I LIBRARY Michigan State University This is to certify that the dissertation entitled CHARACTERIZATION OF FOUR MEMBERS OF THE FERROUS ION AND a-KETOGLUTARATE DEPENDENT HYDROXYLASE FAMILY FROM TRYPANOSOMA BRUCE]: TWO THYMINE HYDROXYLASE-LIKE PROTEINS, J- BINDING PROTEIN 1, AND AN ALKB HOMOLOG presented by JANA M SIMMONS has been accepted towards fulfillment of the requirements for the Ph.D. degree in Biochemistry and Molecular Bioloqu Major Professor’s Siénature AW/ 7) 90/0 Date MSU is an Affirmative Action/Equal Opportunity Employer 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 5/08 KLIProj/AccsnPres/CIRCIDaIeDue.indd CHARACTERIZATION OF FOUR MEMBERS OF THE FERROUS ION AND a- KETOGLUTARATE DEPENDENT HYDROXYLASE FAMILY FROM TRYPANOSOMA BRUCE]: TWO THYMINE HYDROXYLASE-LIKE PROTEINS, J— BINDING PROTEIN 1, AND AN ALKB HOMOLOG By Jana M Simmons A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Biochemistry and Molecular Biology 2010 ABSTRACT CHARACTERIZATION OF FOUR MEMBERS OF THE FERROUS ION AND a- KETOGLUTARATE DEPENDENT HYDROXYLASE FAMILY FROM TRYPANOSOMA BRUCE]: TWO THYMINE HYDROXYLASE—LIKE PROTEINS, J- BINDING PROTEIN 1, AND AN ALKB HOMOLOG By Jana M Simmons Trypanosomes are eukaryotic parasites and the causative agents of several mammalian diseases. They are phylogenetically divergent making study of their biology pivotal to understanding their relationships to both ‘lower’ and ‘higher’ organisms. Trypanosomes contain many open reading fi'ames predicted to encode proteins of the Fen/(x-ketoglutarate (aKG) hydroxylase family including two thymine 7-hydroxylase-like proteins (TLPS & TLP7), J-binding protein 1 (JBPI), and an AlkB homolog (TbABH). TLPS & TLP7 transcript levels are 4 and 2.5 fold higher in the bloodstream form over the procyclic form parasites, indicating a role for the respective proteins in the former life stage. Protein production was verified in both life stages by western blot with a polyclonal anti-TLPS antibody and suggested that these proteins may undergo some posttranslational modification in vivo. Recombinant TLPS & TLP7 were confirmed to be members of the Fen/qKG hydroxylase family based upon formation of a characteristic metal—to—ligand charge-transfer (MLCT) chromophore when Feu and 0tKG were added to anaerobic samples. A homology model of TLPS was created and a library of small molecules docked with this model, indicating a propensity of the protein to bind heterocyclic molecules resembling nucleotides. Unfortunately, no DNA binding or enzymatic activity was observed for these proteins, leaving their cellular function unknown. JBPI is known to be involved in the hypermodification of thymine in nuclear DNA resulting in the formation of B-D-glucosylhydroxymethyldeoxyuracil, or base I. The first step of base I synthesis involves hydroxylation of the C5 methyl group of thymidine, a reaction type known to be catalyzed by the Fen/OKG hydroxylases in other organisms. Recombinant JBPI was confirmed to be a member of this enzyme family based on the characteristic MLCT chromophore formation. Unfortunately, no enzymatic turnover was observed by the in vitro methods employed leaving the direct detection of activity elusive. TbABH is a homolog of AlkB from Escherichia coli, an enzyme that oxidatively repairs alkylated DNA, consistent with such a mechanism of DNA repair existing in trypanosomatids. Sequence analysis suggested that TbABH is a nuclear, DNA-binding protein, and electrophoretic mobility shift assays using recombinant TbABH demonstrated preferential binding to alkylated double-stranded DNA. Membership in the Fen/0K6 hydroxylase family was verified by MLCT chromophore formation, however, attempts to detect in vi tro activity were unsuccessful. TbABH was shown to be capable of complementing an alkB mutant of E. coli subjected to alkylation stress. The complemented cell line was approximately 2-fold more resistant to alkylation-induced growth inhibition than the mutant, confirming the assignment of TbABH as a functional AlkB-like DNA repair protein in T. brucei. To my husband and children, with respect, gratitude, and love iv ACKNOWLEDGEMENTS I am grateful to the many people who have been a part of my life and a part of my graduate journey in the last six years. First, my utmost thanks go to my advisor, Dr Robert Hausinger, for his ever-present and immensely helpful guidance, encouragement, inspiration, and support. I also want to thank the members of my committee: Dr. Michael Feig, Dr. Charlie Hoogstraten, Dr. Donna Koslowsky, and Dr. Leslie Kuhn. Special thanks goes to Dr. Koslowsky for her assistance with several of the studies presented in this thesis including the epitope tagging of TLPS, the DNA binding studies with TbABH, and the qRT-PCR. I would also like to thank Dr. Kuhn for her help in optimizing and validating the homology model of TLPS. I have had the pleasure and privilege to work with many wonderful people in the lab over the years including Dr. Scott Mulrooney, Dr. Tina Miiller, Dr. Piotr Gryzska, Dr. Lee Macomber, Dr. William Kittleman, Dr. Hajime Masukawa, Dr. Soledad Quiroz, Dr. Meng Li, Dr. Efthalia Kalliri, Dr. Jong Kyong Kim, Mukta Rohankhedkar, Eric Carter, Jodi Boer, Nicholas Flugga, Saundra Hempel, Greg Moyerbrailean, Mike Howard, Megan Andrzejak, Brittnie DeVries, Scott Taber, Chris Towns, Rachel Morr, Andrea Silva, Bruce Fraser, Kim Anderson, Melody Campbell, Scott Elsey, and Matt Fountain. For all of their camaraderie, technical help, and support, I am grateful. I want to especially thank Piotr for all his help with the spectroscopy studies, Greg for his help maintaining the parasite cultures, William for his help with the mass spectrometry studies, Megan for help with the EMSAs using abasic site-containing DNA, and Mike for performing the docking studies with TLPS. I also need to thank Dr. Henri van Luenen of the Borst lab in the Netherlands for providing JBPI protein and collaborating on those studies. I would like to thank Dr. Joe Beckmann for encouraging and motivating me and giving me excellent training at Alma College before I began my Ph.D. studies. I cannot adequately express my love and gratitude toward my family for their support and encouragement. To my husband, Aaron, I could have never done this without you. Finally, I thank God for granting me the ability to undertake such a challenging journey, and the fortitude to complete it. vi TABLE OF CONTENTS LIST OF FIGURES ...................................................................................... - ............. i x ABBREVIATIONS ................................................................................................... xi CHAPTER 1 Introduction ................................................................................................................ l 1.1 Overview of trypanosome biology .................................................................. 2 1.1.] T brucei life cycle .................................................................................. 3 1.1.2 Antigenic variation .................................................................................. 5 1.1.3 General features of kinetoplastids ........................................................... 5 1.1.4 T. brucei replication, chromosome arrangements, and transcription ...... 6 1.1.5 Subtelomeric expression of VSG genes .................................................. 11 1.2 F e"/aKG-dependent hydroxylases .................................................................. 14 1.2.1 Mechanism of Fe I/oiKG hydroxylases .................................................... 14 1.2.2 Hydroxylases in nucleic acid and chromatin metabolism ....................... 18 1.3 Organization of thesis and open questions ...................................................... 20 References .................................................................................................................. 22 CHAPTER 2 Trypanosoma brucei brucei: Thymine 7-hydroxylase-like proteins ......................... 28 Abstract ...................................................................................................................... 29 2.1 Introduction ..................................................................................................... 30 2.2 Materials and Methods .................................................................................... 32 2.2.1 Candidate gene identification ................................................................. 32 2.2.2 Parasites and quantitative real-time PCR ................................................ 32 2.2.3 Construction of TLPS- and TLP7—expression Escherichia coli strains ..33 2.2.4 Over-expression and purification of TLP5 and TLP7 ............................ 34 2.2.5 Antibody production and Western blotting ............................................. 35 2.2.6 Spectroscopy ........................................................................................... 36 2.2.7 Enzyme assays ........................................................................................ 36 2.3 Results and Discussion ................................................................................... 38 References .................................................................................................................. 47 CHAPTER 2 APPENDIX 2A.1 Introduction .................................................................................................. 51 2A.2 Oligomeric state determination .................................................................... 52 . 2A.3 Homology model .......................................................................................... 54 2A.4 Relocation of purification tag ...................................................................... 58 2A.S C-terminal truncation ................................................................................... 59 2A.6 DNA binding ................................................................................................ 60 2A.7 In vivo epitope tagging of the TLP5 gene .................................................... 61 2A.8 Pull-down assays with TLP5 and trypanosome cell extracts ....................... 66 vii 2A.9 Conclusions .................................................................................................. 67 References .................................................................................................................. 68 CHAPTER 3 Search for hydroxylase activity in the trypanosomal protein, JBPI .......................... 69 3.1 Introduction ..................................................................................................... 70 3.2 Materials and Methods .................................................................................... 74 3.2.1 Determination of succinate production as a measure of uncoupled enzymatic turnover .................................................................................. 74 3.2.2 Oxygen consumption as a measure of uncoupled enzymatic tumover...76 3.2.3 Spectroscopic detection of a metal-to-ligand charge-transfer feature ..................................................................................................... 76 3.3 Results and Discussion ................................................................................... 78 References .................................................................................................................. 81 CHAPTER 4 Characterization of a T typanosoma brucei AlkB homolog capable of repairing alkylated DNA ........................................................................................................... 83 Abstract ...................................................................................................................... 84 4.1 Introduction ..................................................................................................... 85 4.2 Materials and Methods .................................................................................... 91 4.2.1 Gene identification and multiple sequence alignment ............................ 91 4.2.2 Cloning .................................................................................................... 91 4.2.3 Protein production and purification ........................................................ 92 4.2.4 Gel filtration chromatography ................................................................. 93 4.2.5 Spectroscopy ........................................................................................... 93 4. 2. 6 Electrophoretic mobility shift assays ...................................................... 94 4. 2. 7 In vitro enzyme assays ............................................................................ 97 4.2.8 Complementation assays ......................................................................... 98 4.3 Results and Discussion ................................................................................... 101 4.3.] Analysis of the TbABH sequence ............................................................ 101 4.3.2 General biochemical properties of TbABH ............................................ 103 4.3.3 DNA binding by TbABH ........................................................................ 105 4.3.4 Examination of DNA repair by TbABH ................................................. 108 4.3.5 Conclusions ............................................................................................. 112 References .................................................................................................................. 113 CHAPTER 5 Perspectives ................................................................................................................ 1 18 5.1 Introduction ..................................................................................................... 119 5.2 Summary of Results ........................................................................................ 120 5.2.1 TLP5 and TLP7 are unlikely to participate in pyrimidine salvage ......... 120 5.2.2 JBPl is inactive in its recombinant form ................................................ 121 5.2.3 TbABH is a DNA-binding protein homologous to E. coli AlkB ............ 122 5.3 Conclusions and Future Directions ................................................................. 124 References .................................................................................................................. 127 viii LIST OF FIGURES (Images in this dissertation are presented in color) CHAPTER 1 1.1 Anatomy of T Iypanosoma brucez' .................................................................. 8 1.2 Schematic of discontinuous transcription and the production of mature mRNA by trans splicing ................................................................................. 10 1.3 Bloodstream form subtelomeric expression site organization ........................ 13 1.4 General mechanism of F e" and aKG dependent hydroxylases ....................... 16 1.5 Reaction schemes for nucleic acid modifying hydroxylases .......................... 19 CHAPTER 2 2.] Sequence comparison of TLP5, TLP7, and T7H from R. glutinis .................. 39 2.2 Quantitative real-time PCR analysis of TLP5 and TLP7 expression in BF and PF stages of T. brucez' ............................................................................... 40 2.3 Electrophoretic analysis of purified TLP5, TLP7, and extracts of PF and BF cells ........................................................................................................... 43 2.4 Spectroscopic evidence for binding of Fe" and aKG by TLP5 ...................... 45 CHAPTER 2 APPENDIX 2A.1 Native size and Oligomeric state determination of TLP5 ............................. 53 2A.2 Homology model of TLP5 ........................................................................... 56 2A.3 Results of docking a subset of the ZINC small molecule database into the modeled active site of TLP5 ........................................................................ 57 2A4 Hemagglutinin epitope tagging of TLP5 ..................................................... 65 CHAPTER 3 ' 3.1 Synthesis of base I .......................................................................................... 72 3.2 uKG and succinate detection by HPLC .......................................................... 75 ix 3.3 Spectroscopic evidence for binding of F e" and aKG by JBPl ....................... 80 CHAPTER 4 4.1 Reaction ofAlkB with lmeA and 3meC in DNA or RNA ............................. 87 4.2 Structure of AlkB (PDB 2fd8) ........................................................................ 88 4.3 Multiple sequence alignment of selected bacterial and eukaryotic AlkB-like proteins ............................................................................................................ 102 4.4 Native size and Oligomeric state determination of TbABH ............................ 104 4.5 Spectroscopic evidence for binding of Fe" and aKG by TbABH .................. 106 4.6 EMSA studies of TbABH and various DNA substrates ................................. 107 4.7 Time course viability test with wild-type and AalkB BW251 13 cell lines stressed with 0.5% MMS ................................................................................ 110 4.8 Complementation of an E. coli alkB mutant with TbABH under alkylation stress ................................................................................................................ 111 1 meA 3meC aKG ABHl ABH2 ABH3 ApoLl ATP Base J BC IP/N BT BF CF U Cth3 dNTP ds EDTA EMSA ESAG GC/MS HA HPLC ABBREVIATIONS l —methy1-adenine 3-methyl-cytocine alpha-ketoglutarate AlkB homolog 1 AlkB homolog 2 AlkB homolog 3 apolipoprotein Ll adenosine triphosphate [3-D-glucosylhydroxyrnethyldeoxyuraciI 5-bromo-4-chloro-3-indolyl phosphate/nitro blue tetrazolium chloride bloodstream form colony forming unit cytochrome c3 deoxy-nucleotide triphosphate double stranded ethylenediamine tetraacetic acid electrophoretic mobility shift assay expression site associated gene gas chromatography/mass spectrometry hemagglutinin high performance liquid chromatography xi HOMedU IPTG JBPl JPBZ ijC kDa LB MLCT AMMS NAD*an NCBI NLNTA OPDA ORF PCR PDB PF PVDF RNAP RNAi RT SDS-PAGE hydroxymethyldeoxyuracil isopropyl thiogalactopyranoside see base I J-binding protein 1 JBPl related protein Jumonji C kilodalton lysogeny broth metal-to-ligand charge-transfer methylmethane sulfonate nicotine adenine dinucleotide (oxidized or reduced) National Center for Biotechnology Information nickel-bound nitrilotriacetic acid ortho-phenylenediamine open reading frame polymerase chain reaction protein data bank procyclic form polyvinylidene fluoride RNA polymerase RNA interference reverse transcribed sodium dodecyl sulfate - polyacrylamide gel electrophoresis xii SL RNA splice leader RNA 33 single stranded SWI-2/SNF-2 switch/sucrose nonfermentable TauD taurine/aKG dioxygenase TbABH T rypanosoma brucei AlkB homolog T7H thymine 7-hydroxylase TLP5 thymine 7-hydroxylase-like protein (from chromosome 5) TLP5AC13 13 residue C-terminal truncation of TLP5 TLP7 thymine 7-hydroxylase-like protein (from chromosome 7) UTR untranslated region VSG variable surface glycoprotein xiii CHAPTER 1 INTRODUCTION This dissertation focuses on trypanosomal enzymes of the non-heme iron (Fen)- and a—ketoglutarate (aKG)—dependent hydroxylase family, with particular emphasis on proteins that act on nucleic acids and those catalyzing the modification or repair of DNA. To provide a framework for understanding these studies, I divide my introduction into two major sections; first I discuss trypanosomes and their unique biology including key features of gene expression, and then I describe the Fell/(1K6 hydroxylase family emphasizing its members involved in nucleic acid modification. 1.1. Overview of T rypanosome Biology Trypanosomes are eukaryotic unicellular parasites that are the causative agents of several diseases affecting mammals including Human Afiican Trypanosomiasis (commonly called Sleeping Sickness), Chagas disease in humans, and Nagana in cattle (1). These diseases affect millions of people each year and pose a heavy economic burden to the endemic countries due to the loss of livestock. The World Health Organization estimates near 100,000 new cases of Sleeping Sickness each year with only a fraction of those being reported or treated since the most affected populations are mainly rural and among the poorest in Africa. Illustrative of the severity of the problem, the BBC World News produced a documentary aired on October 11, 2008 calling Sleeping Sickness “The Deadliest Disease” on Earth (2) (http://www.survival.tv/documentaries/sleeping_sickness.php). Given the prevalence of trypanosomal diseases, there is great interest in the development of drugs to combat infection. Still, treatments are sorely limited and continued active research is needed to better understand these organisms and to develop improved methods of control. For example, detailed biochemical analyses of enzymes specific to trypanosomes could suggest targets for future drug development. Trypanosoma sp. are members of the order kinetoplastidae which also include organisms of the genera Leishmania, responsible for additional vertebrate diseases (3), and Crithz'dia, responsible for various invertebrate diseases (4). The Kinetoplastids are a phylogenetically distinct group that diverged early from the central eukaryotic domain before the branching of fungi, plants, and animals (5,6). Therefore, study of their physiology and biological processes provide useful insight not only for the purpose of disease treatment and prevention, but also for understanding the development of this ancient branch of life. Because my work involves Trypanosoma brucei, the following ' sections focus on this microorganism. 1.1.1. T. brucei Life Cycle T. brucei has a complex multi-phase life cycle including a morphologically distinct proliferative stage in an insect vector (Tsetse fly) and another in the targeted mammalian host. The insect stage or procyclic form (PF) of the parasite begins in the gut of the fly when the insect takes a blood meal from an infected mammal. Most parasites are cleared from mature insects and very few actually end up transmitting the trypanosomes to a new host (7). Surviving parasites migrate to the salivary glands of the fly over the course of several days where they transform into a non-proliferative metacyclic form awaiting transfer to a new mammalian host. When the T. brucei-carrying fly bites another mammal, the trypanosomes are introduced into the bloodstream and another major life stage of the parasite begins. The bloodstream form (BF) is the stage where human disease is manifest. There are several forms of disease caused by the strains of T. brucei. T brucei brucei is the strain that infects livestock since humans have an innate immunity to this particular parasite due to apolipoprotein L1 (ApoLl), also known as “trypanosome lytic factor” (8). T. brucei rhodesiense is most prevalent in eastern Africa and causes an acute form of the disease in humans that often is fatal after only a few weeks of infection (1). T. brucei gambiense is most prevalent in central and southern Africa and causes a chronic form of the disease where the human victim may go undiagnosed for weeks to months (1). In a chronic infection, the parasites eventually cross the blood-brain barrier and colonize the cerebrospinal fluid while also invading other internal organs. Personality changes, impaired mental functions, and severely disrupted circadian rhythms (hence the term Sleeping Sickness) are all symptoms of this second stage trypanosomiasis. If left untreated, the infection leads to coma and death (9). The ability of T. b. rhodesz'ense and T. b. gambiense to effectively colonize humans relates to a serum-resistance-associated gene (10,11). In particular, humans produce ApoLl which is taken up into the trypanosome cell as part of a high density lipoprotein complex with haptoglobin and hemoglobin (12,13). ApoLl associates with the cell’s lysosome, forming pores in the membrane and leading ultimately to cell lysis (8). T. b. rhodesz‘ense and T. b. gambiense produce the serum-resistance-associated protein from an active variable surface glycoprotein (VSG) expression site which binds to the C-terminus of ApoLl and neutralizes this effect. Recently, it was shown that minor mutations to the C-terrninal helix of ApoLl can ablate this resistance in T. b. rhodesiense making it sensitive to recombinant ApoLl in vitro and in transfected mice (14). 1.1.2. Antigenic Variation In order to maintain an effective colonization of the host, the strictly extracellular parasite must continually deal with the immune system responses to which it is exposed. BF T. brucei utilizes a process called antigenic variation that allows the trypanosome to evade the host immune system by periodically changing its expressed VSG coat glycoprotein in a coordinated fashion (10). The genetic arrangement of the VSG— encoding regions and process of expression will be discussed in section 1.1.5. In the PF trypanosomes the surface coat is dominated by proteins called procyclins (or procyclic acidic repetitive proteins). Unlike the BF protists, the PF cells can express several different procyclins at one time and these proteins remain invariant through the course of this life stage. There are two major forms of procyclins termed EP and GPEET. These designations refer to the single amino acid code for the residues found in the repeated regions of the glycoproteins (15). 1.1.3. General Features of Kinetoplastids T. brucei’s surface coatings in its two life stages are characteristic of this species, whereas other features of their biology are shared more broadly by the order kinetoplastidae. Namely, members of this order contain a single mitochondrion (Figure 1.1.6) containing a DNA compartment termed a kinetoplast (Figure 1.1.7) that has a unique genetic content made up of several maxicircles and several hundred concatenated minicircle DNAs. Through elaborate mechanisms beyond the scope of this introduction, these serve to generate the required mRN As to allow expression of mitochondrial proteins. In T. brucei, the function of the mitochondrion varies greatly throughout the life cycle. In PF parasites, the mitochondrion is large and active with a functional tricarboxcylic acid cycle responsible for generation of ATP. In contrast, nutrients are plentiful in the environment of the host bloodstream where the parasite’s mitochondrion is greatly reduced in size and quiescent. Glycolysis alone provides ATP to the protist and pyruvate is excreted from the cell (9). All kinetoplastids are characterized by a single flagellum that extends down the length of the parasite and provides for motility (Figure 1.1.2). The flagellar pocket includes a portal providing the sole means for import and export of cellular metabolites and proteins across the cell wall (Figure 1.1.4) (16). This exclusivity is due in part to the previously mentioned dense coating of glycoproteins covering the rest of the cell surface. In addition to the dense protein coat precluding access from the outside, the subpellicular microtubules are too closely spaced to allow transport vesicles access to the plasma membrane from the inside (Figure 1.1.3). T. brucei undergoes very high rates of non- specific, exclusively clathrin-dependent endocytosis through this pocket so that the surface proteins are constantly recycled. BF parasites also express several transferrin receptors in the flagellar pocket which are responsible for uptake of host ferritin and provide the iron needed by the cell (16). 1.1.4. T. brucei Replication, Chromosome Arrangements, and Transcription T. brucei replication exhibits several unique properties compared to more typical eukaryotes. They contain single c0pies of their Golgi complex (Figure 1.1.9) and lysosome (Figure 1.1.10), thus creating a challenge for replication as each of these organelles needs to be copied and dutifully partitioned to the daughter cells (1 7). The nuclear genetic arrangement in these cells is very streamlined, and the kinetoplast nearly lacks intronic regions and possesses some overlapping genes. T. brucei contains 11 linear megabase-sized chromosomes containing the vast majority of the protein-coding genes and gene arrays, several intermediate chromosomes that contain repetitive elements and occasionally a VSG expression site, and up to hundreds of mini-chromosomes containing mostly pseudogenes and repertoires of VSG-encoding genes amongst highly repetitive sequences (18,19). The latter regions are believed to provide a repertoire of nearly inexhaustible options for recombination to create new varieties of VSGs. Trypanosomes are diploid for their large chromosomes. These contain most, if not all, protein-encoding genes except for some VSG-encoding genes. The exact sizes of the chromosomes vary greatly between trypanosome populations due to large repetitive elements and tandem gene arrays, which give rise to high rates of recombination and unequal crossing over events. Mini-chromosomes also vary greatly in size as they are made up of typical eukaryotic telomeres linked by large tracts of a 177-bp repeat with usually one VSG-encoding gene or pseudogene. The ploidy of the mini-chromosomes is less certain due to a lack of distinct chromosomal markers (18). Figure 1.1: Anatomy of T rypanosoma brucei. l. nucleus 2. flagellum 3. subpelicular microtubules 4. flagellar pocket 5. endoplasmic reticulum 6. mitochondrion 7. kinetoplast 8. basal body 9. Golgi body 10. lysosome. Figure adapted with permission from Dr. Markus Engstler. One thing markedly lacking from the nuclear genome of T. brucei and other trypanosomatids is the presence of typical RNA polymerase (RNAP) II promoters. Conventional eukaryotic protein-coding gene transcription involves the formation of initiation complexes that congregate at an RNAP 11 promoter with many genetic regulatory elements affecting the transcription initiation of each open reading frame (ORF) (20). In contrast, trypanosomes have long poly—cistronic genetic arrangements on each chromosome with a strand switch region somewhere in the middle from which transcription initiates and radiates outwards toward the telomeres, with one set of ORFs on one strand and another set on the other (21). This leaves only the 5’ and 3’ untranslated regions between the ORFs to provide regulation, almost all of which occurs post—transcriptionally (22). Therefore, gene products needed in abundance, such as tubulin, are arranged in repetitive gene arrays in order to obtain greater amounts of the mRNA. That mRNA is then stabilized to allow for greater levels of translation. Due to this unique mechanism for generation of transcripts, trypanosomes process pre-mRNAs by trans-splicing. As depicted in Figure 1.2, a 39—nucleotide spliced leader sequence (SL-RNA) is transcribed from the RNAP II promoter (using an RNAP that is altered in its subunit composition when compared to that involved in poly-cistron transcription) in great abundance (100-200 tandem copies per genome) and is trans-spliced onto the 5’ end of every mRN A produced in the nucleus. These are then polyadenylated based on the location in the intergenic region of the 5’ splice acceptor site (23). Figure 1.2 Schematic of discontinuous transcription and the production of mature mRNA by trans splicing. Transcription of short discrete tandemly repeated genes (SL RNA) and polycistronic protein-coding regions (exemplified by L. major chromosome 1 (24)) by RNAP H is indicated at the left. The primary transcripts from the two classes of gene are shown in the center intersecting with steps in the trans splicing pathway (right panel). The bimolecular trans splicing reaction resembles the unimolecular cis splicing pathway in many aspects, including the initial cleavage of the SL RNA splice donor site, the formation of 5’—2' phosphodiester bond on a branch—point A residue, cleavage at the pre- mRNA splice acceptor site and ligation of the two exons. Differences between the two processes include the formation of a Y-branched intermediate (rather that a lariat-like intermediate) and the coupled polyadenylation of the upstream mRNA. Figure adapted with permission from (23). SPLICED LEADER GENE ARRAY , SLf RNA Q) _ m7G[-—m7rl monomsrnomc ._~ - TRANS-SPLICING TRANSCRIPTION ‘5' AT Two Exrraames PoLYCISTReuie' G_AM m7cl ‘ » H POLYADENYLATIO LEISHMANIA FRIEDLIN P°'-Y°'5TR°N TRANSLATION CHROMOSOME 1 or GENE *x' mRNA v 10 1.1.5. Subtelomeric Expression of VSG genes Another unique genetic feature of T. brucei relates to its expression of genes in subtelomeric regions of the megabase chromosomes. These regions, spanning up to 2 kb upstream of the telomeric repeats, include the VSG expression sites (Fig. 1.3). Trypanosomes contain approximately 20 of these sites which are made up of various repetitive DNA satellites, the VSG gene, and several other expression site-associated genes (ESAGs). These genes encode variations of transferrin receptors (ESAG 6 and 7) for iron sequestration from the host, a homolog of a biopterin transporter (ESAG 10), several proteins likely involved in the differentiation between life stages (ESAG 4 and 8), other protein-encoding genes of unknown function, and the serum-resistance—associated protein responsible for the ability of T. b. rhodesiense and T. b. gambiense to effectively colonize humans (10,1 1,25). During trypanosome infection, only one VSG expression site is active at any given time, allowing the population of trypanosomes to express a single coat protein. Transcription of the VSG gene is a—amanitin insensitive (26), implicating the involvement of RNAP I which does not typically transcribe protein-encoding genes. RNAP I was shown to be directly responsible for transcription of the VSG and procyclin surface protein-encoding genes of trypanosomes (27). Although each VSG expression site has its own RNAP I promoter and seems to initiate transcription constitutively (28,29), transcription aborts before reaching the VSG gene in silent expression sites (30). The silencing mechanisms employed by these parasites to accomplish such coordinated gene expression have been studied for many years, though they remain ll largely a mystery. Positioning effects mediated by the subtelomeric localization have been investigated with some contradictory results, but it does seem that there is some promoter restraint, especially in BF trypanosomes (31,32). In addition, a modified base found in telomeric and subtelomeric regions has also been implicated in gene silencing. B-D—glucosylhydroxymethyluridine, known as base I (33), replaces up to 1% of the thymidine in trypanosomal DNA (34). This novel base is localized to telomeric repeats, subtelomeric repeats surrounding the VSG expression sites, and sequences within VSG expression sites not being expressed (Fig. 1.3). However, the one active VSG expression site is void of the modification (35). Further, the active VSG expression site is sequestered in the “expression site body”, an area of the nucleus that is separate from the nucleolus; this subnuclear localization may also play a role in its discriminatory expression (36). Trypanosomes can switch the expression of their VSG by several mechanisms. The most common mode of switching is by keeping the same active VSG expression site and swapping out the VSG gene with another by homologous recombination. Alternatively, trypanosomes can splice together new VSGs by recombining part of the currently expressed VSG with a pseudo gene from a minichromosome or intermediate chromosome. Another mechanism is to silence the current VSG expression site while activating another. This approach may be less common as it would require more chromatin rearrangement, though it would also allow the expression of variant transferrin receptors and other ESAGs. 12 Figure 1.3 Bloodstream form subtelomeric expression site organization Figure adapted from (25). ESAGs are depicted by numbered open boxes (those genes present in only some expression sites are shown in parentheses). Relative location of base I is shown above the expression site diagrams and is based on immunoprecipitation of fragmented DNA with an anti-J antibody (3 5). Filled triangles represent 50 bp repeats, hatched boxes represent 70 bp repeats, and filled ovals represent telomeric repeats. The top diagram depicts a silent expression site with base J found throughout the coding sequences while the center diagram depicts an active expression site, devoid of base I and fully . transcribed (dashed arrow). Finally, the bottom diagram depicts a specialized expression site of T. b. rhodesiense containing the serum resistance associated gene. J J I J JJJ JJJJJJJJJJJJ 8 3 2 11 a uummm- J JJ _____,__-_____ ___________________ JJJJJJJ a 3 11 1 9 vse J JJ JJJJJJJJJJ l. B 5 111111111 Mlllllllll l3 1.2. F ell/aK G-Dependent Hydroxylases. The F eu/aKG-dependent hydroxylases use a mononuclear ferrous ion to catalyze the oxidative decarboxylation of aKG while activating molecular oxygen in order to hydroxylate a target substrate. This broad enzyme superfamily has representatives throughout the domains of bacteria and eukaryota, with other members found in viruses, and they catalyze a wide variety of reactions (3 7),(3 8). These enzymes are characterized by a double-stranded fl-helix core fold, often termed a B—jellyroll structure that is made up of 8 [3 strands within which lies the active site (39). Section 1.2.1 will discuss the overall reaction mechanism and Section 1.2.2 will focus on those members whose primary function is the modification of nucleic acids or chromatin. 1.2.1. Mechanism of F e1 1/ aK G hydroxylases. The current view of the enzyme mechanism for these enzymes, summarized in Figure 1.4, is a modification of that first proposed over 25 years ago (40). Two histidines and one carboxylic acid residue (usually an aspartate) coordinate one face of the iron atom (41). This “facial triad” usually occurs in a His-X—Asp/Glu-Xn-His motif where n is typically around 50 residues, with the remaining 3 coordination sites on the metal atom taken up by water molecules in the resting state (Fig. 1.4 panel A). Upon addition of the cosubstrate, uKG, two waters are displaced by the bidentate binding of the aKG molecule via its C1 carboxylate and C2 keto groups (Fig. 1.4 panel B). The C5 carboxylate of aKG often is stabilized by a conserved arginine located approximately a dozen residues C-terminal to the second His residue of the facial triad (38). This enzyme state leaves 14 one iron coordination site occupied by a water molecule that is displaced by the binding of the primary substrate (Fig. 1.4 panel C). The two aKG-bound states can be visualized spectroscopically. Thus, in the absence of primary substrate and oxygen, but the presence of iron and aKG, these enzymes exhibit a metal-to-ligand charge-transfer electronic transition that is observed at around 500 to 530 nm (42). For example, taurine/aKG dioxygenase (TauD), a well- studied member responsible for the oxidative cleavage of taurine to release sulfite during sulfur starvation in Escherichia coli, forms this characteristic chromophore at 530 nm with an extinction coefficient of 140 M'1 cm'1 (42). This feature can be used to diagnose proper protein folding and cofactor/cosubstrate binding. It is also used to confirm membership in this enzyme family for proteins without a verified function or a known primary substrate. Once the primary substrate binds to the protein, near the active-site but not coordinating the iron atom, this feature often shifis. In the case of TauD, taurine binding causes a 10 nm shift in the absorbance to higher energy and increases the intensity of the chromophore to give an extinction coefficient of 180 M'l cm'1 at 520 nm (42). Displacement of the final water molecule leaves an open coordination site on the iron atom that provides a site for molecular oxygen to coordinate the iron. This process likely creates two very short-lived species including an F em-superoxo species and a bridging Few-peroxo species that have never been observed (Fig. 1.4 panels D and E). Decomposition of the latter species leads to cleavage of the cosubstrate into the products succinate and C02 as well as the formation of a highly reactive Few-0x0 species which 15 Figure 1.4 General mechanism of Fe" and uKG dependent hydroxylases. Primary substrate is generalized and abbreviated by R-H. R+I R H 'O o B H210 ' O’ o C ASP""-Fell/O O ASp""'Fe"\/ / 0 His’ H"\O/ H 0 His, lilrso _ IS - 2 " 03/1 COO “A 2 H20 R‘H A H2|O r ‘02 “D ASP"; Fell {H20 ASP'M em /0 O HIS i H20 HISI I \0/ HS l. His J - z R-OH COO K -0 + I 3 H20 R OHCOO— R-H G OH" O 'O—-O E ASp"--.. III Asp: iv’: 0 Fe\- Fe His/ I OJLL His’ |\'o HIS coo R‘H His F 0: COO Asp' ..... Fl l IV 0 . / e\ 16 was directly observed first for TauD (43 -45), and later for prolyl hydroxylase (46) and a related halogenase, Cth3 (47) (Fig. 1.4 panel F). The highly reactive F e‘V oxo species then abstracts a hydrogen atom from the primary substrate generating a likely Fem-hydroxo species and a substrate radical (Fig. 1.4 panel G). The traditional view is that the hydroxyl radical then rebounds with the carbon radical, thus hydroxylating the substrate (which spontaneously degrades into sulfite and arninoacetaldehyde in the case of taurine) and regenerating the ferrous ion at the active—site. Very recent studies suggest this step is more complex, involving an F em- oxo species and an alkoxo species (not depicted), ultimately yielding the same products (48). The products are then released from the active-site and are replaced by three waters to regenerate the resting state for the enzyme (Fig. 1.4 panel A). This reaction cycle can be tightly coupled, resulting in the utilization of stoichiometric amounts of primary substrate, aKG, and 02 for each turnover (3 8). However, in the absence of the primary substrate or the presence of alternate substrates or inhibitors, these enzymes can decarboxylate aKG and activate oxygen to generate reactive oxygen species that can pose a threat to the cell. Some of this damage seems to be avoided by “fail safe” mechanisms involving side chain self-hydroxylation (49,50) or reaction of the activated species with a cellular reductant rather than releasing deleterious oxygen species into the milieu of the cell (51). This “uncoupling” of oxygen activation and substrate hydroxylation can be observed by monitoring oxygen or aKG consumption and succinate production by various methods and has been a useful tool to aid in the characterization of proteins of unknown function. 17 1.2.2. Hydroxylases in Nucleic Acid and Chromatin Metabolism. Several members of the F eu/aKG hydroxylase enzyme family utilize this powerful oxidizing reaction to modify nucleic acids. One of the best studied enzymes in this subgroup is AlkB, an E. coli protein, that repairs alkyl lesions on DNA and RNA (52,53). This enzyme follows the same basic mechanism described above to oxidize methyl adducts on the N—l position of adenine (lmeA), the N-3 position of cytosine (3meC), and selected other lesions in the context of DNA (Fig. 1.5 panel A). The hydroxylated methyl groups are spontaneously released from the molecule as formaldehyde, regenerating the unmodified bases. Multiple complexes of AlkB along with its cofactor, cosubstrate, and primary substrates have been crystallized and show that single-stranded nucleic acids are preferred by this enzyme with the strand bent into the active site, thus flipping the alkylated base deep into the pocket where chemistry takes place (54). Further, this protein has a ‘nucleotide recognition lid’ that alternates between an open and closed state upon binding of the substrate, providing more stability for the transition state and facilitating the enclosed environment needed for the oxygenation reaction. AlkB has been demonstrated to function in the repair of both DNA and RNA, with methylated, ethylated, and 1,N6-etheno modified bases. The physiological relevance of reactions involving more bulky lesions is not clear given other pathways in the cell, such as base excision repair; however, the substrate versatility (plasticity of the active site) suggests that AlkB may be able to serve as a back up system of repair for these lesions (55). Another example of nucleic acid hydroxylation is found in a firngal enzyme, thymine 7-hydroxylase, which participates in pyrimidine salvage. This enzyme catalyzes 18 Figure 1.5 Reaction schemes for nucleic acid modifying hydroxylases. A. Predominant reactions catalyzed by E. coli AlkB. B. Reactions catalyzed by thymine 7-hydroxylase. HMU is 5-hydroxymethyluracil. EU is S-formyluracil. A NH2 NH2 NH? ”R Fe(11) d“ d“ . 1-meA AlkB * + Adenine NH2 / NH2 NH, + CH “KG CO + CHZOH FEN/ 3 O2 Succmate l \N/ HCHOI \N 1k /k N/K til 0 til 0 I 0 dB dR dFI 3-meC Cytosine B. O 0H 0 I NH ' NH N/Ko o co N/KO H al295% homogeneous (Fig. 2.3 panel A, lanes 2 and 3) and of the size predicted by the sequences (37.5 kDa when accounting for the His tag), with slight differences in electrophoretic mobility observed. The proteins behaved as monomers when examined by Blue Native gels (Invitrogen) or Superdex 75 gel filtration chromatography (data not shown). Both proteins precipitated when exchanged into a low salt buffer after purification, but exchange back into the column binding buffer provided enhanced stability. 41 Polyclonal anti-TLP5 antibody was used to perform Western blots of purified recombinant TLP5 and TLP7 along with cell extracts of both PF and BF trypanosomes (Fig. 2.3 panel B). The antibody detected both proteins, as expected from their close similarity. A major cross-reactive species in both BF and PF electrOphoresed more slowly than the TLP5/TLP7 doublet; this species may be an unrelated contaminant or it might arise from post-translational modification of these proteins (leading to an apparent ~10- kDa increase in size) in the trypanosome, but lacking in the E. coli expression system. Future work could include efforts to identify this band and determine what, if any, modifications are added to the protein in vivo. In contrast to the RT-PCR results that indicate higher transcript levels in the BF cells, the immunological analysis suggests that TLP5/7 is more abundant in PF than BF cells; these results are compatible with the protein(s) being turned over more rapidly in the BF life stage or being translated at a greater rate in the PF life stage. Of primary importance, these results indicate that both proteins appear to be present in both life stages. To confirm proper folding of TLP5 and TLP7, the purified proteins were examined for their ability to form a characteristic chromophore seen in other members of the Fen/aKG—dependent dioxygenases and associated with binding of the iron cofactor and the aKG cosubstrate (15). The proteins were monitored by UV-visible spectroscopy under anaerobic conditions, using low temperature to maintain stability, while titrating in orKG and F e". As shown in Figure 2.4, anaerobic TLP5 generated the diagnostic metal- to-ligand charge-transfer transition at 505 nm when both metal and cofactor were present. The extinction coefficient for this feature was approximately 40 M'l cm", indicating that the chromophore was incompletely formed compared with previously described 42 Figure 2.3. Electrophoretic analysis of purified TLP5, TLP7, and extracts of PF and BF cells. Samples were subjected to SDS-PAGE and examined by A. Coomassie staining or B. Western blot analysis with anti-TLP5 antibodies. Lanes: 1, molecular weight ladder; 2, TLP5; 3, TLP7; 4, extracts of PF cells; 5, extracts of BF cells. A B 97 -. .. 66 L I 45 e. a '1; -';‘I ...e ... . 31* -- . —— 21 14 43 intensities (15), but addition of more metal ions led to protein precipitation. TLP7 also exhibited the diagnostic transition (data not shown), confirming correct folding of the protein, but it was less stable than TLP5 and precipitation during the experiment precluded estimation of the extinction coefficient. Despite extensive efforts, no in vitro thymine hydroxylase activity was detected for purified recombinant TLP5 or TLP7 when assayed by using an oxygen electrode to monitor oxygen consumption, HPLC to assess succinate production, or GC-MS to measure thymine hydroxylation. We cannot exclude the possibility that a post- translational modification, not occurring in E. coli, is required for activity. It is also conceivable that the His-tag hinders activity in the recombinant proteins or that, despite formation of the Fell/aKG chromophore indicating appropriate folding of the active site domains, the full length proteins do not fold properly. On the other hand, these results are compatible with the proteins utilizing alternative substrates. Other members of the Fen/OLKG dioxygenase family recognize a wide array of substrates ranging from small molecules to proteins and DNA (16). Several additional small molecules were tested as putative substrates including other bases, nucleosides, and amino acids, but again no activity was detected. With regard to the possible use of DNA as a substrate, trypanosomes are known to contain the hyper-modified base J in their nuclear DNA (17,18), which requires hydroxylation of an unactivated methyl group of thymidine in DNA to create hydroxymethyl-deoxyuracil (19,20) that is subsequently glucosylated. Thus, we examined whether TLP5 or TLP7 reacted with polynucleotides or thymidine; however no such activity was observed. Furthermore, neither protein bound to linear DNA fragments under any conditions examined (incubation with or without 50 uM Fe", 44 Figure 2.4. Spectroscopic evidence for binding of F e" and aKG by TLP5. The anaerobic UV/visible spectrum of TLP5 (250 uM in binding buffer) was examined for the sample as isolated, after adding 1 mM aKG, and while titrating in FeII with stirring at 9 °C. The (F ell-aKG-protein minus aKG-protein) difference spectra shown correspond to the addition of 0.5, 1, 2, and 3 equivalents of metal ions. 0.015 — g A It: .9 h- 0 1n .n < 0.005 A < o . r 350 450 550 650 - 750 Wavelength (nm) 45 150 uM aKG, and 100 uM MgCl2 at 37 °C for 30 min) according to gel band shift studies. Thus, TLP5 and TLP7 are unlikely to possess DNA modifying activity, consistent with the recent proposals that J -binding protein 1 (JBPl ), identified on the basis of its ability to bind to J -containing chromatin, and JBP2, related to JBPI, are critically involved in base J synthesis (10,21-24). Extracts of PF and BF cells were directly assayed for thymine hydroxylase activity by using GC-MS. NO significant hydroxylase activity could be detected (i.e., less than 0.5% product formed from 250 uM thymine) after a 1 h incubation. In conclusion, we have identified two genes encoding T7H-like proteins in T. b. brucei, demonstrated that the genes are expressed at greater levels in the BF over the PF stage, shown that both proteins are present in each life stage (possibly with greater abundance in the PF life stage), and expressed the genes in E. coli. We purified the two recombinant proteins, demonstrated that they form an Fell/OLKG chromophore (indicating proper folding), and tested for catalytic activity and DNA binding. Our results provide no indication of the recombinant proteins being able to hydroxylate free thymine or thymidine within DNA or to bind DNA. Furthermore, no hydroxylase activity was detected in extracts from trypanosomes of either life stage. The roles of these proteins in T. b. brucei remain unknown, but our evidence provides strong arguments against their participation in pyrimidine salvage. 46 REFERENCES Barrett, MP, Burchmore, RJ, Stich, A, Lazzari, JO, Frasch, AC, Cazzulo, JJ, Krishna, S. 2003. The trypanosomiases. Lancet 362: 1469-1480 Berriman, M, Ghedin, E, Hertz-Fowler, C, Blandin, G, Renauld, H, Bartholomeu, DC, Lennard, NJ, Caler, E, Hamlin, NE, Haas, B, Bohme, U, Hannick, L, Aslett, MA, Shallom, J, Marcello, L, Hou, L, Wickstead, B, Alsmark, UCM, Arrowsmith, C, Atkin, RJ, Barron, AJ, Bringaud, F, Brooks, K, Carrington, M, Cherevach, I, Chillingworth, T-J, Churcher, C, Clark, LN, Carton, CH, Cronin, A, Moule, S, Martin, DMA, Morgan, GW, Mungall, K, Norbertczak, H, Ormond, D, Pai, G, Peacock, CS, Peterson, J, Quail, MA, Rabbinowitsch, E, Rajandream, M- A, Reitter, C, Salzberg, SL, Sanders, M, Schobel, S, Sharp, S, Simmonds, M, Simpson, AJ, Tallon, L, Turner, CMR, Tait, A, Tivey, AR, Van Aken, S, Walker, D, Wanless, D, Wang, S, White, B, White, 0, Whitehead, S, Woodward, J, Wortman, J, Adams, MD, Embley, TM, Gull, K, Ullu, E, Barry, JD, Fairlamb, AH, Opperdoes, F, Barrel], BG, Donelson, JE, Hall, N, Fraser, CM, Melville, SE, El-Sayed, NM. 2005 . The genome of the African trypanosome T rypanosoma brucei. Science 309: 416-422 Smiley, JA, Kundracik, M, Landfiied, DA, Barnes, VR, Sr., Axhemi, AA. 2005. Genes of the thymidine salvage pathway: thymine-7-hydroxylase from a Rhodatorula glutinis cDNA library and iso-orotate decarboxylase from Neurospora crassa. Biochim. Biophys. Acta 1723: 256-264 Neidigh, JW, Darwanto, A, Williams, AA, Wall, NR, Sowers, LC. 2009. Cloning and characterization othodotorula glutinus thymine hydroxylase. Chem. Res. T oxicol. 22: 885-893 Holme, E, Lindstedt, G, Lindstedt, S, Tofft, M. 1970. 7-Hydroxylation of thymine in a Neurospora strain coupled to oxidative decarboxylation of 2-ketoglutarate. Biochim. Biophys. Acta 212: 50-57 Shaffer, PM, Arst, HN. 1984. Regulation of pyrimidine salvage-in Aspergillus . nidulans: a role for the major regulatory gene areA medratrng nitrogen metabolite repression. Mol. Gen. Genet. 198: 139-145 Gudin, S, Quashie, NB, Candlish, D, Al-Salabi, MI, Jarvis,'SM, Ranford- Cartwright, LC, de Koning, HP. 2006. T rypanosoma brucei: A survey of pyrimidine transport activities. Exp. Parasitol. 114: 118-125 47 10. ll. 12. l3. 14. 15. l6. 17. 18. van den Born, E, Omelchenko, MV, Bekkelund, A, Leihne, V, Koonin, EV, Dolja, W, F alnes, P0. 2008. Viral AlkB proteins repair RNA damage by oxidative demethylation. Nuc. Acids Res. 36: 5451-5456 Cliffe, LJ, Kieft, R, Southern, T, Birkeland, SR, Marshall, M, Sweeney, K, Sabatini, R. 2009. JBPl and JBP2 are two distinct thymidine hydroxylases involved in J biosynthesis in genomic DNA of Afiican trypanosomes. Nuc. Acids Res. 37: 1452-1462 Iyer, LM, Tahiliani, M, Rao, A, Aravind, L. 2009. Prediction ofnovel families of enzymes involved in oxidative and other complex modifications of bases in nucleic acids. Cell Cycle 8: 1698-1710 Altschul, SF, Gish, W, Miller, W, Myers, EW, Lipman, DJ. 1990. Basic local alignment search tool. J. Mol. Biol. 215: 403-410 Laemmli, UK. 1970. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature (London) 227: 680-685 Brems, S, Guilbride, DL, Gundlesdodjir-Planck, D, Busold, C, Luu, VD, Schanne, M, Hoheisel, J, Clayton, C. 2005. The transcriptomes of T rypanosoma brucei Lister 427 and TREU927 bloodstream and procyclic trypomastigotes. Mol. Biochem. Parasitol. 139: 163-172 Ryle, MJ, Padmakumar, R, Hausinger, RP. 1999. Stopped-flow kinetic analysis of Escherichia coli taurine/a-ketoglutarate dioxygenase: interactions with or- ketoglutarate, taurine, and oxygen. Biochemistry 38 : 15278-15286 Simmons, JM, Mijller, TA, Hausinger, RP. 2008. Fen/OL-ketoglutarate . hydroxylases involved in nucleobase, nucleoside, nucleotide, and chromatin metabolism. Dalton Trans. 38: 5132-5142 Gommers-Ampt, JH, Van Leeuwen, F, de Beer, AL, Vliegenthart, JF, Dizdaroglu, M, Kowalak, JA, Crain, PF, Borst, P. 1993. B-D-glucosyl-hydroxymethyluracr1: a novel modified base present in the DNA of the parasitic protozoan T. brucei. Cell 75:1129-1136 Borst, P, Sabatini, R. 2008. Base I: discovery, biosynthesis, and possible functions. Annu. Rev. Microbial. 62: 235-251 48 19. 20. 21. 22. 23. 24. Ulbert, S, Cross, M, Boorstein, RJ, Teebor, GW, Borst, P. 2002. Expression of the human DNA glycosylase hSMUGl in Trypanosoma brucei causes DNA damage - and interferes with J biosynthesis. Nuc. Acids Res. 30: 3919-3926 Gommers-Ampt, J H, Teixeira, AJ, van de Werken, G, van Dijk, WI, Borst, P. 1993. The identification of hydroxymethyluracil in DNA of T rypanosama brucei. Nuc. Acids Res. 21: 2039-2043 Cross, M, Kieft, R, Sabatini, R, Wilm, M, de Kort, M, van der Marel, GA, van Boom, J H, van Leeuwen, F, Borst, P. 1999. The modified base J is the target for a novel DNA-binding protein in kinetoplastid protozoans. EMBO J. 18: 6573-6581 DiPaolo, C, Kieft, R, Cross, M, Sabatini, R. 2005. Regulation of trypanosome DNA glycosylation by a SW12/SNF2-like protein. Mol. Cell 17: 441-451 Yu, Z, Genest, PA, ter Riet, B, Sweeney, K, DiPaolo, C, Kieft, R, Christodoulou, E, Perrakis, A, Simmons, JM, Hausinger, RP, van Luenen, HG, Rigden, DJ, Sabatini, R, Borst, P. 2007. The protein that binds to DNA base J in trypanosomatids has features of a thymidine hydroxylase. Nuc. Acids Res. 35: 2107-2115 Vainio, S, Genest, PA, ter Riet, B, van Luenen, H, Borst, P. 2009. Evidence that J-binding protein 2 is a thymidine hydroxylase catalyzing the first step in the biosynthesis of DNA base J. Mol. Biochem. Parasitol. 164: 157-161 49 CHAPTER 2 APPENDIX The docking studies were performed by Mike Howard. 50 2A.1. INTRODUCTION Chapter 2 investigated the possible functions of two trypanosomal thymine hydroxylase-like proteins, TLP5 and TLP7 from T rypanasoma brucei brucei. These two peptide sequences are 85% identical to each other and about 30% identical to a fungal (Rhodatorula glutinis) thymine 7-hydroxylase (T7H), which functions in a pyrimidine salvage pathway (1,2). Trypanosomes are not known to possess a pyrimidine salvage pathway, but they have been shown to employ the activity of a thymidine hydroxylase in the first step of the synthesis of a modified base, B-D-glucosylhydroxymethyldeoxyuracil or base J, in their nuclear DNA (3,4). Thus, I tested whether TLP5 and TLP7 may be involved in this nucleotide modification in trypanosomes. The genes were cloned from the T. b. brucei genome and the proteins were overproduced in E. coli and purified by column chromatography. The properties of these recombinant proteins were characterized by several biochemical methods and the expression of the genes was compared for the bloodstream and procyclic forms of the protozoan. Much of that work was compiled and published in Experimental Parasitology (see main body of Chapter 2); the experiments omitted fi'om that publication are described here. This appendix is organized in a modular fashion. Each module explains the rationale for that method and describes the procedure, results, and any conclusions drawn from the experiments. 51 2A.2. OLIGOMERIC STATE DETERMINATION To determine their native sizes and Oligomeric states, the purified and concentrated recombinant TLP5 and TLP7 proteins were chromato graphed on a Superdex®75 size exclusion column and the elution positions were compared to those of standard proteins (BioRad). The collected fractions also were examined by polyacrylamide gel electrophoresis using a 3-12% Bis-Tris Blue native gradient gel from lnvitrogen to verify the major Oligomeric form of both TLP5 and TLP7. Figure 2A.1 shows that multiple forms of TLP5 were present as purified from E. coli. The major peak in the chromatograrn (panel A) eluted at a position corresponding to 36.6 :1: 4 kDa and contained a monomer as the predominant form, as seen on the native gel (panel B), although significant levels of the dimeric or trimeric species also were. present during electrophoresis, consistent with an equilibrium mixture. The minor feature preceding the main peak in the chromatograrn corresponded exclusively to a dimer or trimer. Finally, the first small peak in the chromatograrn appeared to consist of a high molecular mass contaminant that electrOphoresed as a smear on the gel. These results show that the TLP5 protein is predominantly a monomer as purified, though with some dimer or trimer present. Similar results were observed with TLP7 (data not shown). 52 Figure 2A.1. Native size and Oligomeric state determination of TLP5. A. Chromatogram of TLP5 run on a Superdex®75 column and compared with BioRad gel filtration standards (retention times of y-globulin, 158 kDa; ovalbumin, 44 kDa; and myoglobin, 17 kDa, shown as inverted triangles above chromatogram). B. Native polyacrylamide gel showing the Oligomeric state and native size of TLP5. Dashed lines correspond to those on the chromatogram to indicate the fractions run on the gel. A. V V V 50 55 50 65 70 75 so B. 1236__ . .. 1048—‘ .. 720-— “'7'” 480—- - 242“— 146—'“ 66—— ~ 20 ‘ 53 2A.3. HOMOLOGY MODEL There is no crystal structure for TLP5 or TLP7, nor has the fungal T7H been structurally characterized. To choose a suitable parent structure for the construction of a homology model, the protein data bank was searched using the Bioinfo Meta Server (http://metgbioinfopl) and using the amino acid sequence of TLP5 as a query. Based on sequence identity to proteins of known structure, anthocyanidin synthase from Arabidopsis thaliana (PDB 1GP4), with a 33 % identity, was used as a parent structure and the sequence of TLP5 was mapped directly onto the backbone (C01) atom coordinates from the crystal structure. Side chains were further modeled into the structure using the program SCWRL (5). Finally, the iron atom and the aKG were modeled into the structure based on the coordinates of the anthocyanidin synthase structure containing those elements. The basic structure of the catalytic core, conserved among all iron/aKG dioxygenases, was observed in TLP5 (Figure 2A.2 panel A). The facial triad residues made up of His 200, Asp 202, and His 257 in TLP5 were correctly positioned to coordinate a metal atom. Further, when the iron atom and the aKG cosubstrate were modeled into the active site, the coordination distances observed were within the normal range seen in this family of enzymes (6). In addition, a highly conserved arginine known to participate in the coordination of the C5 carboxyl group of aKG in other members of this enzyme family also was present here, at position 266. The coordinates for the model were provided to undergraduate student Mike Howard, who submitted them as a protein target to the program Dock Blaster (http://blaster.docking.org) to identify possible classes of ligands that may fit into the 54 modeled active site (7). A specified active site, using the residues pictured in Figure 2A.3, also was submitted in PDB format. This program used the ZINC database of commercially available small ligands (8) and docked them into the active site based on the lowest energy binding modes. Ligand candidates were then scored and ranked by binding energies. The program identified the three best ligands for the TLP5 homology model active site, which were all ring-based structures resembling nucleic acids. The C-terminal a-helix of the TLP5 homology model was predicted to rest against the putative opening to the active site and appeared poised to act as a gate, limiting access to the oxygen-activating iron atom (Figure 2A.2 panel B). This model led to two hypotheses that were experimentally tested. First, the polyhistidine tag, added during the cloning process to facilitate protein purification, may have interfered with movement of the C-terminal helix required for activity and would thus explain the lack of activity observed to that point. Therefore, the purification tag was moved to the N-terrninus and attempts to characterize the alternatively tagged protein were repeated (subsection 4). Second, one could postulate that another factor in the trypanosome, but not present in my in vitro system, could induce movement of the C-terrninal helix and allow activity. To test this hypothesis, a truncation mutant was created that eliminated the C-terminal 13 residues making up that helix and the corresponding protein was characterized (subsection 5). 55 Figure 2A.2. Homology model of TLP5. A. Ribbon diagram colored as a spectrum from the N-terminus in blue to the C-terminus in red. Breaks in the model represent areas where there was insufficient homology to model. The iron-coordinating facial triad and aKG-stabilizing arginine are shown as sticks and colored by element (carbon-green, oxygen-red, nitrogen-blue). The aKG is also shown as sticks and colored by element (carbon-cyan, oxygen-red). The iron atom is shown as a magenta sphere. B. Surface representation of the same model colored by element (carbon-white, oxygen-red, nitrogen-blue, sulfur-yellow). Figures were created using PyMol. A. B. 56 Figure 2A.3. Results of docking a subset of the ZINC small molecule database into the modeled active site of TLP5. Each panel has a chemical structure and a modeled binding depiction of a low-energy, docked ligand molecule. TLP5 is shown as magenta lines, the iron atom as an orange sphere, the aKG as either yellow or purple sticks, and the docked ligand as blue sticks. Panel C is rotated 60° from the angle of panels A and B to better show the possible molecular interactions. Binding figures were created using PyMol. 57 2A.4. RELOCATION OF PURIFICATION TAG To investigate whether the C-terminal histidine tag interfered with the activity assays, the purification tag was moved to the N-terrninus of the protein. The gene encoding TLP5 was cloned out of its initial expression plasmid, pET28b (Novagen), by using forward (5’-AGGATATAGC TAGCATGGCT CACGGCTCGA T-3’) and reverse (5’-GTGGTGGTGC TCGAGTCACA TCT'I‘TGTI‘TT G-3 ’) primers to reintroduce the stop codon before the C-terminal purification tag and move the start site downstream of a sequence encoding the N-terrninal polyhistidine purification tag. This process introduced six extra residues between the purification tag and the initial methionine of the actual protein sequence. The recombinant protein was overproduced in E. coli and purified as before (see main body of Chapter 2) though the protein was less stable and precipitated after even brief storage at 4 °C. The recombinant, N-terrninally-tagged TLP5 protein was concentrated to about 180 uM and assayed for formation of the diagnostic metal-to-ligand charge-transfer chromophore in the presence of Fell and OLKG as seen in the original C-terrninally tagged protein preparation. The spectroscopic assay was performed as described in the main body of Chapter 2 and no chromophore was observed. This result indicated that the protein with the extra N-terminal residues did not fold properly and was thus unable to bind the iron and aKG in the correct orientation. Therefore, the N-terminally tagged protein was not used for further experiments. 58 2A.5. C-TERMINAL TRUNCATION The C-terrninal a-helix of TLP5 appeared to occlude access to the active site in the homology model, fireling the hypothesis that removing those residues may provide access to the active site in the absence of some required in viva post translational modification or protein-induced activation. To test this hypothesis, a 13 residue truncation mutant was created to remove this helix. The gene encoding TLP5 was cloned out of genomic trypanosome DNA using the original forward primer and the new reverse primer (5’-GAGCATCCTC GAGAGTCTTC AGCAACCAAT CC—3’) to replace the sequence encoding the C-terminal 13 residues with a stop codon. Truncated protein, termed TLP5AC13, was produced in E. coli and purified as described in the main body of Chapter 2. The shortened protein was concentrated to 65 pM and assayed anaerobically for the characteristic metal-to-ligand charge-transfer chromophore as before. The feature was present, with an estimated extinction coefficient of 60 M'1 cm'l indicating that the feature was incompletely formed compared with previously reported intensities (9). TLP5AC13 also was assayed for enzymatic activity by measuring oxygen consumption with an oxygen electrode, but no activity was observed for any of the substrates tested including thymine, thymidine, and oligo polyT. Finally, the truncated protein was assayed for DNA binding (see subsection 6 for method). No binding was observed with any of the substrates tested including supercoiled and linearized plasmids, with or without alkylation, or HindIII lambda DNA fragments. This protein tended to precipitate during experiments; therefore, further experiments were not performed with this mutant. 59 2A.6. DNA BINDING The sequence similarity of TLP5 and TLP7 to the fungal T7H led to the hypothesis that the trypanosomal proteins may be nucleotide-modifying enzymes. One possibility is that the substrate is a polynucleotide, therefore the proteins were tested for their abilities to bind to various DNA substrates via an electrophoretic mobility shift assay using agarose gels. Briefly, proteins were incubated with DNA and various supplements for 30 min at 37 °C. Glycerol was added to the samples to 5% final concentration and they were loaded onto a 0.8 % agarose gel and run in 89 mM Tris borate buffer. Additives tested in the binding assays included binding buffer (30 mM imidazole and 10 mM Tris, pH 7.9, containing 150 mM NaCl), Tris buffer (25 mM Tris, pH 8.0, containing 100 mM KCl and with or without 5 mM MgCl2, 0.5 mM DTT, or 0.2 mM EDTA), or Hepes buffer (50 mM Hepes, pH 8.0, containing 40 uM CoCl2, 100 pM ascorbate, and 200 uM aKG). Other assay components included in some of the assays included up to 200 uM uKG, 50 uM F e", and 100 uM ascorbate. DNA substrates included supercoiled plasmid, linearized plasmid, methylated plasmid, and HindlII A DNA fragments. No shift in the DNA bands was observed that was greater than the variation in band positions present in lanes without protein. This result, taken together with the lack of detectable enzymatic activity challenged the hypothesis that TLP5 and TLP7 were polynucleotide-modifying enzymes. Therefore, other methods were employed to look for possible substrates and functions of these proteins. 60 2A.7. IN WVO EPITOPE TAGGING OF THE TLP5 GENE Data acquired by quantitative reverse transcriptase PCR amplification of T LP5 and T LP7 indicated that the steady state mRNA levels were higher in the bloodstream form trypanosomes than the procyclic forms. While interesting, the picture remained incomplete as most of the genetic regulation in trypanosomes takes place at the level of protein expression and turnover (10). Therefore, a construct was designed to add sequence coding for a nine residue hemagglutinin (HA) epitope tag to the chromosomal copy of T LP5. This would allow the analysis of in viva protein levels under various stress conditions as well as determination of the subcellular localization for the protein, and it would provide a method to observe changes in the protein production levels in concert with other procedures such as RNAi. The initial HA tagging construct was acquired from Professor Donna Koslowsky. It consisted of the sequence coding for the HA tag, an a/B tubulin spacer region, and a blasticidin resistance gene. About 100 nucleotides corresponding to the 3’ end of the T LP5 gene were added to the 5’ end of the construct by PCR amplification using the primer (5’- CCA'I'TCGCCA AGCAATCCCC CCAAATATCC ACCAGTCCGT GCTGTGGATT GGTTGCTGAA GCGTTTCGCG GAAACATATG CCCATCGCAA AACAAAGATG TACCCATACG ACGTCCCAGA CTACGCTIAA- 3’) where the stop cation is underlined. In addition, about 100 nucleotides corresponding to the beginning of the 3’ untranslated region was added to the 3’ end of the construct by PCR amplificationusing the primer (5’- GAAAAGAAAA AAAAAGAGTT TGAAGACACA "ITCATTCATI’ CATTGCCACC GCATCCGTAC CGACGGGGGC CTAACGAAAA GATTACGTTC 61 AGTTATTGAT ILAGCCCTCC CACACATAAC CAGAG -3’) where the reverse complement of the stop codon for the blasticidin resistance gene is underlined. This first round of PCR amplification created the unique tagging construct needed for my tagging procedure. Next, this unique construct was amplified by a second round of PCR using the forward primer (5’- CCATTCGCCA AGCAATCCCC CCAAA -3 ’) and the reverse primer (5’- GAAAAGAAAA AAAAAGAG'IT TGAAGACACA TTCATTCATT -3 ’) (Figure 2A.4). The PCR products were separated from unused reagents by using the Qiagen PCR clean-up kit and used directly for trypanosome transfections. In replicate trials, 2.5 or 5 pg of the DNA-tagging construct were added to 108 procyclic trypanosome cells in 0.5 mL phosphate-buffered sucrose in a BioRad electroporation cuvette. Cells were treated in a BioRad electroporator at 1.5 kV, 25 uF, and infinite resistance for two pulses Spaced by 10 sec. Cells were transferred immediately to a small culture flask containing 4.5 mL of SDM-79 medium with 10 % fetal bovine serum and 7.5 ug/mL hemin (as an iron source) along with penicillin and streptomycin to select against bacteria. Cultures were allowed to grow at 27 °C overnight with gentle shaking before adding the eukaryotic selection drugs: 15 ug/mL neomycin (to maintain the tet repressor), 100 ug/mL hygromycin (to maintain the T7 polymerase), and 10 ug/mL blasticidin (to maintain our tagging construct). Cells were grown for an additional two days and then serially diluted in the same selective media to create a more clonal population. One of the dilution cultures was grown for approximately a week before some of the cells were harvested and genomic DNA extracted to test the incorporation of the tagging construct. 62 The entire T LP5 gene segment of chromosome 5 was amplified by PCR using the forward primer (5’- GCCATTTTT CTGTGAGCCT AATCC -3 ’) and the reverse primer (5’- TCAGG ACAGATACGA GCAAAAGAAG-3’). With the incorporated tag, the lengths of the products would be a mixture of approximately 1 kb for the tagged allele and 300 bases for the untagged allele, since trypanosomes are diploid. In addition, the segment was amplified by using primers internal to the tagging sequence that would further test its placement when paired with the primers described above; these hybridize to regions just upstream and downstream of the sequences included in the original unique-tagging construct. The internal primers corresponded to a forward segment near the end of the blasticidin resistance gene (5’-GGGATTCGTGAATTGCTGCCCTCT-3’) and a reverse segment (5’-TGTACCACGCTGCAACAGTGTGAAAATTCGA-3’) near the beginning of the 01/0 tubulin spacer (Figure 2A.4). One culture out of six separate tagging experiments, each with six transfections, appeared to have the correct tag incorporation in at least one allele as seen by the PCR verification. Therefore, a whole cell lysate was made from a sample of that culture and was Western blotted using the anti-HA antibody conjugated to alkaline phosphatase as a probe. Briefly, 107 cells were harvested by centrifugation and resuspended in 1 mL cell wash buffer (20 mM Tris, pH 7.5, containing 100 mM NaCl and 3 mM MgCl2) and placed in a bath sonicator with ice for about 20 min. Debris was removed by centrifugation at 16,000 g for about 10 min. The supernatant was electrOphoresed on a denaturing 12% polyacrylamide gel, proteins were transferred to an ImmobilinP PVDF membrane. (Millipore), and the membrane was probed with rabbit anti-HA antibody from Sigma. Unfortunately, no protein band of the appropriate size was detected. Several high 63 molecular weight bands were detected that are commonly seen in blots of trypanosome extracts probed with the anti-HA antibody (Donna Koslowsky personal communication). 64 Figure 2A.4. Hemagglutinin epitope tagging of T LP5. This figure is a construction schematic where the nine residue hemagglutinin (HA) epitope is shown in green, the a/B tubulin spacer in yellow, and the blasticidin resistance gene in blue. The 3’ end of the T LP5 gene is shown in red and the 3’ untranslated region in brown as parts of the primers used to make the unique tagging construct by PCR. This construct was amplified by a 2° PCR reaction using primers whose positions are indicated by the red and brown lines above the scheme. Finally, primers whose positions are indicated by the red and brown open boxes and the grey arrows were used to test the incorporation of the tag into chromosome 5 of the trypanosome. 1 =- Tx‘q- A ‘0‘? . ‘3‘! ’3: “a. ._, 01/0 Tubulin —__ _ _ _B,SR . _ YPYDVPDYA l1° PCR . 2° PCR , :; .. . . 3 m '- - HA}- a/p Tubulin -_ BSR :3 +— -——-> 65 2A.8. PULL-DOWN ASSAYS WITH TLP5 AND TRYPANOSOME CELL EXTRACT All attempts to detect activity with various nucleic acid substrates failed, therefore pull down assays with trypanosome cell extracts were used in an effort to find potential protein interaction partners of TLP5. E. coli cells over-expressing TLP5 were lysed, sonicated, and ultracentrifuged to generate soluble cell extracts. This sample was added to Ni-NTA cellulose beads and the histidine tagged TLP5 allowed to bind to the resin. The beads were washed with binding buffer twice and then trypanosome soluble cell extracts (prepared as before) were added and incubated at room temperature for 20 min to allow binding. Again, the resin was washed with binding buffer and eluted in elution buffer (150 mM imidazole and 10 mM Tris, pH 7.9, containing 150 mM NaCl). Extract, wash, and elution samples were electrOphoresed on a denaturing 12% polyacrylamide gel and silver stained to detect proteins of low abundance. The gel lanes were compared as described above for E. coli extracts, T. b. brucei extracts, and mixtures of the extracts. With three separate experiments, including analyses using free resin in an eppendorf centrifuge tube and resin packed into a small column, no new bands were identified in the combined extract runs that were not also present in the control runs. Thus, the pull-down experiments failed to identify potential interaction partners of TLP5. 66 2A.9. CONCLUSIONS The thymine hydroxylase-like proteins TLP5 and TLP7 from T. b. brucei have been characterized and determined to be members of the non-heme iron and aKG dependent dioxygenase superfamily of enzymes. Extensive efforts were undertaken to identify the activity of these proteins, though none of the tested hypotheses were supported. Under the conditions used, no DNA binding was observed and no activity was demonstrated using various substrates, even when examining modified proteins to eliminate possible active site access restrictions. Pull-down assays failed to identify protein interaction partners and docking studies did not yield obvious ligand choices to pursue. While the true purpose of these proteins in the cell remains enigmatic, they probably do not participate in polynucleotide modification but are very likely to catalyze some other type of hydroxylation reaction in the cells. 67 10. REFERENCES Smiley, JA, Kundracik, M, Landfried, DA, Barnes, VR, Sr., Axhemi, AA. 2005. Genes of the thymidine salvage pathway: thymine-7-hydroxylase from a Rhodatorula glutinis cDNA library and iso-orotate decarboxylase from Neurospora crassa. Biochim Biophys Acta 1723: 256-264 Neidigh, JW, Darwanto, A, Williams, AA, Wall, NR, Sowers, LC. 2009. Cloning and characterization of Rhodatorula glutinis thymine hydroxylase. Chem Res T oxicol 22: 885-893 Gommers-Ampt, JH, Van Leeuwen, F, de Beer, AL, Vliegenthart, JF, Dizdaroglu, M, Kowalak, JA, Crain, PF, Borst, P. 1993. beta-D-glucosyl- hydroxymethyluracil: a novel modified base present in the DNA of the parasitic protozoan T. brucei. Cell 75: 1129-1136 Gommers-Ampt, J H, Teixeira, AJ, van de Werken, G, van Dijk, WJ, Borst, P. 1993. The identification of hydroxymethyluracil in DNA of T rypanosoma brucei. Nucleic Acids Res 21 : 2039-2043 Canutescu, AA, Shelenkov, AA, Dunbrack, RL, Jr. 2003. A graph-theory algorithm for rapid protein side-chain prediction. Protein Sci 12: 2001-2014 Hausinger, RP. 2004. Fe(II)/or-ketoglutarate-dependent hydroxylases and related enzymes. Crit. Rev. Biochem. Mal. Biol. 39: 21-68 Irwin, JJ, Shoichet, BK, Mysinger, MM, Huang, N, Colizzi, F, Wassam, P, Cao, Y. 2009. Automated docking screens: a feasibility study. J Med Chem 52: 5712- 5720 Irwin, IJ, Shoichet, BK. 2005. ZINC-~a free database of commercially available compounds for virtual screening. J Chem Inf Model 45: 177-182 Ryle, MJ, Padmakumar, R, Hausinger, RP. 1999. Stopped-flow kinetic analysis of Escherichia coli taurine/oc-ketoglutarate dioxygenase: interactions with or- ketoglutarate, taurine, and oxygen. Biochemistry 38: 15278-15286 Clayton, C, Shapira, M. 2007. Post-transcriptional regulation of gene expression in trypanosomes and leishmanias. Mol Biochem Parasitol 156: 93-101 68 CHAPTER 3 SEARCH FOR HYDROXYLASE ACTIVITY IN THE TRYPANOSOMAL PROTEIN, JBPl Portions of these studies were published along with experiments conducted by others in: Yu, Z, Genest, PA, ter Riet, B, Sweeney, K, DiPaolo, C, Kieft, R, Christodoulou, E, Perrakis, A, Simmons, JM, Hausinger, RP, van Luenen, HG, Rigden, DJ, SabatiniR, Borst, P. 2007. The protein that binds to DNA base J in trypanosomatids has features of a thymidine hydroxylase. Nuc. Acids Res. 35: 2107-21 15 The introduction for this chapter mentions some of the other studies, citing this reference, as well as prior background and more recent results by others. 69 3.1 INTRODUCTION Trypanosomes are eukaryotic kinetoplastid flagellates and the causative agents of several neurological diseases affecting mammals. Chagas Disease, caused by T rypanosama cruzi and transmitted by redurid bugs, leishmaniasis, associated with various strains of Leishmania and transmitted by sandflies, and African Sleeping Sickness, caused by T rypanosoma brucei and carried by the tsetse fly are examples in humans (1,2). About one percent of the thymine in the telomeric repeats of nuclear DNA in the mammalian bloodstream form (BF) of T. brucei is replaced with B-D-glucosyl- hydroxymethyldeoxyuracil (base J) (3 ,4). Even smaller amounts of the modification are found in subtelomeric regions and very little in chromosome internal locations. This modified base has several possible functions. First, it is proposed to induce chromatin compaction and thereby stabilize the chromosome from rearrangements within these repetitive sequences (5 ,6). Second, base J has a proposed role in the process of antigenic variation characteristic of T. brucei and other related parasites. Base J is found throughout the silent variable surface glycoprotein (VSG) expression Sites and the flanking repetitive sequences, but no I is present in the one active expression site, suggesting a specific role for J in gene silencing (7). Finally, base I has been identified in the chromosomal internal polymerase II polycistronic transcription unit strand switch regions of several kinetoplastids, including T. brucei and L. major, as well as the spacer regions between individual open reading frames, suggesting a role for base J in the wider family of kinetoplastids that do not all undergo antigenic variation (8). Base J is synthesized in two steps. First, the methyl group on the C5 position of a thymidine already incorporated into DNA must be hydroxylated to create the 70 intermediate hydroxyrnethyldeoxyuridine (HOMedU) (Figure 3.1 panel A), as evidenced by the presence of low levels of HOMedU in BF trypanosome DNA (9). Next, the hydroxymethyl moiety must be glucosylated. This reaction is not specific to the sequence or life cycle-stage as evidenced by the random placement of base I in the bloodstream and procyclic forms of cells grown on HOMedU (5). Recently, a protein was identified in T. brucei that specifically binds to base I in DNA (10). This I binding protein 1 (JBPI) is ~90 kDa protein whose N terminal domain shows some sequence Similarity to other non-heme iron and a-ketoglutarate (aKG) dependent hydroxylases by manual alignment (1]). Secondary structure prediction further places this protein in the double-stranded beta helix or “jelly-roll” core fold family of proteins (11). This homology and the obvious firnctional similarity to the methylated DNA hydroxylase AlkB (a DNA repair enzyme) and the fungal thymine-7- hydroxylase, which is involved in the pyrimidine salvage pathway and acts only on the free base, fueled the hypothesis that JBPl may be the thymine hydroxylase in trypanosomes. JBP2, identified in silico based on its sequence identity to JBPl, does not bind to J -containing DNA, but does possess a SWI-2/SNF-2 chromatin remodeling domain at its C—terminal end and is capable of inducing de novo base J synthesis when expressed in procyclic T. brucei cells (12,13). These cells normally do not express JBPl or JBP2 and lack base I (3). JBPl is suggested to maintain 1 levels by stimulating additional J synthesis in nearby sequences while cells containing modified DNA, but lacking JBPl , lose base J by dilution during replication (14). Further work to establish the roles of these proteins has shown that a JBPl knockout in Crithidiafasciculata (a non-pathogenic 71 Figure 3.1. Synthesis of base J. A. Scheme showing the two step molecular synthesis of base J. The R group represents the continuing strands of DNA. B. Cartoon depicting the proposed chromatin specific introduction and maintenance of base J. J BP2 is responsible for the initial introduction of telomeric and subtelomeric base I, while JBPl is responsible for the initial introduction of chromosome internal base I (8). JBPl then binds to these regions and stimulates amplification of the modified base (15). GT- glucosyl transferase. Figure modified from (1 l). A: NH aKG succinate H NH o co A N 2 2 N R O , I R O JBP1/JBP2 " ll 0R containing Fe OR dT HMdU Glucose B-glucosyl transferase O Ho/yé/O/TLNH H OH o NAG OR dJ strain) reduces J levels to 5% of the wild-type cells (14), a JBP2 knockout reduces base J levels to 20% of the wild-type cells in studies involving T. brucei and L. tarentolae (12,16), and a double knockout creates a J-null cell line in T. brucei (8,15). Surprisingly, both J BPl and JBP2 proteins are dispensable in T. brucei with no phenotypic or growth effects (8,15), while JBP 1 is essential in L. tarentolae ( l 7) and JBP2 is not (16). Finally, it has been shown that mutation of the key conserved residues making up the “facial tria ” of the active site or the conserved arginine believed to stabilize the aKG cosubstrate in these proteins abolishes their ability to stimulate base J synthesis (1 1,15,16). Understanding the enzymes involved in J biosynthesis may lead to the development of drugs to treat trypanosomiases, as this modified base is found only in the kinetoplastid flagellates. Yet, no direct in vitro hydroxylase activity has been detected for either JBPI or J BP2. Therefore, this study sought to demonstrate enzymatic or uncoupled turnover of J BPI. To test JBPl for aKG dependent hydroxylase activity, I used recombinant protein with in vitro assays to look for product formation and substrate consumption. Other members of this enzyme family have been shown to decarboxylate aKG and consume oxygen in the absence of their primary substrate, termed uncoupled turnover. These studies primarily focused on the uncoupled reactions of JBPl due to the lack of sufficient amounts of primary substrate, genomic trypanosome DNA containing base J. Some putative alternate substrates were tested in attempts to measure the coupled turnover or stimulate uncoupled turnover. 73 3.2 MATERIALS AND METHODS 3.2.] Determination of succinate production as a measure of uncoupled enzymatic turnover. Purified recombinant JBPI was supplied by Henri van Luenen of Piet Borst’s lab at The Netherlands Cancer Institute in Amsterdam. JBPl was from C. fasciculata (not pathogenic to mammals) or Leishmania tarentolae in wild type or mutated forms. One mL reactions in 13 x 100 glass tubes, to provide for efficient aeration, were incubated at either 30 or 37 °C in either 25 mM Tris or 50 mM Hepes buffer, with the pH ranging from 7.0 to 8.5. Conditions tested included assays containing ferrous iron (0.05-1 mM), ascorbate (0.05-5 mM), aKG (0.5-20 mM), NaCl (20 mM), MgCl2 (0.5-10 mM), KCl (IO-100 mM), EDTA(0 or 20 uM), glycerol (0 or 5%), or Triton X-100 (0 or 0.02%), and as potential substrates, thymidine (20 mM), DNA oli go containing base J (20 uM), and genomic trypanosome DNA (40 uM). Aliquots of 300 pL were taken at various time points and quenched with 5 uL of 6 M sulfuric acid. Samples were centrifuged at 16,000 g for 5 min to remove any protein. 250 uL was loaded onto a 0.45 pm cutoff spin filter and centrifuged. 200 uL of the flow through fraction was loaded onto an Aminex HPX-87H ion exchange organic acids column (BioRad) (300 mm x 7.8 mm) and analyzed by high performance liquid chromatography (HPLC). Organic acids were eluted with 0.013 M H2304 as the mobile phase and detected by using a Waters refractive index detector set at 35 °C and a , sensitivity level of 16. The standards of aKG and succinate eluted at 19.8 and 26.6 min, 74 Figure 3.2. aKG and succinate detection by HPLC. A. A representative chromatogram showing the retention times for aKG (19.8 min) and succinate (26.6 min). B. Standard curve showing the detection of succinate down to a lower limit of 5 uM in the injected volume. The R2 value was 0.9999 and the fitted line was used to determine the amount of succinate produced in an assay based on the integrated area of the detected peak. A. 25.002 20004 I 15.0041 > 1 2 1000-; , 1 coo—lfrfrrrrrrT'rrTurrrunrfirr TTT 'Tu . ' ‘ ' ltf’ffif'l ' TTTFTTTTTTTT r-TTTT 19.00 20.00 21.00 22.00 23.00 24.00 25.00 20.00 27.00 28.00 29.00 30.00 Retention time (min) B. A 400 - 8 o 350 T I? v 300 4 8 I- 250 a < .K a 200 - 01 °- 150 1 8 *6 100 1 a a, 50 . ‘E -' 0 m T T l_— LL; 0 50 100 150 200 250 Succinate (pM) 75 respectively, and succinate was detectable to a lower limit of 1 nmol or 5 uM in the injected volume (Figure 3.2). 3. 2.2 Oxygen consumption as a measure of uncoupled enzymatic turnover. Assays were performed by using a micro oxygen electrode in an Instech chamber coupled to a circulating water bath to maintain a temperature of 33° C and a Diamond Electra-Tech chemical microsensor. Percent oxygen saturation was continuously monitored throughout the assay. The upper and lower saturation limits were set by flushing the system with buffer (25 mM imidazole, pH 7.0) that was bubbled with air or degassed buffer bubbled with nitrogen, respectively. JBPl, as received from the Netherlands, was in 25 mM Hepes, pH 7.0, with 150 mM NaCl and 1 mM EDTA. A portion of the protein was dialyzed against 25 mM imidazole, pH 7.0, and 150 mM NaCl to remove the EDTA. JBPl with or without EDTA was assayed at 1 uM using variations of the standard conditions: 50 uM ferrous iron, 1 mM aKG, 400 uM ascorbate, 6.5 mM NaCl, and 1 mM of either thymine, thymidine, or oligo polyT. Assays were carried out in 25 mM imidazole, pH 7.0, in the micro-oxygen electrode with an assay volume of 600 uL. 3.2.3 Spectroscopic detection of a Metal-to-Ligand Charge- Transfer (MLC T) Feature. UV/visible spectra were obtained by using an HP 8453 spectrophotometer (Hewlett Packard) equipped with a circulating water bath. Stock solutions of 10 mM Fe“ and 5 mM 01KG were made by adding the dry reagents to vials, subjecting them to repeated cycles of vacuum and argon, and adding anaerobic water by using a syringe. 76 The JBPl protein was provided in a high salt buffer with a reducing agent for shipping, therefore to prevent possible interference in these experiments it was first exchanged (at 4 °C by repeating concentration/dilution by using an Amicon double-membrane centrifiigal concentrator with a 10 kDa cutofl) into 25 mM Tris, pH 8.0, containing 50 mM KCl. 50 uM protein was added to a l—cm path length, 300 uL, black-walled cuvette and made anaerobic by the same method, followed by scanning at 9 °C. Four equivalents of aKG were added followed by a titration of one equivalent of Fe" into the cuvette in 0.2 equivalent increments with scans after each addition. Data were analyzed by using the software IGOR6. First, data were corrected for baseline shifts arising from cuvette repositioning to have a uniform absorbance at 850 nm. Then, data points over a 5 nm range of wavelengths on each side of every point were averaged to smooth the data and the wavelength maxima of the observed features were determined by taking the second derivative. 77 3.3 RESULTS AND DISCUSSION The trypanosome protein J BPl is known to bind to J -containing DNA and has been implicated in the process of antigenic variation and chromatin maintenance. It was hypothesized that the C-terminal domain functions in J base recognition and binding while the N-terrninal domain serves as a non-heme iron and aKG-dependent hydroxylase responsible for the initial step in the Synthesis of newly modified bases in adjacent DNA regions. If this is the case, then oxygen and aKG would be consumed in the reaction and succinate would be produced. Assays for oxygen consumption and aKG decomposition were performed by using an oxygen electrode and HPLC, respectively, with purified recombinant JBPl that was known to bind DNA oligos containing base J (as evidenced by gel band Shifts). Methods included assays for both coupled and uncoupled turnover. The assay was verified using taurine hydroxylase. Various nucleic acid substrates were tested, though the amounts available were severely limiting. None of the assays analyzed by oxygen electrode exhibited oxygen consumption greater than the background levels measured in the absence of protein. The background oxygen consumption is thought to arise from the oxidation of ascorbate in the presence of iron (18). In a similar manner, none of the assays analyzed by HPLC Showed appreciable production of succinate over a basal level that was measurable when no protein was present. This background amount of succinate is thought to arise from the spontaneous breakdown of OLKG in the presence of iron, oxygen, and ascorbate. 78 To confirm proper folding of J BPl , the purified recombinant protein was examined for its ability to form a characteristic MLCT chromophore seen in other members of the Fen/(IKG-dependent dioxygenases and associated with binding of the iron cofactor and the aKG cosubstrate (19). The protein was monitored by UV-visible spectroscopy under anaerobic conditions, using low temperature to maintain stability, while titrating in ocKG and Fe". As shown in Figure 3.3, anaerobic JBPl generated the diagnostic MLCT transition at 504 nm when both metal and cofactor were present. Due to the light scattering underneath the feature, an extinction coefficient was not estimated from this data. While J BPl and J BP2 have been shown by mutagenesis studies to be responsible for the specific introduction of base J into kinetoplastid DNA, in vitro activity has not been directly detected for either protein. JBPl purified from recombinant cells was shown to be properly folded and capable of binding iron and aKG in a manner that formed a characteristic MLCT spectrum. This result verifies the protein’s membership in the non-heme iron and aKG dependent dioxygenase superfamily of enzymes. Since the activity assays performed here were unsuccessful, it is possible that the substrate for the J BPl enzyme may be a Specific form of chromatin that is recognized by its structure rather than sequence. Therefore, even the genomic trypanosome DNA containing base J, would not elicit enzymatic turnover under the conditions we tested. 79 Figure 3.3. Spectroscopic evidence for binding of F e" and aKG by JBPl. The anaerobic UV/visible spetrum of J BPl (50 uM in 25 mM Tris, pH 8.0 containing 50 mM KCl) was examined for the sample as prepared, after adding 200 uM aKG, and while titrating in Fe]I with stirring at 9 °C. The (Fell-aKG-protein minus aKG-protein) difference spectra shown correspond to the addition of 0 (baseline), 0.2, 0.4, 0.6, 0.8, and 1 equivalent of metal ions. 0.16 0.14 0.12 0.10 " 0.08 d A Absorbance 0.06 - I II/ 0.04 a 0.02 b . K-n... ODD-4r T I """"*‘“* W ' I 400 500 600 700 800 wavelength (nm) 80 10. 11. REFERENCES Barrett, MP, Burchmore, RJ, Stich, A, Lazzari, J O, Frasch, AC, Cazzulo, JJ, Krishna, S. 2003. The trypanosomiases. Lancet 362: 1469-1480 Herwaldt, BL. 1999. Leishmaniasis. Lancet 354: 1191-1199 Gommers-Ampt, J H, Van Leeuwen, F, de Beer, AL, Vliegenthart, J F, Dizdaroglu, M, Kowalak, JA, Crain, PF, Borst, P. 1993. beta-D-glucosyl- hydroxymethyluracil: a novel modified base present in the DNA of the parasitic protozoan T. brucei. Cell 75: 1129-1136 van Leeuwen, F, Dirks—Mulder, A, Dirks, RW, Borst, P, Gibson, W. 1998. The modified DNA base beta—D-glucosyl-hydroxymethyluracil is not found in the tsetse fly stages of Trypanosoma brucei. Mol Biochem Parasitol 94: 127-130 van Leeuwen, F, Kiefi, R, Cross, M, Borst, P. 1998. Biosynthesis and function of the modified DNA base betavD-glucosyl-hydroxymethyluracil in Trypanosoma brucei. Mol Cell Biol 18: 5643-5651 van Leeuwen, F, Kiefi, R, Cross, M, Borst, P. 2000. Tandemly repeated DNA is a target for the partial replacement of thymine by beta-D-glucosyl- hydroxymethyluracil in Trypanosoma brucei. Mol Biochem Parasitol 109: 133- 145 van Leeuwen, F, Wijsman, ER, Kiefi, R, van der Mare], GA, van Boom, J H, Borst, P. 1997. Localization of the modified base J in telomeric VSG gene expression sites of Trypanosoma brucei. Genes Dev 11: 3232-3241 Cliffe, LJ, Siegel, TN, Marshall, M, Cross, GA, Sabatini, R. 2010. Two thymidine hydroxylases differentially regulate the formation of glucosylated DNA at regions flanking polymerase II polycistronic transcription units throughout the genome of Trypanosoma brucei. Nucleic Acids Res 38: 3923-3 935 Gommers-Ampt, J H, Teixeira, AJ, van de Werken, G, van Dijk, WJ, Borst, P. 1993. The identification of hydroxymethyluracil in DNA of Trypanosoma brucei. Nucleic Acids Res 21 : 2039-2043 Cross, M, Kiefi, R, Sabatini, R, Wilm, M, de Kort, M, van der Marel, GA, van Boom, J H, van Leeuwen, F, Borst, P. 1999. The modified base J is the target for a novel DNA-binding protein in kinetoplastid protozoans. EMBO J 18: 6573-6581 Yu, Z, Genest, PA, ter Riet, B, Sweeney, K, DiPaolo, C, Kieft, R, Christodoulou, E, Perrakis, A, Simmons, JM, Hausinger, RP, van Luenen, HG, Rigden, DJ, 81 12. l3. 14. 15. 16. 17. 18. 19. Sabatini, R, Borst, P. 2007. The protein that binds to DNA base J in trypanosomatids has features of a thymidine hydroxylase. Nucleic Acids Res 35 : 2107-2115 DiPaolo, C, Kiefi, R, Cross, M, Sabatini, R. 2005. Regulation of trypanosome DNA glycosylation by a SWIZ/SNFZ-like protein. Mol Cell 17: 441—451 Kieft, R, Brand, V, Ekanayake, DK, Sweeney, K, DiPaolo, C, Reznikoff, WS, Sabatini, R. 2007. J BP2, a SW12/SNF2-like protein, regulates de novo telomeric DNA glycosylation in bloodstream form Trypanosoma brucei. Mol Biochem Parasitol 156: 24-31 Cross, M, Kiefi, R, Sabatini, R, Dirks-Mulder, A, Chaves, I, Borst, P. 2002. J - binding protein increases the level and retention of the unusual base J in trypanosome DNA. Mol Microbiol 46: 37-47 Cliffe, LJ, Kieft, R, Southern, T, Birkeland, SR, Marshall, M, Sweeney, K, Sabatini, R. 2009. JBPl and J BP2 are two distinct thymidine hydroxylases involved in J biosynthesis in genomic DNA of African trypanosomes. Nucleic Acids Res 37: 1452-1462 Vainio, S, Genest, PA, ter Riet, B, van Luenen, H, Borst, P. 2009. Evidence that J -binding protein 2 is a thymidine hydroxylase catalyzing the first step in the biosynthesis of DNA base J. Mol Biochem Parasitol 164: 157-161 Genest, PA, ter Riet, B, Dumas, C, Papadopoulou, B, van Luenen, HG, Borst, P. 2005. F orrnation of linear inverted repeat amplicons following targeting of an essential gene in Leishmania. Nucleic Acids Res 33: 1699—1709 Klein, SM, Cohen, G, Cederbaum, A1. 1981. Production of formaldehyde during metabolism of dimethyl sulfoxide by hydroxyl radical generating systems. Biochemistry 20: 6006-6012 Ryle, MJ, Padmakumar, R, Hausinger, RP. 1999. Stopped-flow kinetic analysis of Escherichia coli taurine/or-ketoglutarate dioxygenase: interactions with (X.- ketoglutarate, taurine, and oxygen. Biochemistry 38: 15278-15286 82 CHAPTER 4 CHARACTERIZATION OF A T RYPANOSOMA BRUCE] ALKB HOMOLOG CAPABLE OF REPAIRING ALKYLATED DNA Portions of the introduction to this chapter were published in: Simmons, J M, Muller, TA, Hausinger, RP. 2008. Fe(II)/alpha-ketoglutarate hydroxylases involved in nucleobase, nucleoside, nucleotide, and chromatin metabolism. Dalton Trans. 5132-5142. 83 ABSTRACT T rypanosoma brucei encodes a protein homologous to AlkB of Escherichia coli, raising the possibility that trypanosomes catalyze oxidative repair of alkylation-damaged DNA. The gene encoding the T. brucei AlkB homolog (TbABH) was cloned and expressed in E. coli, the recombinant protein was purified and characterized, and the capacity of the protozoa] gene to complement alkB bacteria cells was examined. When anaerobic TbABH was incubated with Fe" and a-ketoglutarate (orKG), the sample produced a characteristic metal-to-ligand charge-transfer chromophore, confirming its membership in the Fell/aKG dioxygenase superfamily. TbABH was Shown to bind to DNA, exhibiting a clear preference for binding to alkylated DNA according to results derived by electrophoretic mobility shift assays. Further, TbABH partially complemented an alkB knockout in E. coli cells stressed with methylmethanesulfonate, confirming the assignment of TbABH as a functional AlkB protein in T. brucei. 84 4.1 INTRODUCTION Trypanosomes are eukaryotic extracellular parasites that cause diseases in various mammalian hosts (1). They are evolutionarily divergent organisms that branched early from the phylogenetic tree, before fungi, plants, or animals (2). Trypanosoma brucei, the causative agent of African Sleeping Sickness in humans, has a dense exterior protein coat that limits access to the parasite’s cellular membrane (3,4). In bloodstream form cells, the coat protein is expressed from a single allele and changes periodically in a population to evade detection by the host immune system; a process called antigenic variation (5). The genes encoding the repertoire of coat proteins are located primarily in the subtelomeric regions among large patches of repetitive sequence and are relocated by homologous recombination into a promoter-containing expression site as needed (6,7). The many repetitive regions in the T. brucei genome and the high frequency of recombination events lead to the need for efficient DNA repair and maintenance mechanisms. Components for most of the common DNA repair pathways have been identified in trypanosomes and some have been characterized. Proteins homologous to certain elements of the mammalian mismatch repair pathway (8), base excision repair (9), nucleotide excision repair (10) and homologous recombination (7) are present. One method of DNA repair not yet described in trypanosomes is direct repair which can be catalyzed by several DNA glycosylases, alkyl transferases, and hydroxylating enzymes in other systems. One such hydroxylase is AlkB, which has been best characterized in Escherichia coli. 85 E. coli alkB has been studied for its role in the adaptive response to alkylation damage since the 19808 (11,12). This gene was long known to confer resistance to certain methylating agents, and in 2002 the encoded protein was discovered to be a member of the F ell/a-ketoglutarate (aKG) dioxygenases (13,14). The enzyme catalyzes N-dealkylation of l-methyladenine (lmeA) and 3-methylcytosine (3meC) in DNA by the mechanism shown in Figure 4.1, creating an unstable intermediate that spontaneously releases an aldehyde to regenerate the native base. AlkB repairs the analogous lesions in RNA (15), including mRNA and tRNA (16). Furthermore, the protein dealkylates 1- methylguanine, 3-methylthymine and several etheno adducts of DNA (17-20). The crystal structure of E. coli AlkB in complex with F e", OLKG, and the tri- deoxynucleotide substrate T-lmeA-T (Fig. 4.2) reveals details of the metallocenteri active site and helps to clarify how the enzyme can accommodate its various substrates that include single-strand (ss)-DNA, double-strand (ds)—DNA, and various forms of RNA (21). Residues His 131, Asp 133, and His 187 bind the metal, aKG coordinates Fe" in a bidentate manner (via the C1 carboxylate and C2 keto groups), and a salt bridge from Arg 204 stabilizes the aKG C5 carboxylate. The open coordination site for oxygen binding does not point toward the substrate (a situation termed “off-line” geometry) (22); thus requiring a Shift in position of the C1 carboxylate prior to oxygen binding or a “ferryl flip” to occur after generating the F e'v—oxo intermediate (23). The trimer substrate is bent to extend the damaged base deep into the active-site pocket, making ten identified hydrogen bonds and many more van der Waals contacts with the protein. 86 Figure 4.1. Reactions of AlkB with lmeA and 3meC in DNA or RNA. NHZ NHz N + N N N2 02 C02 HCHO DNA or RNA aKG succinate DNA or RNA DNA or RNA NH2 OH (lib. 002 HCHO DNA or RNA GOKG 911990319 DNA 0, RNA DNA or RNA 87 Figure 4.2. Structure of AlkB (PDB 2fd8). The left panel depicts the overall fold of the enzyme, while the right panel highlights the active site residues. Fe" is an orange sphere, aKG has yellow carbon atoms, metal ligands have blue carbon atoms, a cofactor- stabilizing residue is shown with orange carbon atoms, and other postulated active site residues are indicated with white carbon atoms. The bound substrate is shown with magenta carbon atoms (abbreviated in the right panel). The nucleotide-binding lid (residues 45-90) is comprised of three B-strands (the left-most strand in the pair of 8- sheets and two short B-strands at the bottom) along with several loops (bottom left) lacking well-defined secondary structures. 88 The AlkB N-terminus is responsible for about half of the identified contacts with the substrate via a highly flexible nucleotide recognition domain or lid made up of 3 B- strands (residues 45-90). This protein region is flexible based upon the superimposed structures of the bound and nucleotide-free states, thus allowing movement and reorientation without losing critical contacts. Substrate specificity at the base level is provided deep in the pocket; a hydrogen bond between Arg 210 and the adenine moiety could also be made with cytosine, but not with the other two bases, explaining why AlkB prefers lmeA and 3meC as substrates. Finally, AlkB’s modest preference for SS- DNA/RN A substrates (15,24) is explained by the nucleotide polymer being oriented to extend into a hydrophobic pocket with stabilization provided by base stacking with nearby aromatic residues. By contrast, ds—DNA would need to unwind partially in order to allow the section of DNA containing the lesion to extend into the active site. Kinetic studies have examined AlkB activity using a wide range of substrates. A rate of 12 min'1 was reported for randomly methylated poly(dA) (25), or about 4 min’1 for two specifically mono-methylated oligonucleotides (26). Catalytic rates with the tri- deoxynucleotide T-lmeA-T used in the crystal structure studies were determined separately by two groups and found to be 7.4 and 4.5 min", respectively (21,25). A phosphate 5’ of the methylated base is critical for proper recognition based upon a 6-fold increase in the apparenth when it is missing (25), and this result is compatible with the crystal structure which shows both a hydrogen bond and a salt bridge involving this 5’ phosphate (2 1). In addition to the dealkylation reactions observed with the above substrates, AlkB is known to exhibit additional types of reactivity. The minimal substrate that could be 89 demethylated was identified as lme-dAMP (25), whereas the free nucleosides lmeA, lme-2’-dA, 3meC, and 3me-2’-dC were shown to stimulate oKG decomposition without undergoing demethylation (27). In the absence of nucleotide, but the presence of aKG and 02, AlkB hydroxylates its own Trp 178 (located near the active site, see Fig. 4.2) to generate a hydroxy-tryptophan side chain. A ligand—to-metal charge-transfer transition between this modified residue and the oxidized metal site results in a blue protein that absorbs maximally at 595 nm (28). This chapter focuses on an AlkB homolog of T. brucei and describes the DNA repair activity observed for this member of the non-heme F e"- and aKG-dependent hydroxylase family of enzymes in kinetoplastids. 90 4.2 MATERIALS AND METHODS 4.2.1 Gene identification and multiple sequence alignment The Basic Local Alignment Search Tool (29) was utilized to search the protein- encoding sequences of the T rypanosoma brucei brucei genome with the protein sequence of E. coli AlkB as the query, resulting in the identification of a sequence with the NCBI accession number XP_844196. The sequence of the identified trypanosomal AlkB homolog (TbABH) was aligned to representative group 1A (30) AlkB sequences of E. coli (NP_416716), Brucella abortus (ZP__05894130.1), Pseudomonas putida (AAN69003.1), Pseudomonas syringae (NP_792910.1), and the first three human AlkB homologs (ABHI, AAH25787.1; ABH2, Q6NS38.1; ABH3, Q96Q83.1) by using Clustal W (31) for analysis. Further, the TbABH sequence was analyzed to predict the subcellular location by several online servers including LOCTree (32), PSORTII (http://psort.hgc.jp/fonn2.html), SubLoc (33), and ESLPred (34). 4. 2.2 Cloning A 991 -bp DNA fragment containing TbABH was amplified by PCR using genomic T. b. brucei strain 427 DNA as a template with “forward” (5’-AGGATATA_C_Q ALQGAAGACC CCGTGC-3’) and “reverse” (5’-GA GCATCCTCGAG TTCGTTAAG GAACTCAC-3 ’) primers which introduce NcoI and Xhol restriction sites (underlined), and T aq polymerase master mix kit (Promega) which leaves a single 3 ’ adenine nucleotide overhang. The PCR product was treated with a PCR clean up kit (Qiagen, Inc.) and ligated into pGEM-T Easy (Promega) to create pGEM-TbABH. The pGEM- 91 TbABH plasmid was transformed into E. coli DHSa (Invitrogen), isolated from several transformants, and sequenced (Davis Sequencing). TbABH was excised from the pGEM- T backbone by N001 and Xhol restriction and ligated into pET28b (Novagen) which had been cut previously with the same enzymes, creating pET-TbABH and putting the coding sequence in frame with a sequence encoding a C-terminal 6-histidine tag. This plasmid was transformed into the expression strain E. coli BL21 (DE3). 4. 2.3 Protein production and purification E. coli BL21 (DE3) cells containing pET-TbABH encoding TbABH-His, (hereafter referred to simply as TbABH) were grown at 30 °C in lysogeny broth (LB) medium supplemented with 100 ug/mL kanamycin while shaking at ~l60 rpm to an optical density of 0.4 to 0.6 at 600 nm. Cultures were induced to overexpress the desired gene by addition of isopropyl-B-D-thiogalactopyranoside (IPTG) to 0.1 mM, and grown for an additional 4 h, then harvested at 4 °C by centrifugation at ~8,000 g for 8 min. The cell paste was either used immediately for protein purification or stored at -80 °C. In a typical purification, 3 mL of binding buffer (30 mM imidazole, 10 mM Tris, 150 mM NaCl, pH 7.9) was added per g of cell paste for resuspension. The protease inhibitor phenylmethylsulfonyl fluoride was added to 0.5 mM, cells were lysed by sonication (Branson Sonifier, 3 pulses of l min each, 3 W output power, duty cycle 50%, with cooling on ice), and the cell lysates were ultracentrifuged at 100,000 g for l h. Soluble cell extracts were loaded onto a Ni-bound nitrilotriacetic acid column (Qiagen) pre-equilibrated with binding buffer. The column was washed with binding buffer until the baseline was reestablished, and proteins were released with elution buffer (150 mM 92 imidazole, 10 mM Tris, 150 mM NaCl, pH 7.9). Fractions containing the purified proteins, as determined by denaturing sodium dodecyl sulfate—polyacrylamide gel electrophoresis (SDS-PAGE, 12% acrylamide) (35) and Coomassie staining, were pooled and dialyzed back into binding buffer by using 12-14 kDa molecular weight cutoff dialysis tubing (Fisher) at 4 °C overnight with stirring. The concentration of protein was determined by using its calculated molar absorptivity at 280 nm (45,350 M'1 cm”l according to the ExPasy protein parameters prediction server (http://ca.expasy.org/tools/protparam.html) (36). The protein was either used immediately for assays or was stored at 4 °C and discarded after two weeks or if precipitation developed. A typical purification yielded approximately 3 mg protein per g of cell paste. 4.2.4 Gel filtration chromatography To determine the native Size and Oligomeric state of the protein, purified TbABH was concentrated to 200 uM (7.4 mg/mL) of protomer in an Amicon centrifugal filter unit with a 10 kDa molecular weight cutoff and was chromatographed on a Superdex®75 size exclusion column preequilibrated with binding buffer. Retention times were compared to those of gel filtration standards (BioRad). The collected fractions also were examined by PAGE using a 3-12% Bis-Tris Blue native gradient gel (Invitrogen) to verify the major Oligomeric forms of TbABH. 93 4.2.5 Spectroscopy UV/visible spectra were obtained by using an HP 8453 spectrophotometer (Hewlett Packard) equipped with a circulating water bath and magnetic stirrer. TbABH was concentrated in an Amicon centrifugal filter unit with a 10 kDa cutoff to 265 uM of protomer in binding buffer. Stock solutions of 10 mM F e" and 50 mM aKG were prepared by adding the dry reagents to vials, subjecting them to repeated cycles of vacuum and argon, and adding anaerobic water by using a syringe. Protein was added to a 1-cm path length, 1.5 mL, quartz cuvette, the sample was made anaerobic by gentle vacuum/argon cycling, and it was scanned while stirring at 9 °C. Four equivalents of aKG (to 1 mM final concentration) were added and the sample was re-scanned, followed by titration with iron in 8 increments up to 500 uM with scanning after each addition. Data were corrected for dilution in Excel and plotted as difference curves with the absorbance from the protein alone set as the baseline. The difference absorbance of the feature observed at 530 nm (AA) was plotted versus the concentration of total added iron (14) to calculate a Kd for the binding of iron to the aKG-bound active Site using equation 1 (37). In this equation, AAmax is the predicted maximal absorbance change, [Ed is the total enzyme concentration, and n is the number of iron binding sites per protein molecule. Eq. 1 AA = AAma;((Kd+r.+an.n-(a 280 290 300 . .... .... ... . .. .. ....[....l....l. .l. ..l....l... E.coli#A1kB ~;T “ IQPLKAGFHP ------------------- LTID B.abortus_AlkB ‘1' ' ILPLKSGEHE ------------------- RLGP P.putida_A1kB «fi' “. PIKPGVHP ——————————————————— RLGE P.5yringae AlkB ifl‘fi ILPIKQABHP ------------------- LLGE T.brucei_ABH “ PRIMDDCPQYLCPQNETEEEEGEFYWREQMRH H.sapien ABHl VPRVLPNPEGEGLPHCLEAPLPAVL(l9)YLKT H. sapi en ABH2 P‘I‘NTHWYHSLPVRK ---------------- KVLA H. sapi en ABH3 ——————— RKKPPPEEAI’YTYVERVKI PL LLIMEGATQADWQHRVPKEY -------- HSRE O O O O 0.. O O O *0 102 biosynthetic pathway (44), phospholipase A1 (45) and isopropyl alcohol dehydrogenase (46). Despite TbABH’S greater similarity to the bacterial AlkB-like proteins than to homologs in eukaryotes, it is distinct in possessing a much longer amino terminus and inserts of 37, 1, and 19 residues compared to the E. coli protein. The largest insert extends from within the nucleotide-binding lid of AlkB, the extra amino acid at residue 193 corresponds to an unstructured region on the back face of the E. coli protein in the view of Figure 4.2, and the 19 amino acid insertion (residues 281 ~299) corresponds to an unstructured region at the very top of AlkB in the view shown. 4.3.2 General biochemical properties of TbABH To characterize this protein, TbABH was cloned from T. b. brucei genomic DNA, expressed recombinantly in E. coli, and the protein purified by affinity chromatography to about 90% purity as visualized via SDS-PAGE and Coomassie staining (data not Shown). Figure 4.4 shows that multiple forms of the protein were present as purified from E. coli. The predominant peak in the chromatogram (panel A) chromatographed as a monomer while the less prominent peak eluted as a dimer; however, native gel electrophoresis of the fractions was most consistent with both protein Species forming an equilibrium mixture of 37 kDa monomers, 74 kDa dimers, and 148 kDa tetramers. To confirm prOper folding of TbABH, the purified protein was examined for its ability to form a characteristic chromophore seen in other members of the F ell/aKG- dependent dioxygenases and associated with binding of the iron cofactor and the aKG cosubstrate (47). The protein was monitored by UV-visible spectroscopy under anaerobic conditions, using low temperature to maintain stability, while titrating in OLKG 103 Figure 4.4. Native size and Oligomeric state determination of TbABH. A. Chromatogram of TbABH analyzed by using a Superdex®75 column and compared with BioRad gel filtration standards (retention times of y-globulin, 158 kDa; ovalbumin, 44 kDa; and myoglobin, 17 kDa, are shown as inverted triangles above the chromatogram). B. Native polyacrylamide gradient gel Showing the Oligomeric states and native sizes of TbABH. The bar on the chromatogram indicates the fractions shown on the gel. A. V V V 0.20 1 1 0.151 ° 1 a 0.10 1 < 0.05.? 0.00 : ¢ ; LL j; 4 L r l L :L 4 L . _. 4 fiL _. J i 1 4o 45 50 - 55 so 65 70 75 Retention Time (mm) ' . .‘H " ‘ ' ‘ ‘ . _. v r c > _ rf‘Jr—‘g, -,. . ...“ . . . . 21' ,r-G'rw, I. 1“ 3* 10" Li). 4" ‘1 "‘ r- ticmfi‘fir‘: Atari .G‘E'i-{r’o ti... ,3; . «Jens-"rfinzvth‘ir: .k-ll 1 ‘T' '4 .. A“ .. ‘_ ' . ' , v . r _ . . . ' 31' ‘ < - . “3'57, 1“ _. . . ' ' -* l ‘ ‘ r e ' ‘ . V . . , r .t. . , 7' " ‘ ',.- 5 r r‘ ’r _ . ‘ - w . ‘ '( m‘r ,~' . v J‘rrzx .fi ~ . 4.1.‘A’Krf-AJIPIW. . Wigwm ,) I U.” 3.; 1,. ., , p. :r .. ... ~ ‘ . r, y . , . ' .. 1. ‘ », j" y. A ' . :- .. r r' 1‘ ..- All 11 A 1 <9 , ~n r- w. r _ v . .;.‘ -. ‘ ’ \ A. v. ‘1 . A, n. g l :1 4 V'. . v : a w ' i 7: a u L‘ 1 .,' o C‘. r“.. . A . s u .. _ ‘3 , ...:- . ,x _ - r. ‘ r a .. V r :'. ‘7 "i. {3. .4. . e"- .\ 104 and Fe". AS shown in Figure 4.5, anaerobic TbABH generated the diagnostic metal-to- ligand charge-transfer transition at 530 run when both metal and cofactor were present. The extinction coefficient for this feature was approximately 190 M’1 cm", consistent with values previously described for members of this enzyme family (37,48). The relative absorbance change at 530 nm was plotted versus the total concentration of iron titrated into the protein sample and fit using equation 1. This allowed for calculation of an Fe'I Kd of ~4 11M with 1.3 i 0.07 iron atoms binding per active Site. 4.3.3 DNA binding by T bABH To test the hypothesis that TbABH is a DNA repair protein, the ability of this protein to bind to potential DNA substrates was tested. TbABH was able to bind to supercoiled dS-plasmid DNA as evidenced by a shift in the electrophoretic mobility of the DNA band in an agarose gel (Fig. 4.6 A). The development of multiple TbABH-DNA complexes confounded efforts to quantify the thermodynamics of this interaction. Therefore, to obtain more quantitative binding data, EMSA assays were performed using 3‘ZP-labeled oligonucleotides that had been left untreated or had been subjected to methylation by MMS. While TbABH shifted all polynucleotides to some extent when tested by this method, a clear preference was observed for ds-DNA and especially for methylated dS-DNA over other compounds tested (Fi g 4.6 B). These band intensities were analyzed, normalized to the total 32P counts in each lane, and converted to nM DNA bound. These values were plotted against the concentration of added TbABH and the resulting curve was fit to a hyperbolic single Site binding equation (equation 2). This allowed the estimation of a K; of 7.1 i 1.9 uM (Fig 4.6 C). Although these assays 105 Figure 4.5. Spectroscopic evidence for binding of Fe‘1 and aKG by TbABH. A. The anaerobic UV/visible spectrum of TbABH (266 uM protomer in binding buffer) was examined for the sample as isolated (baseline), after adding 1 mM aKG, and while titrating in F ell with stirring at 9 °C. The (Fen-aKG-protein minus protein) difference spectra shown correspond to the addition of 0, 63 uM, 125 uM, 188 uM, 250 pM, 313 uM, 375 pM, 438 uM, and 500 uM metal ions. B. The intensity of the absorbance difference at 530 nm was examined as a function of added Fe". The data were fit to equation 1. 0.1 - 9’ 0.08 1 0.06 ~ 0.04 r 0.02 J A Absorbance 3' 0.07 '1 0.06 a ,_/’" 0.05 5 ./ A Absorbance fil T l I l o 100 200 300 400 500 Iron (11M) 106 Figure 4.6 EMSA studies of TbABH and various DNA substrates. A. Agarose gel depicting the shift in mobility of supercoiled pGEXS plasmid (7.5 nM) with increasing TbABH concentration (shown by the wedge, 0 to 20 nM). B. Native polyacrylamide gel Showing the Shift in mobility of 32P-labeled ds-oligonucleotide (7.5 nM; untreated on left and methylated on right) with increasing TbABH concentration (0 to 30 nM). C. Fit of data from B to equation 2. C. Untreated Methylated r ' ' 10 i5 i0 25 30 TbABH (pM) O (”I 107 contained 10 nM DNA, the process used for methylating the substrate typically exhibits poor efficiency (~10%); thus, the observation that saturation (Bmax) corresponded to 0.79 :1: 0.08 nM DNA indicates that nearly stoichiometric binding (close to 80%) of the methylated substrate was bound. In contrast to the above results Showing TbABH’s preference for binding to methylated ds-DNA, other oligonucleotides were bound with lower affinity. Very little binding was observed for ss-DNA with no differences noted between the untreated and the methylated substances (data not Shown). Furthermore, TbABH did not exhibit enhanced binding to DNA containing an abasic site; where as this binding has been noted for human ABHl (49). 4. 3.4 Examination of DNA repair by TbABH In an attempt to obtain direct evidence for the demethylation reaction expected of a functional AlkB, two in vitro assays were examined. The purified recombinant protein was used in a formaldehyde dehydrogenase-coupled reaction that allows for detection of released formaldehyde by monitoring an increase in absorbance at 340 nm due to the conversion of NAD+ to NADH as part of the coupled reaction. While there was an indication of turnover in one experiment using this assay, only background rates were found in follow-up studies where the protein-free control also yielded a slightly positive reaction. Efforts to demonstrate OLKG consumption by an OPDA assay also were inconclusive due to the low sensitivity of measuring substrate loss. As an alternative approach, TbABH was tested for the ability to complement an alkB knockout in an E. coli cell line. The alkB gene of BW251 13AalkB cells is replaced with a kanamycin resistance cassette resulting in 3.5-fold greater susceptibility to 108 methylation damage by MMS as evidenced by the decreased viable cell counts over time when exposed to 0.5% of this methylating agent (Fig. 4.7). The alkB knockout cell line was transformed with pUC-TbABHZ (which placed the trypanosomal gene in the correct reading frame while adding 7 extra residues to the N-terminus of the protein) and tested for MMS sensitivity by the Kirby-Bauer method. At 1% MMS, wild type cells Showed no inhibition of growth while the alkB knockout exhibited an average zone of inhibition (the distance between the edge of the paper disc and the edge of non-growth) of 4.0 i 0.7 mm. The complemented strain containing pUC-TbABH2 partially complemented the knockout exhibiting a zone of inhibition of 1.5 i 0.3 mm (Fig. 4.8). To extend the alkB complementation approach, the trypanosomal gene was cloned into several other vectors and the disc diffusion assays were repeated. The BW25113 cell line does not contain the DE3 lysogen and thus cannot support T7 dependent expression plasmids, So efforts focused on three alternative vectors which would use the natural methionine start of TbABH. However derivatives of pEXT20, pMMB67, and pWKS30 containing TbABH failed to produce soluble protein in this cell line so the complementation studies could not be performed. 109 Figure 4.7 Time course viability test with wild-type and AalkB BW251 13 cell lines stressed with 0.5% MMS. BW25113 wild type (filled symbols) or BW25113AalkB (open symbols) cultures were grown to exponential phase, stressed with 0.5% MMS, sampled in a time course, diluted and plated to determine viable cell count. The time of exposure to MMS required to reduce viability by half was 3.5 fold greater for the wild type than the knockout cultures. 1.E+09 1.E+08 I 1.9071 ‘ ...l 1.E+06 - é 1.E+05 J a 1.E+04 4 I U 1.E+03 i 1.E+02 . T I 1.E+01 l r 1.E+00 ‘Wfir—Q—m—Q 010 20 30 40 50 60 70 80 90100110120 Time of exposure (min) 110 Figure 4.8 Complementation of an E. coli alkB mutant with T bABH under alkylation stress. BW25113 cells containing either pUC18 or the derivative pUC-TbABH2 were stressed with 1% MMS according to the Kirby-Bauer method (39). Plates were incubated overnight at 37 °C and the resulting zones of inhibition were measured. Below is a representative experiment showing the wild type cells containing pUC18 (labeled WT), AalkB cells containing pUC18 (AalkB), and AalkB cells containing pUC-TbABH2 (TbABH). The zones of inhibition are shown by the black lines and white dashed circles. WT AalkB TbA BH lll 4.3.5 Conclusions Trypanosomes contain a full length ortholog of the E. coli AlkB which exhibits characteristics representative of the Fe" and aKG dependent dioxygenase superfamily of enzymes including the formation of a diagnostic metal-to-ligand charge-transfer chromophore when incubated with the metal and cofactor under anaerobic conditions. Further, while E. coli AlkB binds DNA only very weakly (50), TbABH forms a tight complex and exhibits a clear preference for alkylated ds-DNA. Although direct evidence for DNA repair was not obtained in vitro, an E. coli cell line deficient in alkB was complemented by the expression of TbABH upon alkylation stress conditions confirming its assignment as an AlkB protein. 112 10. 11. REFERENCES Barrett, MP, Burchmore, RJ, Stich, A, Lazzari, JO, Frasch, AC, Cazzulo, JJ, Krishna, S. 2003. The trypanosomiases. Lancet 362: 1469-1480 Pace, NR. 2009. Mapping the tree of life: progress and prospects. Microbial Mal Biol Rev 73: 565-576 Roditi, I, Furger, A, Ruepp, S, Schfirch, N, Biitikofer, P. 1998. Unravelling the procyclin coat of T rypanasama brucei. Mol Biochem Parasitol 91: 117-130 Field, MC, Carrington, M. 2009. The trypanosome flagellar pocket. Nat Rev Microbiol 7: 775-786 Cross, GA, Wirtz, LE, Navarro, M. 1998. Regulation of vsg expression site transcription and switching in T rypanasama brucei. Mal Biochem Parasitol 91: 77-91 Horn, D, Barry, JD. 2005. The central roles of telomeres and subtelomeres in antigenic variation in Afiican trypanosomes. Chromosome Res 13: 525-533 Glover, L, McCulloch, R, Horn, D. 2008. Sequence homology and microhomology dominate chromosomal double-strand break repair in African trypanosomes. Nucleic Acids Res 36: 2608-2618 Bell, J S, Harvey, TI, Sims, AM, McCulloch, R. 2004. Characterization of components of the mismatch repair machinery in T rypanasama brucei. Mol Microbiol 51: 159-173 Castillo-Acosta, VM, Estevez, AM, Vidal, AE, Ruiz-Perez, LM, Gonzalez- Pacanowska, D. 2008. Depletion of dimeric all-alpha dUTPase induces DNA strand breaks and impairs cell cycle progression in T rypanasama brucei. Int J Biochem Cell Biol 40: 2901-2913 Lee, J H, Nguyen, TN, Schimanski, B, Gunzl, A. 2007. Spliced leader RNA gene transcription in T rypanasama brucei requires transcription factor TFIIH. Eukaryot Cell 6: 641-649 Kataoka, H, Yamamoto, Y, Sekiguchi, M. 1983. A new gene (alkB) of Escherichia coli that controls sensitivity to methyl methane sulfonate. J. Bacteriol. 153: 1301-1307 113 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. Kondo, H, Nakabeppu, Y, Kataoka, H, Kuhara, S, Kawabata, S, Sekiguchi, M. 1986. Structure and expression of the alkB gene of Escherichia coli related to the repair of alkylated DNA. J. Biol. Chem. 261: 15772-15777 Trewick, SC, Henshaw, TF, Hausinger, RP, Lindahl, T, Sedgwick, B. 2002. Oxidative demethylation by Escherichia coli AlkB directly reverts DNA base damage. Nature 419: 174-178 Falnes, PO, J ohansen, RF, Seeberg, E. 2002. AlkB-mediated oxidative demethylation reverses DNA damage in Escherichia coli. Nature 419: 178-182 Aas, PA, Otterlei, M, Falnes, PO, Vagbo, CB, Skorpen, F, Akbari, M, Sundheim, O, Bjoras, M, Slupphaug, G, Seeberg, E, Krokan, HE. 2003. Human and bacterial oxidative demethylases repair alkylation damage in both RNA and DNA. Nature 421: 859-863 Ougland, R, Zhang, CM, Liiv, A, Johansen, RF, Seeberg, E, Hou, YM, Remme, J, Falnes, PO. 2004. AlkB restores the biological function of mRNA and tRNA inactivated by chemical methylation. Molec. Cell 16: 107-116 Koivisto, P, Robins, P, Lindahl, T, Sedgwick, B. 2004. Demethylation of 3- methylthyrnine in DNA by bacterial and human DNA dioxygenases. J. Biol. Chem. 279: 40470-40474 Delaney, J C, Essigrnann, JM. 2004. Mutagenesis, genotoxicity, and repair of 1- methyladenine, 3-alkylcytosines, l-methylguanine, and 3-methylthymine in alkB Escherichia coli. Proc. Natl. Acad. Sci. USA 101 Mishina, Y, Yang, C-G, He, C. 2005. Direct repair of the exocyclic DNA adduct 1,N6-ethenoadenine by the DNA repair AlkB proteins. J. Am. Chem. Soc. 127: 14594-14595 Delaney, J C, Smeester, L, Wong, C, Prick, LE, Taghizdeh, K, Wishnok, J S, Drennan, CL, Samson, LD, Essigmann, J M. 2005. AlkB reverses etheno DNA lesions caused by lipid oxidation in vitro and in vivo. Nature Struct. Molec. Biol. 12: 855-860 Yu, B, Edstrom, WC, Hamuro, Y, Weber, PC, Gibney, BR, Hunt, JF. 2006. Crystal structures of catalytic complexes of the oxidative DNA/RNA repair enzyme AlkB. Nature 439: 879-884 Hausinger, RP. 2004. Fe(II)/a-ketoglutarate-dependent hydroxylases and related enzymes. Crit. Rev. Biochem. Mol. Biol. 39: 21-68 114 29. 30. 31. 32. 33. 34. Clifton. 1.1. McDon ugh. -\IA. Siren-13.11:." i.- . Kc.-. .3..\ \. . GT: .322? \ Schofieid. CJ- 2006. S.. .rc..;. ' «1...: es 0:: :A‘\eg.;;:-=.:3:e cxyg-srsscs 331: mixes . double-stranded B-helix fold pretext. j are: 3‘;-(.35., 2.1 ewe-‘3 Falnes- PO.Bior.1s..\1-A.1s PA.S'-... nine-1:1: .0. \ee:\.;;-': 2.1-‘4 S..':~."r=:e specificities of bacterial and ..u. 1.111 A.‘ 2.3. are: -‘ .ns \ $3-1: 3-3. 3 - 433 3.461 Koivisto. P. Duncan. T. Lindahl- T. Sedcui k. 8 33(3- -\{:::::t:3.I :tteftyisrai substrate and extended substrate range or r:- cci: r:.-F:.-.: 2; A33 , r.‘:c:::. .r .- meth} ladenine-DNA dioxgensse. J. 3.93. L 5:. w:- ‘ ’ Roy. TW, Bhagwat. ..AS 200". Kinetic stu res ot 3.5.2. r: .‘5:.‘.: .‘z‘I: .‘\:‘.\R Lzszztc .1 men fluorescence-based assm for D.\' A deme t-‘t_\i.1:: on -\ .... . .. t. ...- :- Res 3 e147 Welford. RWD. Schlemminger. I. -\lc.\'eill. LA. liemtson. KS. Schofieid. L11. 2003. The selectixity and inhibition of AlkB. J. Biol. L‘in'»: 2‘.\‘: 1023310201 Henshaw. TF. Feig. M. Hausinger. RP. 2004. Aberrant .icnxity ot‘thc DNA remit enzyme AlkB. J. Inorg. Biochem. 98: 856-801 Altschul, SF, Madden, TL, Schaffer. AA. Zhang. J H. Zhang. 7.. Miller. \V. Lipman, DJ. 1997. Gapped BLAST and PSI-BL AST: A new generation ofprotein database search programs. Nucleic Acids Res 25 : 33 89 - 3402 van den Born, E, Bekkelund, A, Moen. MN. Omelchenko. .\1\'. Klimt-gland. A. F alnes, PO. 2009. Bioinformatics and functional analysis define. four distinct groups of AlkB DNA-dioxygenases in bacteria. .\'uclcic.1ci.is Res 3": "124-‘136 Thompson, JD, Higgins, DG, Gibson. TJ. 1993. CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. .\‘ucl. Acids Res. 22: 4673-4680 Nair, R, Rost, B. 2005. Mimicking Cellular Sorting Improves Prediction of Subcellular Localization. J. Mol. Biol. 348: 85-100 Hua, S, Sun, Z. 2010. Support vector machine approach for protein subcellular localization prediction. Bioinformatics: in press Bhasin, M, Raghava, GP. 2004. ESLpred: SVM-based method for subcellular localization of eukaryotic proteins using dipeptide composition and PSI-BLAST. Nucleic Acids Res 32: W414-4 l 9 115 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. Laemmli, UK. 1970. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature (London) 227: 680-685 Hulo, N, Bairoch, A, Bulliard, V, Cerutti, L, De Castro, E, Langendijk-Genevaux, PS, Pagni, M, Sigrist, CJ. 2006. The PROSITE database. Nucleic Acids Res 34: D227-230 Ryle, MJ, Padmakumar, R, Hausinger, RP. 1999. StOpped—flow kinetic analysis of Escherichia coli taurine/(x-ketoglutarate dioxygenase: interactions with or- ketoglutarate, taurine, and oxygen. Biochemistry 38: 15278-15286 Datsenko, KA, Wanner, BL. 2000. One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640- 6645 Bauer, AW, Kirby, WM, Sherris, J C, Turck, M. 1966. Antibiotic susceptibility testing by a standardized single disk method. Am J Clin Pathol 45 : 493-496 Dykxhoom, DM, St. Pierre, R, Linn, T. 1996. A set of compatible tac promoter expression vectors. Gene 177: 133-136 Wang, RF, Kushner, SR. 1991. Construction of versatile low-copy-number vectors for cloning, sequencing and gene expression in Escherichia coli. Gene 100: 195-199 Lizcano, JM, Unzeta, M, Tipton, KF. 2000. A Spectrophotometric method for determining the oxidative deamination of methylamine by the amine oxidases. Anal Biochem 286: 75-79 Hayashi, T, Tsuchiya, H, Todoriki, H, Naruse, H. 1982. High—performance liquid chromatographic determination of alpha-keto acids in human urine and plasma. Anal. Biochem. 122: 173-179 Koreny, L, Lukes, J, Obomik, M. 2010. Evolution of the haem synthetic pathway in kinetoplastid flagellates: an essential pathway that is not essential after all? Int J Parasitol 40: 149-156 Richmond, GS, Smith, TK. 2007. A novel phospholipase from Trypanosoma brucei. Mol Microbiol 63: 1078-1095 Molinas, SM, Altabe, SG, Opperdoes, FR, Rider, MH, Michels, PA, Uttaro, AD. 2003. The multifunctional isopropyl alcohol dehydrogenase of Phytomanas Sp. could be the result of a horizontal gene transfer from a bacterium to the trypanosomatid lineage. J Biol Chem 278: 36169-36175 116 47. 48. 49. 50. Ryle, MJ, Padmakumar, R, Hausinger, RP. 1999. Stopped—flow kinetic analysis of Escherichia coli taurine/or-ketoglutarate dioxygenase: interactions with or- ketoglutarate, taurine, and oxygen. Biochemistry 38 : 15278-15286 Hegg, EL, Whiting, AK, Saari, RE, McCracken, J, Hausinger, RP, Que, L, Jr. 1999. Herbicide-degrading or—keto acid-dependent enzyme deA: metal coordination environment and mechanistic insights. Biochemistry 38: 16714- 16726 Muller, TA, Meek, K, Hausinger, RP. 2010. Human AlkB homologue l (ABHI) exhibits DNA lyase activity at abasic sites. DNA Repair (Amst) 9: 58-65 Dinglay, S, Trewick, SC, Lindahl, T, Sedgwick, B. 2000. Defective processing of methylated single-stranded DNA by E. coli alkB mutants. Genes Develop. 14: 2097-21 05 117 CHAPTER 5 PERSPECTIVES 118 5.1 INTRODUCTION Enzymes of the non-heme iron and a-ketoglutarate (dKG) dependent dioxygenase superfamily are ubiquitous in nature and versatile in catalytic activity. Members have been identified in nearly every branch of life and catalyze oxidative reactions ranging from hydroxylations to desaturations, ring formation, or ring expansion to name a few (1). The defining features of this family are the double-stranded B-helix core fold and the requirement for a ferrous ion at the active site. Most members oxidatively decarboxylate aKG to form C02 and succinate while creating a highly reactive ferryl-oxo species that then carries out the primary oxidative reaction (1). One branch of phylogeny for which no direct activity for this enzyme family has been Shown is the kinetoplastid flagellates including trypanosomes. Trypanosomes are the source of medical and economic challenges in their endemic areas, causing disease in humans and livestock (2). These parasites have unique anatomy and physiology (see Chapter 1) and complex relationships with their mammalian and insect hosts. They undergo drastic physiological changes during their life cycle and persist in mammalian infection through host immune system evasion involving a long studied, yet poorly understood, process called antigenic variation (3). For all of these reasons, trypanosomes are interesting topics of study. The experiments described in this thesis sought to identify and characterize several putative F e" and aKG- dependent hydroxylases from trypanosomes in order to increase understanding of these parasites and expand knowledge of this remarkable enzyme family. 119 5.2 SUMMARY OF RESULTS 5.2.1 T LP5 and TLP7 are unlikely to participate in pyrimidine salvage Two T rypanosoma brucei genes were identified based on sequence similarity to thymine 7-hydroxylase from Rhodatorula glutinis and termed T LP5 and T LP7 . The genes were cloned into Escherichia coli and the recombinant proteins expressed, purified by column chromatography, and characterized. Both proteins were soluble as expressed and existed predominantly as monomers in solution. In addition, proper folding and binding of the Fe" and aKG cosubstrates were verified by observation of a characteristic metal-to-ligand charge-transfer (MLCT) feature by UV-visible spectroscopy. However, neither protein bound to the tested nucleic acids or exhibited enzymatic activity as measured by oxygen consumption or succinate production. Steady state mRNA levels of TLP5 and TLP7 were tested by quantitative reverse transcriptase PCR and were found to be several fold higher in the bloodstream form (BF) of the parasite over the procyclic form (PF) for both genes, suggesting that the RNAs are stabilized in this life stage. Epitope tagging of the chromosomal copy of TLP5 was attempted in order to be able to test the subcellular localization and expression levels of the protein, but this effort failed on six separate attempts. Protein production was verified in trypanosome cell extracts by western blotting with a polyclonal antibody raised against TLP5. The antibodies were Shown to be cross reactive against TLP7, and both proteins were identified in extracts of both BF and PF parasites. Each cell form also contained a cross reactive band of higher molecular weight suggesting possible posttranslational modification of these proteins in the trypanosome 120 cell. Pull-down assays were performed to identify possible protein partners of TLP5 and TLP7 in the cell by passing trypanosome cell extracts over column-bound recombinant TLP5 or TLP7 and examining the eluant on a polyacrylamide gel, but no interacting proteins were observed. In efforts to detect enzymatic activity from these proteins in their natural states, cell extracts were incubated with possible substrates along with cosubstrates and analyzed for product formation by mass spectrometry, but no product formation was observed. In addition, a homology model of TLP5 was created and used in docking studies in an effort to generate hypotheses about the enzyme specificity; although several heterocyclic ring structures that resembled nucleotides were identified as the best ligands, no convincing potential substrates were identified. 5.2.2 JBP1 is inactive in its recombinant farm Trypanosomes contain a unique base in their nuclear DNA, B-D- glucosylhydroxymethyldeoxyuracil or base J, which replaces up to 1% of thymine bases (4). Extensive evidence has been obtained that JBP1, a protein Shown to bind specifically to J -containing DNA, is responsible along with JBP2 for the introduction of base I into chromatin in viva, though no in vitro activity has been reported (5,6). Recombinant J BP1 , purified from E. coli, was shown to fold properly and bind Fe“ and uKG via observation of the MLCT feature; however, no activity was observed when it was tested for in vitro activity by monitoring succinate production via high performance liquid chromatography or oxygen consumption via oxygen electrode. While it has been established that JBP1 and J BP2 are critical to the production of base J in trypanosome 121 DNA, the in vitro systems tested were insufficient to observe any activity of these enzymes. 5.2.3 T bABH is a DNA-binding protein homologous to E. coli AlkB TbABH is an ortholog of E. coli alkB, and was predicted to be a nuclear, DNA- binding protein. Its peptide sequence of the catalytic core aligned well with bacterial AlkBs of group 1A (7) including all metal-coordinating residues and several other highly conserved amino acids known to make contact with the nucleic acid substrate in E. coli AlkB (8). TbABH was cloned into E. coli and the protein expressed and purified. TbABH was found to be soluble and in equilibrium between monomer, dimer, and tetramer forms. Proper folding was verified by the formation of the MLCT feature when anaerobic sample was incubated with F e" and orKG, and titration studies provided a ferrous ion Kd of approximately 4 pM. TbABH bound to supercoiled plasmid DNA, as evidenced by electrophoretic mobility shift (EMSA) assays in agarose gels, and to linear alkylated oligonucleotides, as evidenced by EMSA assays in polyacrylamide gels. The latter EMSAS, performed with varying amounts of TbABH, allowed determination of a methylated DNA Kd of 7 pM with near stoichiometric binding to the alkylated portion of the substrate, given the typical low alkylation efficiency. The specific shift observed with methylated DNA was not observed with untreated DNA, and was not due to the creation of abasic Sites indicating that TbABH binds preferentially to methylation-damaged DNA. TbABH was tested for in vitro activity by monitoring oxygen consumption via oxygen electrode, aKG consumption via the OPDA assay, and formaldehyde production 122 via the FDH-coupled assay. Unfortunately, none of the approaches provided conclusive results. TbABH was also tested for the ability to complement an alkB knockout in an E. coli cell line stressed with methylmethane sulfonate. The complemented cell line containing pUC-TbABHZ was approximately two fold more resistant to MMS growth inhibition than the knockout cell line containing underivatized pUC18. This complementation along with the DNA binding confirms the assignment of TbABH as a functional AlkB-like protein in T. brucei expanding the knowledge of DNA repair mechanisms in this organism. 123 5.3 CONCLUSIONS AND FUTURE DIRECTIONS Trypanosomes contain several representatives of the F e" and aKG-dependent dioxygenase superfamily, and while all the proteins investigated in this study were expressed well in E. coli and yielded soluble protein that was properly folded and capable of binding iron and aKG, none of the in vitro assays detected enzymatic activity. It may be that trypanosomes contain some stabilizing or activating factor that is missing in our assay conditions. Also, it is possible that these proteins require some type of posttranslational modification for activity, consistent with the results obtained in western blots of trypanosome cell extracts with anti-TLP5 antibodies where prominent cross- reactive bands were observed. On the other hand, I was unable to detect thymine 7- hydroxylase activity using a mass Spectrometry assay after incubating thymine with trypanosome cell extracts, perhaps because the protein levels were too low to allow detectable product formation or the substrates tested may have been incorrect. In order to address these problems, one could attempt to purify the proteins directly from trypanosomes, bypassing the recombinant system entirely, so as to maintain the native form of the protein as translated and modified in the cell. This would pose a new problem of yield, however, Since trypanosomes do not grow to a high density and the proteins of interest would not be over expressed. Many culture flasks would be needed to obtain the number of cells required to achieve a comparable amount of protein purified from 1 L of an E. coli culture. Another approach would be to use trypanosome cell extracts as an additive in in vitro assays with recombinant protein. This could possibly provide a needed stabilizing 124 or activating factor, however then the reactions become less defined and any observed activity cannot be directly attributed to the recombinant protein. It would provide a starting point though, and the in vitro assay could be refined and optimized once it was known that something in the cell extract could render the protein active. Finally, one could look into other potential enzyme family members in these amazing parasites. The genome of T rypanasama brucei contains many orthologs of F e" and aKG-dependent dioxygenases and studying several of these could lead to further understanding of trypanosome biology and new therapeutic targets. As one example, locus Tb09.211.3730 (XP_827514) encodes an ortholog of the oxygen-sensing asparaginyl hydroxylase (FIH) of humans (25% identity over 343 amino acids). Whereas the human enzyme is 349 amino acids, the trypanosomal protein is 1145 amino acids in length and consistent with a multi-domain architecture. The role of FIH in humans is oxygen-sensing via hydroxylation of the hypoxia inducible factor (HIF) (9), so it is reasonable to suspect a similar function of the protozoal protein. In this regard, the bloodstream form of the parasite strictly uses glycolysis for its energy needs while the intra-insect or procyclic form uses oxidative phosphorylation; thus, oxygen sensing plays a critical role in protozoan cellular regulation. It would be interesting to investigate this trypanosomal F IH homolog and determine if it has asparaginyl hydroxylase activity by testing several Asn-containing possible substrates by the methods used previously or a more sensitive approach utilizing a radiolabeled cosubstrate such as 1-[ l4C]-OLKG. Another example of potentially interesting proteins to study is a group of highly related sequences arranged in a cluster. Trypanosomes contain large gene arrays within polycistronic units Spanning megabase regions of their chromosomes (10). Some genes 125 whose protein products are needed in abundance, such as tubulin, are known to appear in groups of tandem copies in order to obtain more of the primary transcript. Therefore, the presence of five tandemly repeated genes in T. brucei (Tb927.2.6180, 6210, 2130, 6270, and 6310) that encode nearly identical proteins of 319 amino acid residues and are likely to be Fell/dKG dioxygenases based on sequence comparisons is interesting because it implies that the protein product of these genes is important for the cell and needed in abundance. While there is no close homolog to these sequences to suggest a function for them, it would be interesting to pursue a screening approach to identify possible substrates. This could be accomplished using l4C-labeled aKG that would release l4C02 upon its oxidative decomposition. Any positive results from those assays would have to be followed up with substrate specific assays to confirm its assignment as a substrate. Trypanosomes are of great interest because they present many health threats to society and their unique biology offers exceptional challenges to scientists. They will continue to be studied even after effective and affordable cures of their diseases are available because they offer an interesting window into phylogenetic relationships represented in the tree of life. Having branched so early from the eukaryotic lineage, they are very divergent from the more widely studied model organisms for eukaryotic systems, such as yeast (11). They harbor several genes that appear to have been obtained by gene transfer from bacteria and others that seem to be distantly related to plant sequences based on alignments. The F e"/aKG dioxygenases represent an important enzyme family responsible for many reactions in the cell including several related oxygen sensing functions. Sensing oxygen levels is critical to any organism and especially for trypanosomes as they undergo changes associated with their life cycle which includes 126 forms exhibiting very different oxygen utilization. I am confident that in time, members of this enzyme family will be found to be involved in the regulation of these processes, expanding their roles across nature. 127 10. 11. REFERENCES Hausinger, RP. 2004. F e(II)/0t-ketoglutarate-dependent hydroxylases and related enzymes. Crit. Rev. Biochem. Mol. Biol. 39: 21-68 Barrett, MP, Burchmore, RJ, Stich, A, Lazzari, JO, Frasch, AC, Cazzulo, JJ, Krishna, S. 2003. The trypanosomiases. Lancet 362: 1469-1480 Morrison, LJ, Marcello, L, McCulloch, R. 2009. Antigenic variation in the Afiican trypanosome: molecular mechanisms and phenotypic complexity. Cell Microbiol 11: 1724-1734 Borst, P, Sabatini, R. 2008. Base J: discovery, biosynthesis, and possible functions. Annu Rev Microbiol 62: 235-251 Cliffe, LJ, Kieft, R, Southern, T, Birkeland, SR, Marshall, M, Sweeney, K, Sabatini, R. 2009. JBP1 and J BP2 are two distinct thymidine hydroxylases involved in J biosynthesis in genomic DNA of African trypanosomes. Nucleic Acids Res Yu, Z, Genest, PA, ter Riet, B, Sweeney, K, DiPaolo, C, Kieft, R, Christodoulou, E, Perrakis, A, Simmons, JM, Hausinger, RP, van Luenen, HG, Rigden, DJ, Sabatini, R, Borst, P. 2007. The protein that binds to DNA base J in trypanosomatids has features of a thymidine hydroxylase. Nucleic Acids Res 35: 2107-2115 van den Born, E, Bekkelund, A, Moen, MN, Omelchenko, MV, Klungland, A, F alnes, PO. 2009. Bioinformatics and functional analysis define four distinct groups of AlkB DNA-dioxygenases in bacteria. Nucleic Acids Res 37: 7124-7136 Yu, B, Edstrom, WC, Hamuro, Y, Weber, PC, Gibney, BR, Hunt, JF. 2006. Crystal structures of catalytic complexes of the oxidative DNA/RNA repair enzyme AlkB. Nature 439: 879-884 Webb, JD, Coleman, ML, Pugh, CW. 2009. Hypoxia, hypoxia-inducible factors (HIF), HIF hydroxylases and oxygen sensing. Cell Mal Life Sci 66: 3539-3554 Myler, PJ, Audleman, L, deVos, T, Hixson, G, Kiser, P, Lemley, C, Magness, C, Rickel, E, Sisk, E, Sunkin, S, Swartzell, S, Westlake, T, Bastien, P, Fu, G, Ivens, A, Stuart, K. 1999. Leishmania major F riedlin chromosome 1 has an unusual distribution of protein-coding genes. Proc Natl Acad Sci U S A 96: 2902-2906 Pace, NR. 2009. Mapping the tree of life: progress and prospects. Microbial Mol Biol Rev 73: 565-576 128