, HDAC2 or negative control IgG antibodies (lanes 3 through 8, respectively). Lanes 1 and 2 represent input titration. p53, I-IDACl and HDAC2 occupancy on U1 snRNA promoter (but not on negative control U1 upstream region) was increased after UV light treatment. No change in U1 snRNA promoter occupancy by acetylated p53 was observed after UV light exposure. (B) A portion of untreated or p53 or GST treated U6 in vitro transcription reaction mixtures was cross- linked with formaldehyde and subjected to immunoprecipitation with anti-TBP, IgG, anti-p53, anti-RNA polymerase III, anti-HDACI and anti-HDAC2 antibodies (lanes 2 through 7, respectively). Enrichment of the U6 reporter plasmids or negative control pUC119 DNA was compared by PCR using promoter-specific or pUC-specific primers. Lane 1 represents 10% input DNA. 152 rrRVA gene 7 cells was (2135,) or lies (lanesi IDACI and fl upstream A prometct 1 portion of 3 W35 CIOSS' -lBP. lgG. ies (12111653 tire cont-'01 tic primers. ]U1 snRNA l 3 8 g e m 9'. < o o c — < 00 m < < o\° r'. a? 9 ‘3- 3.: I. G r- O 5 5 5 5 5 9 — - CD- Or- -Uv — . . — N H u. — +UV ~ F‘- -UV - -—~ +UV 1 2 3 4 5 6 7 8 Chromatin IP 6 v- N n. O O O. m < < < a 'T <5 ‘.1 ‘F 3.3 3.: E 5 2’ 5 5 5 5 m - . "" U6 promoter _ ........ ..... _. I... ._..... plasmid - m .- pUC119 — 1 2 3 4 5 6 7 Cross-linking IP 153 U1 Upstream Control Untreated p53 treated GST treated GST treated UV light treatment, suggesting that HDACs are involved in snRNA gene transcription in response to UV light treatment and that p53 may recruit HDACs to U1 snRNA gene promoters during transcription repression. Specific enrichment of HDACl and 2 was also observed on exogenous U6 snRNA gene promoters during p53-mediated repression using in vitro transcription assays (Figure A-lB, lanes 6 and 7 of the p53 treated reactions); however, no recovery of U6 snRNA gene promoters was observed in anti-HDAC] and 2 immunoprecipitated samples from untreated or GST treated transcription reactions. Together, these data suggest that p53 may recruit HDAC 1 and 2 during snRNA gene transcription repression. What are the determinants for p53-mediated recruitment of HDACs during transcription repression? Recently, it has been reported that sumoylation of proteins may promote HDAC recruitment and transcription repression (12). It has been shown that p53 can be sumoylated at Lys3 86 (9) and both positive and negative roles of sumoylation for p53-mediated transactivation of target genes were described (2, 9, 10). Thus, we asked whether p53 sumoylation mediates HDACs recruitment and transcription repression by p53. Interestingly, as UV light exposure results in accumulation of total p53 as well as sumo-modified forms of p53 (Figure A-2A), the concomitant enrichment of p53 and sumoylated protein population was observed on the endogenous U1 snRNA gene promoters in response to UV light treatment in chromatin immunoprecipitation experiments (Figure A-2B). Since there are no commercially available antibodies against sumoylated p53, we can only speculate that U1 snRNA gene promoter occupancy by sumoylated p53 was also enriched in response to UV light exposure and contribute to the signal from the total sumoylated protein population recognized by anti-Sumo-l 154 Figure A-2: Sumoylated protein population is enriched on U1 snRNA gene promoters after UV exposure. (A) UV light exposure of MCF-7 cells results in accumulation of total p53 as well as sumo-modified forms of p53 as measured by Western blotting. Whole cell extracts from untransfected and pCDNA-HA-Sumo-l, -2, and -3 cotransfected MCF-7 cell harvested prior and 8 hours after UV light treatment were analyzed by 12.5% SDS-PAGE and Western blot analysis of HA-tagged sumoylated proteins (lanes 1 and 2) and p53 (lanes 3 through 6). Co-migrating bands that were detected by both anti-HA and anti-p53 antibodies were labeled as SUMO-p53. (B) Chromatin from untreated or UV light treated MCF-7 cells was immunoprecipitated with antibodies against total and K320 acetylated p53, antibodies against Sumo-1 or negative control IgG antibodies (lanes 3 through 6, respectively). Lanes 1 and 2 represent input titration. p53 and sumoylated proteins were concomitantly enriched at endogenous U1 snRNA gene promoters after UV light treatment. No change in U1 snRNA promoter occupancy by acetylated p53 was observed after UV light exposure. 155 SDRVA gene €115 lesultsia ‘ ”16215111611113 A-Sumo-l, .1 11 ght treatnent )1 PIA-tagged ting bands Lit IMO-p53. (Bl :cipitated with -1 or negatn'e :present input idogenous 11 NA propels: - ' ' + + uv - + - + - +'Sumo-HA 190.. 120- 85 - i a" . W . -. \-¢ “—- ‘._. ‘. :SUMo_p53 60 — 50 — ‘7 w :1 -2 21953 a-HA a-p53 Western A co g a +-' .D O ’5 3- 3‘. E3 '7 C o (O Q) °\° '7 9 ‘5 “P o H -UV ....... .. .. .. .. +UV]U1 SnRNA L: -UV]U1 Upstream +UV Control 1 Chromatin IP 156 antibodies (lane 5). Thus, we observed direct correlation between levels of p53, sumoylated proteins and HDACl and 2 on U1 snRNA promoters after UV light treatment (Figures A-1 and A-2B), suggesting that sumo-modified p53 may recruit HDACs for repression of snRNA gene transcription. I further tested whether Sumo-modification of p53 is required for p53-mediated snRNA gene transcription repression by comparing the ability of wild type GST-p53 and GST-p53 (K386R) to repress U1 and U6 snRNA gene transcription (Figure A-3). Both GST-p53 and GST-p53 (K386R) were expressed in E. coli and supposedly lack any post- translational modifications. However, during in vitro transcription assays these proteins could undergo covalent modifications by enzymes present in the HeLa nuclear extract. Thus, during in vitro transcription wild type GST-p53 could become sumoylated; in contrast, GST-p53 (K386R) should lack the sumo-modification. These experiments revealed that sumoylation-deficient mutant GST-p53 (K3 86R) repressed both U1 snRNA gene transcription by RNA polymerase II and U6 snRNA gene transcription by RNA polymerase III approximately two fold less efficiently than wild type GST-p53, suggesting that sumoylation of p53 may contribute to p53-mediated snRNA gene transcription repression. A possible explanation of the modest effect of p53 K386R mutation on snRNA gene transcription repression is that in vitro transcription assays were performed using naked DNA, but not a chromatinized template. If p53 sumoylation is indeed important for HDAC recruitment, HDAC-mediated repression of snRNA genes may require a chromatin context. Indeed, we did not observe any effect of the HDAC inhibitor sodium butyratc on p53-mediated snRNA gene transcription repression using non-chromatinized DNA templates (data not shown). In addition, the proportion of wild 157 Figure A-3: p53 sumoylation contributes to p53-mediated snRNA gene transcription repression in vitro. Recombinant wild type GST-p53 (lanes 2 through 4) and GST-p53 (K386R) (lanes 6 through 8) were titrated into U1, U6 and AdML in vitro transcription reactions. 0.5, 1 and 2 pg of proteins were used in U1 and AdML in vitro transcription assays and 0.15, 0.3 and 0.6 pg of proteins were added into U6 in vitro transcription mixtures. Heat-inactivated GST-p53, GST-p53 (K386R) and GST were also tested in these transcription reactions (lanes 5, 9 and 10, respectively). Lane 1 shows level of transcription when no proteins were added. 158 iptiorr I'll-’13 +GST-p53 +GST-p53 (K386R) I I .. “Latina I W HI ('7) ‘ ‘ 1° _ + intinn . ‘ urn- ~-- - . -~- - u..- —-- Orr- ~ ~- ---o *- _U1 5 ~{3.1011 u..— .— “ .... . «n.- 1“ ——- «— r *’ —'. w — U6 5I tedir: m-—-——_.~— —AdM|_ V61 01 1 2 3 4 5 6 7 8 9 10 In vitro transcription 159 Figure A-4: Sumoylated p53 is enriched in acetylated population of p53 in vivo. Whole cells extracts from UV light treated MCF-7 cells expressing HA-Sumo-l, -2 and — 3 proteins were immunoprecipitated with either full length p53 antibodies (lane 4), antibodies against p53 acetylated at K320 (lane 5) or negative control IgG antibodies (lane 3). Presence of p53 and Sumo-p53 was detected by immunoblotting with another p53 (Ab-6) (lefi panel) and anti-HA (right panel) antibodies. 160 p53 in 1111. 10-1, -2 and- lies (lane 4) ;G antibodies with another or-p53 Westem 161 oe-HA Westem «Sumo-p53 type p53 that gets sumoylated during in vitro transcription is low and majority of the wild type GST-p53 was not sumoylated, but may repress transcription via sumoylation- HDAC-independent mechanism. Thus, it would be useful to know what proportion of the wild type p53 (if any) gets sumoylated during in vitro transcription assays. Interestingly, I observed that sumoylated forms of p53 are enriched in acetylated p53 population, suggesting that acetylation may be a prerequisite for a subsequent sumoylation of p53. As shown in figure A-4, despite the fact that more p53 was immunoprecipitated with antibodies against total cellular p53 (lane 4, left panel) as compared to acetylated p53 (lane 5, left panel), levels of sumoylated p53 in these immunoprecipitated materials were comparable. Materials and Methods Cell culture and UV irradiation Human MCF-7 cells were maintained in Dulbecco’s modified Eagle’s medium supplemented with penicillin-streptomycin and 10% fetal bovine serum. Cells grown to 70 to 80% confluence were washed with phosphate-buffered saline and irradiated with 50 J of UV light (254-nm peak)/m2 by using a UV Stratalinker (Stratagene). After irradiation, growth medium was added and cells were incubated at 37°C under 5% C02 for the indicated times. Additionally, MCF-7 cells were grown to 50% confluence in ISO-mm plates and were then transiently co-transfected with 5 pg of pCDNA3-HA-Sumo-l, pCDNA3-HA-Sumo-2 and pCDNA3-HA-Sumo-3 plasmids (gift from Dr. Kwok, University of Michigan) using 3 pl Lipofectin reagent (Invitrogen) per transfection. Transfections were performed for 6 h. Subsequently, the medium was replaced and cells 162 were incubated for 48 h for further analysis. Expression and purification of recombinant proteins Glutathione S—transferase (GST), GST-tagged. full-length human p53 and GST were expressed in Escherichia coli BL21 (DE3) codon+ cells (Stratagene) and were affinity purified by binding to glutathione agarose beads (Sigma). GST-p53 and GST proteins were then eluted from beads in HEMGT-ISO buffer containing 50 mM glutathione for 4 h at 4°C and concentrated by centrifugation using a Centricon YM-3O spin column (Millipore) in HEMGT-80 buffer (20 mM HEPES [pH 7.9], 0.1 mM EDTA, 10 mM MgC12, 10% glycerol [vol/vol], 0.1% Tween 20, 80 mM KCl) containing protease inhibitors and 1 mM dithiothreitol. In vitro transcription assays In vitro transcription assays were performed as described previously (4) using 18, 2, and 10 pL of HeLa cell nuclear extract for the U1 snRNA, U6 snRNA, and adenovirus major late promoter (AdML) transcription reactions, respectively. The pUl-4.0 (1 pg), pU6/Hae/RA.2 (250 ng), and M13-AdML (250 ng) templates were used for the U1 snRNA, U6 snRNA, and AdML transcription reactions, respectively. Purified proteins were added in the amounts indicated in the figure legends. Transcription was performed for l h at 30°C. Transcripts were separated by denaturing 6% polyacrylamide gel electrophoresis and visualized by autoradiography. 163 Chromatin immunoprecipitation assay Chromatin immunoprecipitation, assays were performed as described previously (5). Human MCF-7 cells were grown to 60 to 80% confluence and were then cross-linked with 1% formaldehyde for 30 min at room temperature. After cell lysis and sonication, immunoprecipitation reactions were performed overnight at 4°C using chromatin from approximately 107 cells per reaction mixture and 1 pg of each antibody: anti-SNAP43 (CS48), anti-p53 (21-25) (Ab-6; Oncogene), anti-acetyl-p53 (320) (Upstate), anti- HDACl (Santa Cruz), anti-HDAC2 (Santa Cruz), or pre-immune IgG (Sigma) antibodies. Anti-Sumo-l antibodies were purchased from Zymed. Recovered chromatin was suspended in 50 pL of H20, and PCR analysis was performed using 5 pL of immunoprecipitated chromatin or input chromatin. The primers used for amplification of each gene were described previously (3). PCR products were separated by 2% agarose electrophoresis in Tris-borate-EDTA buffer and were stained with ethidium bromide. Immunoprecipitations from in vitro transcription reactions In vitro transcription assay mixtures containing U1 or U6 promoter plasmids and equal molar amounts of pUC119 were performed as described previously (5) in the absence or presence of full-length wild type p53 or GST. 5 pL of each transcription reaction mixture was diluted to 500 pL and was cross-linked in 1% formaldehyde for 10 min at room temperature, quenched with 125 mM glycine for 10 min at room temperature, and immunoprecipitated with anti-TBP (SL2), pre-immune IgG (Sigma), anti-p53 (Ab-6; Oncogene), anti-RNA polymerase III (TB2) and anti-HDACI (Santa Cruz) or anti- HDAC2 (Santa Cruz) antibodies. Recovered plasmid DNA was analyzed by PCR using 164 primers specific to the U1 and U6 promoter regions or to pUC119 as a negative control. Immunoprecipitation experiments Whole cell extracts from pCDNA-HA-Sumo-l, -2, and -3 cotransfected MCF—7 cell harvested 8 hours after UV light treatment were diluted to 1 mL using HEMGT-ISO buffer containing protease inhibitors and incubated with 1 pg of antibodies directed against goat IgG (Sigma), full length p53 (Upstate) or anti-acetyl-p53 (320) (Upstate) 90 minutes at room temperature. Stable complexes were then affinity purified by incubation with Protein-G Fast Flow sepharose beads (Upstate Biotechnology) for 90 minutes at room temperature. Beads were washed in HEMGT-l 50 buffer and boiled for 5 minutes in Laemmli Buffer. Bound proteins were separated by 12.5% SDS-PAGE and transferred to nitrocellulose. Western blot analyses of recovered proteins were performed using anti- p53 (21-25) (Ab-6; Oncogene) and anti-HA antibodies. 165 References 10. 11. Barlev, N. A., L. Liu, N. H. Chehab, K. Mansfield, K. G. Harris, T. D. Halazonetis, and S. L. Berger. 2001. Acetylation of p53 activates transcription through recruitment of coactivators/histone acetyltransferases. Mol Cell 8:1243- S4. Gostissa, M., A. Hengstermann, V. Fogal, P. Sandy, S. E. Schwarz, M. Scheffner, and G. Del Sal. 1999. Activation of p53 by conjugation to the ubiquitin-like protein SUMO-1. Embo J 18:6462-71. Gridasova, A. A., and R. W. Henry. 2005. The p53 tumor suppressor protein represses human snRNA gene transcription by RNA polymerases II and III independently of sequence-specific DNA binding. Mol Cell Biol 25:3247-60. Henry, R. W., V. Mittal, B. Ma, R. Kobayashi, and N. Hernandez. 1998. SNAP19 mediates the assembly of a functional core promoter complex (SNAPC) shared by RNA polymerases II and III. Genes Dev 12:2664-72. Hirsch, H. A., G. W. Jawdekar, K. A. Lee, L. .Gu, and R. W. Henry. 2004. Distinct mechanisms for repression of RNA polymerase III transcription by the retinoblastoma tumor suppressor protein. Mol Cell Biol 24:5989-99. Ho, J., and S. Benchimol. 2003. Transcriptional repression mediated by the p53 tumour suppressor. Cell Death Differ 10:404-8. Juan, L. J., W. J. Shia, M. H. Chen, W. M. Yang, E. Seto, Y. S. Lin, and C. W. Wu. 2000. Histone deacetylases specifically down-regulate p53-dependent gene activation. J Biol Chem 275:20436-43. Murphy, M., J. Ahn, K. K. Walker, W. H. Hoffman, R. M. Evans, A. J. Levine, and D. L. George. 1999. Transcriptional repression by wild-type p53 utilizes histone deacetylases, mediated by interaction with mSin3a. Genes Dev 13:2490-501. Rodriguez, M. S., J. M. Desterro, S. Lain, C. A. Midgley, D. P. Lane, and R. T. Hay. 1999. SUMO-1 modification activates the transcriptional response of p53. Embo J 18:6455-61. Schmidt, D., and S. Muller. 2002. Members of the PIAS family act as SUMO ligases for c-Jun and p53 and repress p53 activity. Proc Natl Acad Sci U S A 99:2872-7. Thiagalingam, S., K. H. Cheng, H. J. Lee, N. Mineva, A. Thiagalingam, and J. F. Ponte. 2003. Histone deacetylases: unique players in shaping the epigenetic histone code. Ann N Y Acad Sci 983284-100. 166 12. Yang, S. H., and A. D. Sharrocks. 2004. SUMO promotes HDAC-mediated transcriptional repression. Mol Cell 13:611-7. 167 |"Eililljjilflljilliifli11111