Cell Cycle Effects Associated with Human Adenovirus E4orf4 Protein-Induced Tumour Cell Death Neera Sriskandarajah BSc Biochemistry Department of Biochemistry and Rosalind and Morris Goodman Cancer Research Centre McGill University Montreal, Quebec, Canada September 2014 A thesis submitted to McGill University in partial fulfillment of the requirements of the degree of Master of Science © Neera Sriskandarajah, 2014 Neera Sriskandarajah 2 Table of Contents Abstract…………………………………………………………………………………... 5 Abrégé………………..…………………………………………………….……………...6 Acknowledgements ………………………..……………………………………………...7 Preface …………………………..……………………………………………………….. 8 Contribution of Co-Authors ………………..…………………………………………......8 List of Figures……………………………………..………………………………………9 List of Tables……………………………….…………………………………………......9 List of Abbreviations………………………..………………………………………..10-11 Original Contributions to Knowledge……………………………..……………………..12 Publications………………………………………………………………………………12 I. Introduction..................................................................................................................13 1. Human Adenoviruses and their Classification……...……………………………..13-14 1.1. Adenovirus structure…...……...………………………………………...14-15 1.1.2 Adenovirus Infection……...………….…………………...……16-17 1.2. Adenovirus genome and gene expression.……….……………………...18-20 1.2.1. The Adenovirus E1A gene.…….…...………………………....21-22 1.2.2. The E4 transcription unit.……...………………………………….23 1.2.3. Late gene expression. ……...……………………………………...24 1.2.4. Host Shut-off. ……...……………………………………….……..25 1.2.5. Cytopathic Effect (CPE).….…………...………………………….25 2. The Adenovirus E4orf4 protein.……...………………………………….……………26 2.1. The structure of E4orf4…..……….………………………………..…….26-27 2.1.2. The function of E4orf4. ……...……………………………….…...27 2.1.2.1. E4orf4 interacting partners.……...……………….……..28 2.1.2.1.1 Protein Phosphatase 2A………………………..28 2.1.2.1.1.2. PP2A the Cell Cycle……………….………..29 2.1.2.1.1.3. E4orf4-PP2A Interaction……………….. 29-31 2.1.2.2 E4orf4 and splicing..……...……………………………...31 2.2. E4orf4 mediated cell killing and the Cell Cycle………………….....………..….32-36 3. Experimental Rationale ………………………..……………………………………...37 II. Materials and Methods.........................................................................................38-41 III. Results.......................................................................................................................42 3.1. G0G1 Synchronization................................................................................42-47 3.2. G0G1 Cell Synchronization and Release....................................................48-51 3.3. Time Lapse Microscopy of H1299 cells expressing E4orf4 ……...…….52-60 3.4. E4orf4 induces cell death at both G1 diploid and G1 tetraploid states….. 61-64 Neera Sriskandarajah 3 IV. Discussion...................................................................................................................65 4.1. Development of a novel method of G0/G1 synchronization in p53-/- H1299 Human Lung Carcinoma cells……………………………………………………65 4.2. Cellular morphology of E4orf4 expressing cells………………………...65-66 4.3. Cell fate induced by E4orf4……………………………………………...66-67 4.4.Concluding Remarks…………………………….…………………………...69 V. Supplementary Videos…………………..…………………………………………..69 VI. References............................................................................................................ 70-78 Neera Sriskandarajah 4 Abstract The adenovirus E4orf4 protein induces cell death independently of p53 in human cancer cells but not normal primary cells through a mechanism that remains to be elucidated. Previous studies have suggested that E4orf4 induces an arrest or a delay in mitosis, a fate that is heavily dependent on E4orf4’s interaction with the Bα regulatory subunit of protein phosphatase 2A. In this study, a protocol using 0.01% FBS media was developed to synchronize human lung carcinoma (H1299) cells in G0/G1. Time-lapse microscopy was performed using this G0/G1 synchronization as well as S phase arrest and release in mCherry-H2B H1299 cells to look at both morphological changes by phase contrast and cell cycle progression by immunofluorescence. It was found that cells expressing E4orf4 display an overall increased mobility, rounding up, and demonstrate a slow transit through mitosis, a delay and often failure in cytokinesis, and those cells that survive take a prolonged period of time to re-enter another round of the cell cycle. In fact, when E4orf4 was expressed in G0/G1 arrested cells, most cells failed to initiate DNA synthesis, as shown in parallel flow cytometry studies by our group (Cabon, Sriskandarajah et al, 2013). Studies in this thesis using time-lapse microscopy confirmed that in such cells no further nuclear division was evident. However, when E4orf4 was expressed in cells blocked in S phase and then released, cells completed DNA synthesis but often remained tetraploid and by time-lapse microscopy, demonstrated difficulties completing cytokinesis successfully. Studies in this thesis also addressed the issue of whether the cell death induced by E4orf4 occurs in G1 arrested diploid or tetraploid cells. Cell sorting experiments followed by flow cytometry revealed that E4orf4 induced cell death occurred in both tetraploid and diploid cells. Thus my work has confirmed that E4orf4 toxicity is characterized by difficulty in transiting though mitosis to successful completion of cytokinesis. Neera Sriskandarajah 5 Abrégé Il a été démontré que la protéine adénovirale E4orf4 induit la mort cellulaire indépendamment de p53 dans des cellules cancéreuses humaines mais non dans des cellules primaires normales par un mécanisme d'action qui cependant reste largement inconnu. Des études ont suggéré que l'intéraction entre E4orf4 et la sousunité régulatrice Bα de la protéine phosphatase 2A résulte en l'induction d'un arrêt ou un délai dans la progression mitotique. Dans cette thèse, un protocole de synchronisation de cellules de carcinome pulmonaire humaines (H1299, en culture à 0.01%FBS) ) en G0/G1 a été développé. Grâce à cette synchronisation combinée à un arrêt puis libération de la phase S du cycle cellulaire de cellules H1299 exprimant un mCherry-H2B, une étude microscopique par immunofluorescence ainsi que par contraste de phase a permis d'investiguer les changements morphologiques ainsi que la progression à travers le cycle cellulaire. Il a été constaté que les cellules exprimant E4orf4 affichaient une mobilité globale accrue ansi qu'une morphologie arrondie. Ces cellules ont également démontré une progression mitotique plus lente, un retard et même souvent l'échec dans la cytokinèse. De surcroît, les cellules qui survivent prennent une période de temps plus longue pour amorcer un autre cycle cellulaire. En effet, notre groupe a démontré par des études de cytométrie par flux (Cabon, Sriskandarajah et al, 2013) que lorsque E4orf4 est exprimé dans les cellules en arrêt G0/G1, la majorité de ces cellules n'initient pas de synthèse d'ADN, ce qui a été également confirmé par les études dans cette thèse. Cependant, lorsque E4orf4 est exprimé dans des cellules bloquées en phase S puis libérées, ces cellules complètent la synthèse d'ADN, mais demeurent souvent tétraploïdes et ont des difficultées à compléter la cytokinèse. Les études par cytométrie de flux de cette thèse ont également déterminé que la mort cellulaire induite par E4orf4 se produit dans des cellules tétraploïdes ainsi que diploïdes. Ainsi, mon travail a confirmé que la toxicité de E4orf4 se caractérise par une difficulté à transiter de la mitose à la réussite de la cytokinèse. Neera Sriskandarajah 6 Acknowledgements This thesis summarizes my research work carried out in the laboratories of Professors Philip E. Branton and Jose Teodoro. My studies here at McGill University would not be possible without many people and as such I would like to thank each and every one of them. To Dr. Phil Branton who saw something in me when I was a second year Biochemistry student during my Undergraduate degree. Thank you for being the kind person that you are and an extraordinarily passionate and patient scientist. Thank you for lighting this passion for research in me, back when I was not sure where my path in academics would be. I am so very grateful to have been given the chance be your very last graduate student in your long and successful research career. To Dr. Jose Teodoro who equally gave me the opportunity to pursue my interest in biochemical research. Thank you for your attentiveness, and innovative ideas. I also want to thank the present and past members of Phil and Joe’s laboratories, Dr. Paola Blanchette, Dr. Theresa ChiYing Cheng, Dr. Zarina J. D'Costa, Dr. Timra Gilson, Dr. Melissa Z. Mui, Dr. Peter Wimmer, Dr. Dennis Takayesu, Frédéric Dallaire, Thomas Kucharski, David Sharon, Jieyi Yang, Dr. Wissal El-Assaad, Isabelle Gamache, Amro Mohammed, Mohamed Moustafa, Linda Smolders, Dr. Rachid Zagani and YiQing Lü for your all your support and smiles. I would like to give a special thanks to Thomas Kucharski who gave me a head start in a lot of the techniques that I have learned over the past two years but also for the friendly and scientific conversations. Thank you to Ken MacDonald and Dr. Abba Malina for their help teaching me world of flow cytometry. Thank you to Meena and Sophia for the early morning conversations and all the smiles, your work is always appreciated. I also want to thank my Research Advisory Committee, Dr. Gordon Shore, Dr. Anne Roulston and Dr. Jacques Archambault for all the valuable advice, suggestions and critical questions that can only make me improve as a scientist. Specifically, I would like to thank Dr. Archambault for kindly accepting me as a student in his lab for my PhD studies this Fall. My thanks also go to each of my friends for all our conversations both at work and our ventures outside. I would like to give a special thanks to Dr. Sami K. Boualia for giving me his constant loving support, as well as his lending hand in partial reviewing of the manuscript and abstract translation. To my great-uncle Dr. Rajendram Saddanathar who was such an inspiration growing up, and continues to be one of the greatest role models in my life. I remember you every day I’m at the bench. Most importantly, I would like to thank my wonderful parents for teaching me how important education is, and never to give up on your dreams. Thank you for letting me grow into the person I am today, and for loving me and supporting me in every way that you do. Neera Sriskandarajah 7 Preface In compliance with the Guidelines concerning Thesis Preparation of the Faculty of Graduate and Postdoctoral Studies of McGill University; Section 3.3 of this thesis contains parts of a paper that will eventually be submitted for publications; Section 3.4 of this thesis represents parts of the text of a paper published in Journal of Virology 2013 Dec; 87(24): 13168-78. Contribution of Co-Authors All of the figures presented in this thesis were generated by the author, except the following: Figure I-1: from Nemerow, Pache et al. 2009. Figure I-5/6: from Cabon, Sriskandarajah et al. 2013. Neera Sriskandarajah 8 List of Figures Figure I-1: Schematic diagram of the HAdV structure…………………………………15 Figure I-2: Schematic Adenovirus infection……………………………………………17 Figure I-3: Transcriptional map and gene products of adenovirus type 5 (Ad5)…….…20 Figure I-4: APC and the cell cycle…………………………………………………...…33 Figure I-5: Cell cycle analysis by flow cytometry post-synchronization in S phase...…35 Figure I-6: Cell cycle analysis by flow cytometry post-synchronization in G0/G1……..36 Figure III-1: G0/G1 Synchronization by 1% (low) serum conditions………………...…42 Figure III-2: G0/G1 Synchronization by Nutrient depleted serum starvation………...…43 Figure III-3: G0/G1 Synchronization by Amino Acid (aa) free media+/- serum………..44 Figure III-4: H1299 G0/G1 Synchronization using 0.01% FBS media……………….…45 Figure III-5: Varying seeding amounts with 0.01% serum……………………………..46 Figure III-6: G0/G1 Cell Synchronization and Release……………………………….…48 Figure III-7: Validation of G0/G1 release by a BrdU assay………………………….50-51 Figure III-8: Cell cycle fate of Asynchronous mCherry-H2B H1299 cells in the presence of E4orf4…………………………………………………………………………………53 Figure III-9: Aggregation of G0/G1 arrested mCherry-H2B H1299 cells………………55 Figure III-10: Cell Cycle fate of mCherry-H2B H1299 cells post G0/G1 Synchronization and 18 Hour release in the presence of E4orf4………………………………………..…56 Figure III-11: Cell Cycle fate of mCherry-H2B H1299 cells post G1/S Synchronization with Hydroxyurea (HU) in the presence of E4orf4……………………………………...58 Figure III-12A: Analysis of E4orf4 induced cell death in 2n and 4n cell populations....61 Figure III-12B: Analysis of E4orf4 induced cell death in 2n and 4n cell populations....63 List of Tables Table 1. Methods employed to synchronize H1299 cells in G0/G1……………………...47 Table 2. Comparison of E4orf4 expressing mCherry-H2B H1299 cell mitosis transit compared to vector control AdrtTA…………………………………………………...…59 Neera Sriskandarajah 9 List of Abbreviations Abbreviation aa ACF APC/C ARM BrdU BSA cAMP CAR CAV Cdc Cdk1 cDNA CMV Co-IP CPE CRE DAPI DMEM DNA ds/ssDNA E1A E1B E2A E3 E4 E4ARM E4orf4 E4orf4 Point Mutants E4orf6 eIF4E FACS FBS FFU FITC g G418 hr H1299 HAdV HIV-1 HTLV-1 Neera Sriskandarajah Full Name Amino Acid ATP-utilizing Chromatin assembly and remodeling Factor Anaphase Promoting Complex/Cyclosome Arginine Rich Motif Bromodeoxyuridine Bovine Serum Albumin cyclic-adenosine monophosphate Coxsackie and Adenovirus receptor Chicken Anemia Virus Cell Division Cycle Cyclin-Dependent Kinase 1 complementary DNA Cytomegalovirus Co-Immuno Precipitation Cytopathic Effect cAMP Responsive Element 4’,6-diamidino-2-phenylindole Dulbecco's Modification Eagle's Medium Deoxyribonucleic Acid Double/Single-Stranded DNA Adenovirus Early Region 1A Adenovirus Early Region 1B Adenovirus Early Region 2A Adenovirus Early Region 3 Adenovirus Early Region 4 Early Region 4 Arginine-Rich Motif Adenovirus Early Region 4, Open Reading Frame 4 E4orf4 point mutants are named as Original Amino Acid + Position Number + Mutant Amino Acid Adenovirus Early Region 4, Open Reading Frame 6 Eukaryotic translation Initiation Factor 4E Fluorescent-Activated Cell Sorting Fetal Bovine Serum Fluorescent Focus Unit (FFU) Fluorescein isothiocyanate grams Geneticin Hours Human Lung Carcinoma cell line Human Adenovirus Human Immunodeficiency Virus 1 Human T-cell Leukemia Virus 1 10 HU IP kb kDa min MLP MLTU MOI MRN mRNA Mut NES NLS NPC Nup Nup 205 OA orf p.i. p53 PBS PFU PML PP2A PP2ABα PP2ACdc55 pRb RNA SAC S. cerevisiae SH3 S phase TPL TPR Neera Sriskandarajah Hydroxyurea Immunoprecipitation kilo bases kilo Daltons Minutes Major Late Promoter Major Late Transcription Unit Multiplicity of Infection Mre11/Rad50/Nbs1 protein complex messenger RNA Mutant/Mutation Nuclear Export Signal Nuclear Localization Signal Nuclear Pore Complex Nucleoporin Nuclear pore complex protein Nup 205 (nucleoporin 205kDa) Okadaic Acid open reading frame Post-Infection tumor protein 53 Phosphate Buffered Saline Plaque Forming Unit Promyelocytic Leukemia Protein Phosphatase 2A Bα-containing form of PP2A Cdc55-containing form of PP2A retinoblastoma tumour suppressor protein Ribonucleic Acid Spindle Assembly Checkpoint Saccharomyces cerevisiae Src-homology 3 Synthesis Phase Tri-Partite Leader Sequence Tetratricopeptide Repeat 11 Original Contributions to Knowledge 0.01% foetal bovine serum (FBS) in media (DMEM) arrests p53-/- H1299 human lung carcinoma cells in G0/G1. H1299 cells expressing E4orf4 exhibit increased mobility, a major delay in mitosis, and a significant proportion of cells demonstrate a failure in cytokinesis. Confirmation by time-lapse imaging that cells expressing E4orf4 following release from G0/G1 arrest largely fail to divide. H1299 cells overexpressing E4orf4 undergo cell death at both G1 diploid and G1 tetraploid states. Publications Publications immediately relevant to the thesis 1. Cabon L, Sriskandarajah N., et al. Adenovirus E4orf4 Protein-Induced Death of p53-/H1299 Human Cancer Cells Follows a G1 Arrest of Both Tetraploid and Diploid Cells Due to a Failure to Initiate DNA Synthesis. JVI, Dec 2013. 87(24): p. 13168-78. Neera Sriskandarajah 12 I. Introduction 1. Human Adenoviruses and their Classification Adenoviruses are very common in the world’s human population, as 80% of children under the age of 5 have already been exposed to the virus [1]. Wallace Rowe and his colleagues first isolated Adenovirus from adenoid cell cultures in 1953 in the search to identify the infectious agent causing the common cold [1]. Despite not being the sought after common cold causing target, human adenoviruses (HAdV) were later identified as being responsible for many acute infections affecting the respiratory, enteric, and ocular systems [1-6]. In 1962, HAdV12, the first human oncogenic virus to be discovered, was shown to transform hamster cells [7]; however, thus far adenoviruses have not yet been causally implicated in any human malignancy [8]. Adenoviruses are part of the viral family called Adenoviridae which is divided into two subclasses: Mastadenoviruses and Aviadenoviruses. Mastadenoviruses infect mammals such as humans, monkeys, mice, horses and dogs while Aviadenoviruses infect birds [9]. Thus far, there are more than 60 HAdv serotypes identified based on their sensitivity to neutralizing antibodies [1, 9]. Each subtype is classified into different species (A-F) distinguished by their ability to agglutinate red blood cells [10, 11]. Adenovirus subtypes are also classified according to their oncogenicity in rodents, DNA sequence similarity/homology, virion protein electrophoretic mobility, as well as serum profiles based on antibody binding to virion hexon and fiber proteins [3,12-14]. To date, serotypes belonging to species A and B and two serotypes belonging to species D have been shown to cause tumors when subcutaneously injected into rodents whereas the rest of the subgroups display lesser oncogenic potential [10]. Two HAdV types have been most widely studied, Ad2 and Ad5 each of which belong to species C [1,3]. Adenovirus research has advanced the field of molecular biology in many ways, including through studies of messenger RNA splicing, adenoviral gene therapy, and a Neera Sriskandarajah 13 host of other topics. As such adenoviruses have served and will continue to serve as a useful model system to explore fundamental cellular processes, as well as serve as evolutionary models for other viruses [1-2, 4-6]. 1.1. Adenovirus structure Adenoviruses are large non-enveloped viruses of approximately 90nm in diameter and a molecular weight of 150MDa. The adenovirus structure has been resolved by both X-ray crystallography and cryo-electron microscopy (schematic shown in Figure I-1) [3]. Adenovirus virions are the infective form of the virus and consist of a protein shell called the capsid that protects the viral core genome. Adenovirus virions display icosahedral symmetry with 20 equilateral triangle faces and 12 vertices and are studded with knobbed fibers [10]. The HAdV capsid, surrounds the viral core genome, and is comprised of 11 late viral structural proteins, composing the hexon and penton base shown in Figure I-1. The two main functions of the capsid are to protect the core genome and arbitrate virus entry into host cells. The capsid is composed of 252 units called capsomeres. The iscosahedral symmetry of HAdV results from the arrangement of hexons (protein II) form 240 capsomeres, and the pentons (protein III) forming the remaining 12 units. There are twelve vertices in the capsid containing a penton base, which serves as a base for the noncovalent association to a projecting fiber protein. Each of the rod and knob fiber (protein IV) vertex proteins is crucial for adenovirus host cell attachment and entry during infection [3,13]. Proteins II, III and IV are the main structural proteins. Neera Sriskandarajah 14 Figure I.1. Schematic diagram of the HAdV structure The icosahedral capsid of adenovirus protects the core genome. The capsid is composed of capsid proteins, hexons, penton bases, and fibers. Minor capsid proteins include pIX, pIIIa, pVI and pVIII. The core genome consists of the viral DNA bound to proteins V, VII and mu. Adapted from [3]. Neera Sriskandarajah 15 1.1.2. Adenovirus infection Initiation of the HAdV infection begins with the adenoviral knob domain of the fiber proteins making contact with high affinity cellular receptors, termed Coxsackie and Adenovirus Receptor (CAR), as well as with integrin receptors. The binding of the HAdV penton base to secondary cell surface integrin receptors, such as the αV family, leads to migration of virus-receptor complexes to clathrin-coated pits [10]. This permits the internalization of the partially disassembled virions by clathrin-mediated endocytosis. Once in the cytoplasm the virion disintegrates in the acidic environment of the late endosome releasing the vertex proteins (penton base, pIIIa, fiber, peripentonal hexons). The uncoated virus particles are then released into the cytoplasm by liberating the internal capsid protein pVI, which induces the endosomal membrane to break down. The HAdV genome core is transported to the nucleus via interactions between hexons and the microtubule network. After moving along microtubules, the virus particles are imported by translocation through the host cell nuclear pore complex into the nucleus, where finally the viral DNA is transcribed (Figure I-2) [3]. Neera Sriskandarajah 16 Figure I.2. Schematic diagram of Adenovirus Infection Adenovirus knob domain makes contact with cellular Coxsackie and Adenovirus Receptor (CAR) and integrin receptors. HAdV penton base interaction with integrin receptors results in the packaging of virus-receptor complexes into clathrin-coated pits. The virion collapses under low pH of late endosome releasing the vertex proteins. Virus particles in the cytoplasm are transported along the microtubule network and are translocated through the host NPC into the nucleus. Neera Sriskandarajah 17 1.2. Adenovirus genome and gene expression The adenovirus core genome is associated with four proteins bound to its linear, condensed, double-stranded DNA, ranging in size from 30 to 38 kilo base pairs (Kbp) [1]. Three of these core proteins are arginine-rich basic proteins V, mil (protein X), and histone-like protein VII, which function together to protect and maintain folding of the viral DNA. The fourth protein, pTP, binds the 5’ end of the viral dsDNA covalently and acts as a primer for viral replication in the host’s nucleus (Figure 1-3) [15]. The viral DNA contains inverted terminal repeat sequences approximately 100bp which serve as origins of replication [3, 12]. The adenoviral genome is divided into a set of three transcription units: early, delayed early (intermediate), and late phase each of which is defined by the onset of viral DNA replication. During the early phase of the infection as soon as the viral DNA has been translocated into the host-cell nucleus (about 2hpi), E1A, E1B, E2A, E2B, E3, E4 alternatively spliced mRNA’s are produced from six early transcription promoters independently of viral DNA replication that encode regulatory proteins that prepare the cell for optimal viral DNA replication. There are three preparatory steps that have evolved to ensure effective viral replication. First, E1A expression promotes efficient activation of all early viral transcription units. Second, E1A and E4 products induce host cells to enter S phase. And third, E1B, E3 as well as adenovirus associated RNA (VARNA) inactivate interferon-based host-cell antiviral defense mechanisms [12]. Viral DNA replication quickly follows at 8-10 hours post-infection (hpi) along with the expression of delayed early (intermediate) genes such as IX, IVa2, E2 late. Finally, the expression of five late mRNA’s, L1 to L5, by alternative splicing from one Major Late Promoter (MLP) and five polyadenylation sites, induces progression to the late phase of infection (10-24hpi). This expression also gives rise to structural proteins involved in DNA packaging and assembly of viral particles. The host-cell then begins to break down and virion progeny release occurs (24-36 hpi). The assembly of transcription units is an evolutionary advantage first discovered in adenovirus, that results in the production of multiple viral proteins through alternative Neera Sriskandarajah 18 splicing of full-length mRNA. In fact, products of early units often regulate the expression of late products. For example, the Early Region 4 open reading frame 4 (E4orf4) protein promotes the alternative splicing of adenoviral late mRNA’s to enhance late protein production by causing hypo-phosphorylaton and thus activation of cellular SR splicing factors [15]. Neera Sriskandarajah 19 Figure I-3. Transcriptional map and gene products of adenovirus type 5 (Ad5) Families of early (E in purple), intermediate (in black), and late (L, in blue) mRNAs are shown in relation to viral genome, divided into 100 map units (350nt). Names of proteins encoded by mRNAs are labeled adjacent to the respective mRNA. The vertical bar on the 5’ end of mRNA’s correspond to transcriptional promoters while the 3’ end (arrowhead) corresponds to polyadenylation sites. Adapted from Acheson [10]. Neera Sriskandarajah 20 1.2.1. The Adenovirus E1A gene The Adenovirus Early Region 1A (E1A) is the first viral gene to be transcribed following entry into the host cell nucleus [16]. E1A transcription is driven by a transcriptional enhancer positioned at the E1A promoter and relies on transcription factors provided by the host. E1A is essential for viral replication as viruses containing mutations in E1A genes are highly deficient in early gene transcription, and thus yield reduced amounts of progeny virions. [8]. Some adenovirus gene expression is regulated by viral products as, for example, E4orf4 (discussed in section 2.0) autoregulates E4 transcription. Through alternative splicing, the E1A gene produces five distinct mRNAs with sedimentation coefficients (denoted for the slight different sedimentation rates in sucrose gradients) of 9S, 10S, 11S, 12S, and 13S. However the latter two, 12S and 13S encode proteins 243R and 289R, respectively [18-20]. They are the most abundant proteins produced by the E1A transcription unit and differ by an alternatively spliced intron. There are five conserved regions among adenovirus serotypes within E1A proteins, the N-terminal region, CR1 (conserved region 1), CR2, CR3 and CR4 (Figure I4). E1A protein 13S/289R contains all of these domains, whereas 12S/243R lacks CR3 [12, 21]. It is through each of the three conserved regions of E1A that protein-protein interactions occur. CR1 and CR2 are the main domains involved in adenovirus E1Ainduced oncogenic transformation as they contribute to the induction of host cell S phase entry and further progression through the cell cycle [23-27]. CR1 is specifically necessary to finish a round of the cell cycle [22-23] through interaction with chromatinmodifying complex proteins such as p300/CBP, p400, as well as with CDK inhibitors such as p21 and p27, and proteasome components [23, 27]. E1A’s CR2 domain interacts with the Retinoblastoma (Rb) protein family of pocket proteins via an LxCxE motif. Rb is known to bind and inhibit E2F cellular transcription factors. This inhibition prevents E2F from targeting the genes required for activating viral and cellular DNA synthesis and entry into S-phase. During infection, this Neera Sriskandarajah 21 function allows adenovirus to optimally target quiescent epithelial cells [22]. Adenovirus induces cell cycle entry and progression, through interaction between the E1A-CR2 LxCxE motif and Rb family members Rb, p107 and p130 [28-31] thus freeing E2F to activate expression of cell cycle-dependent genes [32-34]. Examples of E2F activated target genes are cyclin-dependent kinase 2 (Cdk2) and cyclins A and E which drive cells from the G0 phase to the S phase [12, 31-33]. Thus E1A can both activate viral genes that are required for viral DNA replication, and E2F-dependent cellular genes that are required for S phase entry [1]. CR3 is vital to activate early viral gene expression by associating with cellular transcription factors present at these promoters rather than by binding DNA directly [3537]. Both the 12S/243R and 13S/289R forms of E1A proteins are important transcriptional activators of several viral and cellular genes [36, 38]. For example, 243R specifically activates the E2 region while 289R activates the transcription of all the remaining early viral genes. E1A also moderates the expression pattern of host-cellular factors required for transcriptional regulation, cell cycle progression, apoptosis, protein degradation and even deoxynucleotide production for viral DNA replication [8, 36, 3839]. E1A is the major oncogene of hAdV. Through its CR1, CR2 domains and the Nterminal region, E1A is capable of promoting S phase entry, DNA synthesis and cell transformation in otherwise non-dividing host cells. Such cells are capable of forming tumours in nude mice, or in some other cases other rodents [8]. Neera Sriskandarajah 22 1.2.2. The E4 transcription unit There are seven different proteins encoded by Early Region 4 (E4) of human adenovirus, E4orf1, E4orf2, E4orf3, E4orf3/4, E4orf4, E4orf6 and E4orf6/7 [40]. E4 proteins are involved in multiple biological processes that enhance viral replication [4145]. The rate of E4 transcription peaks during the early phase (4hpi) of adenovirus infection and is maintained throughout infection, although the rate declines somewhat later in infection [46]. Among all serotypes of HAdV, the E4 transcription units are highly homologous. The adenovirus E4 gene expression is activated by E1A and regulated at two levels: transcriptionally and post-transcriptionally [47]. In addition, the E1A-dependent expression of the E4 transcriptional unit is subject to E4 negative feedback regulation. In fact, E4orf4 has been shown to down-regulate the expression of all E4 genes [40,48]. For example, E4orf4 inhibits the E1A-mediated transactivation of E4 promoter through the cellular serine/threonine protein phosphatase 2A (PP2A) [49] (discussed in section 2.1.2.1.1). E4orf3, E4orf6, and E4orf4 (discussed in section 2) have been best studied in both serotypes Ad2 and Ad5 [1]. Firstly, E4orf3 enhances viral replication by associating with E1B55K and relocalizing it to structures within the nucleus such as the promyelocytic leukemia (PML) nuclear bodies [50]. This relocalization to PML bodies forces the reorganization of these nuclear bodies and of the MRN protein complex (Mre11/Rad50/Nbs1), which is involved in DNA repair, to “track-like” structures [5153]. Sequestering of the MRN complex by E4orf3 expression lowers DNA damage repair, promotes genomic instability, and contributes to uncontrolled cell proliferation. Moreover, E4orf6 promotes p53 degradation by forming cellular Cullin-based E3 ubiquitin ligase complexes that allows interaction with E1B55K (section1.2.4) [54-57]. Along with p53, E4orf6/E1B55K ubiquitin ligase polyubiquitinates and targets substrates such as Mre11, DNA ligase IV, Integrin α3, and Bloom helicase for degradation by the 26S proteasome [52, 58-61]. Both E4orf3 and E4orf6 increase viral DNA replication and late viral protein synthesis as well as induce host cell shut-off [62-66]. Neera Sriskandarajah 23 1.2.3. Late gene expression The majority of structural proteins are produced during the late phse of gene expression from the adenovirus genome [1]. Eighteen late adenovirus mRNAs are produced under the Major Late Transcription Unit (MLTU). In fact, the MLTU is further sub-divided into five sub-regions, L1 to L5. The major late promoter (MLP) is active at low levels during the early phase of infection such that transcription progresses up to the L3 region. Viral DNA replication triggers the full-level activation of MLP in order to produce sufficient levels of late structural and scaffolding proteins as well as a protease resulting in newly synthesized adenovirus genomes packaged into virions [12, 67). Each of the multiple 18 late mRNAs produced by the MLTU, contains: one noncoding 200-nt, the tripartite leader (TPL) sequence (leaders 1, 2, and 3) at their 5’ ends and 3’ splice and poly-adenylation sites. The TPL promotes efficient nuclear export, translation and increases stability of late mRNAs, while the 3’ poly-adenylation sites promote the essential process of alternative splicing [68]. Adenovirus has evolved mechanisms to enhance this splicing activity, in particular, the action of E4orf4 mentioned earlier [15, 34, 49, 67-68]. Neera Sriskandarajah 24 1.2.4. Host Cell Shut-off Adenovirus employs several strategies to control host cell machinery to optimize replication, including the preferential expression of viral products by reducing translation of cellular proteins. This effect has been coined Host Cell Shut-off. Host Cell Shut-off is exerted at two levels: post-transcriptionally and translationally. Post-transcriptionally for example, the host cell selectively exports a higher proportion of newly synthesized late viral mRNAs over cellular mRNA. In fact, host factors such as the nuclear pore complex and early viral products such as E4orf6 and E1B-55K are vital and cooperate to effectively block cellular mRNA transport to the cytoplasm by a mechanism that is not yet understood [69-72]. Viral mRNAs are also preferentially translated over cellular mRNAs at late stages of infection [73]. This effect is achieved by reducing, CAP-dependent translation of cellular mRNA’s. HAdV late 100K protein of the L4 region (L4-100K) binds to scaffold protein eIF4G causing the displacement of a kinase such that translation initiation factor eIF4E (which normally binds to 5’-m7GpppN CAP structure on most cellular mRNAs) is dephosphorylated. Thus the L4-100K/eIF4G interaction inhibits eIF4F complex association with capped cellular mRNAs. The 5’ TPL in late viral mRNAs also contributes to their preferential translation compared to cellular mRNAs. This is due to a process called ribosome shunting, where 5’-UTR scanning is skipped and ribosomes are immediately transferred to the initiation codon. 1.2.5. Cytopathic Effect (CPE) The Cytopathic Effect (CPE) is the sum of the biochemical and morphological changes following the replication of viral DNA and synthesis of all viral components for virion release. Adenovirus specifically, alters causes host cellular morphology to show characteristics of swelling, rounding, basophilic intra-nuclear inclusion bodies, [74] nuclear shrinking (pyknosis), proliferation of nuclear membrane, vacuoles in cytoplasm, syncytia (cell fusion), margination as well as breaking of chromosomes. Infected cells eventually lift from the culture plate and lyse to release viral progeny. Neera Sriskandarajah 25 2. The Adenovirus E4orf4 protein Adenovirus Early Region 4 Open Reading Frame 4 (E4orf4) protein is most well known for its ability to induce p53-dependent-tumour cell specific killing when expressed alone and at high levels [75-78]. E4orf4 is a small protein in the case of Ad2 and Ad5 (in species C), consisting of 114 amino acids and has a molecular mass of 14 kDa. Although E4orf4 does not share any sequence homology with any known eukaryotic protein, it is conserved among all adenovirus serotypes with differences only in the carboxy-terminus [78]. In the past, many studies were performed to determine the role of E4 gene products of adenovirus. Using deletion and insertion mutants of the Ad5 E4 region, some studies found that E4orf4 is not essential for virus growth in human tumor cells [62, 79], although recent studies in human primary cells suggested an important role (P. Blanchette, unpublished). As stated above, E4orf4 has been shown to be an important regulator of adenoviral gene expression by altering viral transcription and splicing of late viral mRNAs [15, 34, 49, 79]. 2.1. The structure of E4orf4 E4orf4 harbors two short motifs; a proline rich sequence at its amino-terminus and an arginine-rich motif (ARM) in the second half of the protein [80]. It is believed that the 11 amino acid proline-rich sequence (in Ad2, M1 VLPALPAPP; consensus MxxPxLPxPP) could function as an SH3-binding site. One of E4orf4’s known interacting partners is c-Src. c-Src normally binds proteins through SH3-motifs. However, mutating the proline-rich motif does not disrupt E4orf4’s interaction with c-Src. The basic arginine-rich motif (E4 ARM) between residues 66/9 and 75 within the E4orf4 protein [81] resembles a sequence found in human immunodeficiency virus 1 (HIV-1)’s Tat and Rev proteins, Chicken Anemia Virus Apoptin (CAV-Ap) [82, 83], as well as Rex from human T-cell leukemia virus 1 (HTLV-1) [84-89]. The E4ARM functions as a strong inducer of nucleolar targeting and is also necessary for E4orf4-mediated cell death [90] The ARM differs from the normal Nuclear Localization Signal (NLS) in that arginines are more prevalent than lysines. Interestingly the ARM is also required for E4orf4 binding to Src-kinase [90,91]. Neera Sriskandarajah 26 In spite of E4orf4’s small size the crystal structure remains to be resolved, due to high insolubility; however, software such as PredictProtein predict that E4orf4 would have mostly a helical structure based on the primary protein sequence. E4orf4 is rendered non functional and often unstable when portions of the protein are deleted or altered [76, 78]. Such a profile implies that E4orf4 may have tertiary and quaternary structure that could be vital for function and stability [78]. …… 2.1.2. The function of E4orf4 Although E4orf4 has been fairly intensely studied, little is known about the function of E4orf4 within a viral context. Using adenovirus mutants lacking the expression of E4orf4 (E4orf4- virus), it was found that E4orf4 plays a role in the regulation of transcription, splicing, and signal transduction (reviewed in [92-93]). For example E4orf4, with respect to adenovirus, down regulates early viral gene expression, increases splicing of late viral mRNA’s, and up-regulates the switch to late viral gene expression (section 2.1.2.3). Most of the research carried out on E4orf4 has focused on its capacity to induce dose-dependent and p53-independent cancer cell specific death, when expressed alone outside of a viral context [77, 94-95]. As such, these studies often employed adenovirus vectors lacking all viral products other than E4orf4. Neera Sriskandarajah 27 2.1.2.1. E4orf4 interacting partners Given the abundance of molecular pathways that viruses hijack in order to efficiently infect the host, viral proteins functionally interact with several host factors. As mentioned earlier, the discovery of an E4orf4 interaction with the host Bα regulatory subunit of PP2A was later been shown to be crucial for E4orf4-induced cell killing [15, 49, 96-97, 98-100]. In 2000, Lavoie and colleagues identified proto-oncogene tyrosine kinase c-Src as yet another interacting partner of E4orf4 [101]. E4orf4-cSrc interaction provided further advancements in the mechanism of cytoplasmic killing pathway activated by E4orf4. Additional studies have revealed a few other interacting partners, such as ASF/SF2 splicing factors, cellular transcription factor AP-1 [15, 34, 49], as well as the ATP-dependent chromatin-remodeling factor ACF [96]. Although many E4orf4 interacting partners such as these have been found, the vast array of pathways that E4orf4 is implicated in during adenovirus replication, gene expression, and cell death still remains to be elucidated. 2.1.2.1.1 Protein Phosphatase 2A (PP2A) Protein phosphatase 2A (PP2A) is the dominant and most abundant cellular serine/threonine phosphatase. PP2A has a wide range of substrate specificities and is involved in the regulation of a multitude of cellular pathways [102-104]. It is a heterotrimeric enzyme comprising a heterodimeric core holoenzyme and a variable regulatory subunit. Subunits A and C represent the structural and catalytic subunits, respectively forming the dimeric core enzyme of PP2A. In mammalian cells, there are 4 classes of regulatory B subunits, B, B’, B’’, B’’’ [105-106]. In addition, these four diverse classes of B regulatory subunits are further divided into 16 members (4 members for each B class). The B class for example has 4 different highly related members Bα, Bβ, Bγ, and Bδ, each with a molecular mass of 55 KDa [107]. The diversity of regulatory B subunits permits substrate specificity spatial and temporal functions of PP2A [105-106, 108]. Neera Sriskandarajah 28 2.1.2.1.1.2. PP2A the Cell Cycle Protein phosphatases and kinases play a significant role in cell cycle regulation such that the cell cycle progresses unidirectionally in a timely and controlled manner. For example, cell cycle entry is triggered by phosphorylation of cyclin D by cyclin dependent kinase (CDK) 4/6. In order for the cell cycle to progress into S phase, cyclin D must be dephosphorylated by a phosphatase and cyclin E must then be phosphorylated by CDK2 [109]. PP2A is amongst many phosphatases involved in multiple aspects of cell cycle regulation. PP2A simultaneously inhibits all forms of Cdc25 phosphorylation and activation and activates Wee1 to impede the formation of CDK1: cyclin B dimers [110, 111]. Rb and p107 pocket proteins are dephosphorylated also by PP2A (among other phosphatases) in response to UV induced DNA damage as a defense mechanism to halt the cell cycle and repair DNA damage [112]. In addition, PP2A negatively regulates entry into mitosis, by dephosphorylating and inactivating Mitosis Promoting Factor (MPF), such that the levels of activated Cdk1-cyclinB do not reach a high enough level to promote G2 to M transition [113]. Moreover, regulation of phosphorylation status by PP2A is also involved in other processes important in cell cycle progression such as disassembly of the nuclear envelope [114]. 2.1.2.1.1.3. E4orf4-PP2A Interaction PP2A was the first protein discovered to bind E4orf4 in co-immunoprecipitation (co-IP) experiments. The Bα isoform of PP2A, belonging to the B/B55 class of regulatory subunits, was shown to be required for PP2A’s interaction with E4orf4 [76-78]. Our group discovered that E4orf4 binds only to the B/B55 class of regulatory subunits. Importantly, in the absence of interactions with the B subunit, E4orf4-induced cell death was found to be greatly reduced, suggesting that this interaction is necessary for this cell death pathway [115]. The association of Bα with E4orf4 allows the co-IP of the A and C subunits [49]. Moreover, E4orf4 complexed to PP2A is associated with phosphatase activity, which can be inhibited by Okadaic Acid (OA), a potent inhibitor of both PP2A and PP1 [49, 116]. It Neera Sriskandarajah 29 was widely believed that E4orf4 utilizes PP2A activity to induce cell death; however, studies by our group showed that inhibition of PP2A activity by E4orf4 binding was the underlying cause of toxicity because: (1) inhibition of PP2A by low levels of OA or a PP2A peptide inhibitor enhanced cell death; and (2) in vitro assays of PP2A activity against protein substrates indicated that E4orf4 reduced phosphatase activity [117]. Recent extensive studies have elucidated the pattern of binding of E4orf4 to B55 subunits [97,118-119]. The crystal structure of the PP2A holoenzyme containing Bα indicated that substrates interact exclusively with Bα [120]. Using a series of point mutants in Bα and the closely related yeast CDC55 protein our group mapped the E4orf4 binding site to regions just “north” and “south” of the putative substrate binding groove of Bα [119]. This interaction was predicted to block access of normal PP2A substrates and in fact at high levels of E4orf4 this was found to be the case with the substrate p107. Thus our group believes that E4orf4 tumour cell toxicity results from the inhibition of the B55 class of PP2A enzymes and the failure to dephosphorylate key substrates necessary for the survival of tumour cells. However, we believe that E4orf4 toxicity is an artifact of overexpression and that E4orf4-PP2A interactions play a considerably different role at lower levels of E4orf4 found during virus infection. In the case of the virus infectious cycle we now believe, that E4orf4 recruits target phosphorylated proteins into the active site of PP2A to be dephosphoylated [15, 49, 78, 96-97, 98-100]. One such target is ASF (see below), section 2.1.2.3. In terms of high level expression and cell killing, the relationship between E4orf4’s function and PP2A interaction has been studied though mutational analysis, in hopes of understanding the mechanism of E4orf4-induced cell death [78]. Marcellus and colleagues were able to create E4orf4 point mutants along the full length of E4orf4, and group these mutant into two classes, Class I or Class II. Class I mutants were identified to have reduced interaction with PP2A-Bα, and therefore defective in inducing cell death by E4orf4. On the other hand, Class II mutants were seen to bind PP2A-Bα similarly to wild type E4orf4, however they exhibited a reduction in killing [78]. As such there may be other players involved working in synergy with PP2A and E4orf4 to promote E4orf4Neera Sriskandarajah 30 induced cytotoxicity [99]. 2.1.2.3 E4orf4 and splicing E4orf4 not only regulates transcription, but also post-transcriptional processes such as splicing [17, 64-65]. One of E4orf4’s functions during virus infection is to stimulate late mRNA production [121]. E4orf4 promotes the shift from early to late viral gene expression by enhancing the alternative splicing and the production of late viral mRNA L1-IIIa [41]. During the early phase of infection, SR (serine/arginine) splicing factors) repress the alternative splicing of the L1 unit of the MLTU, such that only one L1-IIIa pre-mRNA is made. SR factors bind to the 3RE intronic repressor element at the IIIa 3’ splicing site, such that L1-52, 55K mRNA is produced during early phase. During late phase, the SR factors are removed from the 3RE such that the IIIa form of L1 mRNA can be produced. E4orf4 specifically associates with two hyperphosphorylated SR proteins, SF2/ASF and SRp30c through their RNA recognition motif as well as with PP2A [15]. E4orf4 targets hyperphosphorylated SF2/ASF and SRp30c to complex with PP2A and become dephosphorylated such that the SR proteins are released from the 3RE, thus alleviating the repression [15]. Once SR proteins are release, the splicing of IIIa-premRNA inducing the switch from early to late phase of viral gene expression [15, 121]. 2.2. E4orf4 mediated cell killing and cell cycle E4orf4 in cooperation with PP2A, induces p53-independent cell death in many different cancer cells [76-78, 99, 122], as well as in the yeast S. cerevisiae [123]. In the case of human tumour cells generation of a population of G1 tetraploids and evidence of death by mitotic catastrophe was suggested [124, 125]. Earlier work by our group suggested that E4orf4 toxicity in yeast may involve effects on the cellular Anaphase Promoting Complex/Cyclosome (APC/C). APC/C is an E3-Ubiqtuin Ligase involved in cell cycle protein degradation that promotes the progression of cells through mitosis and that is also very critical for a cell’s exit from mitosis and into G1. During normal cell cycle progression there are two forms of APC Neera Sriskandarajah 31 that are sequentially activated, APCCdc20 and APCCdh1. APCCdc20 is active from early mitosis to anaphase while APCCdh1 complex is active from anaphase and into G1 phase of the cell cycle. Each form is activated sequentially to target its corresponding substrates by polyubiquitination for degradation by the 26S proteasome such that cells can go through mitosis and exit successfully into G1 and initiate a new round of DNA synthesis. Previous studies in our lab using S. cerevisiae as a model system suggested that E4orf4 induces the premature activation of APCCdc20 and the failure to activate APCCdh1, such that substrates of APCCdc20 (securin) are degraded as early as S phase and substrates of APCCdh1 (Cdc20 and Plk1) are stable (Figure I-4) [111]. These results suggested that APC may also be involved in E4orf4 killing of tumour cells and that premature activation of APCCdc20 or inhibition of APCCdh1 could be involved. Neera Sriskandarajah 32 Figure I-4. APC and the cell cycle This figure illustrates the steps involved in APC activation, degradation of APC substrates, and the potential role of E4orf4 and PP2ABα in this process. E4orf4 induces Cdk1 activity, and PP2A is believed to positively regulate the stability of Wee1 and may negatively regulate formation of APCCdc20 via dephosphorylation of core proteins. Cdk1 is known to promote the activation of APCCdc20 and to inhibit activation of APCCdh1. Adapted to the mammalian system from the yeast S.cerevisiae system depicted in Mui et al. 2010 [111]. Neera Sriskandarajah 33 In addition, our group performed two types of synchronization experiments performed on H1299 cells [125]. The first case shown in figure I-5, depicts cells arrested , in G1/S using hydroxyurea (HU). Cells were then infected with either the control vector AdrtTA or vector over expressing E4orf4, AdE4orf4, and finally released. It was found that from 2 to 12 hours, mock, control vector, and cells expressing E4orf4 all progressed though S phase and G2M. Interestingly, from 14 hours onwards the controls continued through mitosis and divided normally to yield 2 daughter cells with 2n DNA content, whereas those expressing E4orf4 exhibited a population of cells that remained 4N and G1 tetraploids, with a clear defect in the reappearance of 2n cells. Thus, cells expressing E4orf4 appeared able to continue DNA synthesis once initiated. In the second synchronization experiment (Figure I-6), cells were arrested in G0G1 by nutrient depletion and serum starvation, and then infected and released by the addition of fresh medium. Once released the mock- and AdrtTA –infected cultures continued through the cell cycle; however, cells infected with AdE4orf4 from around 6 hours exhibited very low levels of 4N S-phase cells. In fact most E4orf4 expressing cells remained 2N and therefore failed to initiate DNA synthesis. Thus, depending on when E4orf4 is expressed within the cell cycle, two populations appear to accumulate, one being G1 tetraploid and the other G1 diploid. Expression of E4orf4 at G1S resulted in 4n cells as cells continued to replicate DNA and exhibited a defect in mitosis/cytokinesis. Expression of E4orf4 at G0G1 resulted in more prominently 2n cells and cells that failed to initiate DNA synthesis. So cells expressing E4orf4 can continue DNA synthesis but cannot initiate new rounds of replication. Given the strength of the evidence above pertaining to the many facets of E4orf4’s mode of action, the precise mechanism of E4orf4 induced cell toxicity remains to be identified, and as such further study was needed to reexamine E4orf4’s effect on the cell cycle. Neera Sriskandarajah 34 Figure I-5. Cell cycle analysis by flow cytometry post-synchronization in S phase with hydroxyurea (HU). Mock-, AdrtTA-, or AdE4orf4-infected H1299 cells were examined by flow cytometry following treatment with HU and released by the removal of drug and the addition of fresh media. Cells were harvested at 2 hour intervals following release and analyzed by flow cytometry. Cabon, Sriskandarajah et al. 2013. [125]. Neera Sriskandarajah 35 Figure I-6. Cell cycle analysis by flow cytometry post-synchronization in G0G1 growth arrest. Mock-, AdrtTA-, or AdE4orf4-infected H1299 cells were studied by flow cytometry following growth arrest in spent medium and addition of full medium and fresh serum. Cells were harvested at 2 hour intervals following release and analyzed by flow cytometry. Cabon, Sriskandarajah et al. 2013. [125]. Neera Sriskandarajah 36 3. Experimental Rationale The adenovirus E4orf4 protein induces cell death in human cancer cells, apparently leaving normal primary cells unharmed. For this reason, E4orf4 could be of some therapeutic interest. Given the strength of the evidence supporting the role of E4orf4 in affecting the cell cycle and initiation of DNA synthesis, a more in depth investigation into the processes involved in E4orf4-induced cell killing was warranted. Thus, the objective of this study is to reexamine the effect of E4orf4 on the cell cycle to help delineate a mode of action of E4orf4 induced cell death. The approach taken for the experiments described in this thesis involved first, optimizing a protocol for arresting cells in G0G1 to allow further analysis of E4orf4 action. Upon identifying the proper conditions for synchronization, I further examined the morphology of cells expressing high levels of E4orf4 as well as significant variations in their cell cycle compared to controls. Specifically, I also studied the fate of the E4orf4 expressing G1-arrested diploid and tetraploid cells. Together with future experiments, this research will perhaps reveal novel cellular factors and components of the cell cycle that may synergistically work with E4orf4 to kill tumor cells. Neera Sriskandarajah 37 II. Materials and Methods Cell culture Two types of immortalized human cell lines were used in this research. First, the human lung carcinoma cell line H1299 (ATCC no. CRL-5803) with a homologous deletion of the p53 gene, as well as the fluorescence modified cell line mCherry-H2BH1299. Both cell lines under normal conditions were maintained in Dulbecco’s Modification Eagle’s Medium (DMEM, Multicell, Catalogue Number 319-005-CL), supplemented with 10% v/v Fetal Bovine Serum (HyClone), (without any antibiotics) at 37 °C under 5% CO2 (95% air). The mCherry-H2B-H1299 cell line specifically was treated with 1ug/ul (Geneticin) G418 to maintain the selection of fluorescent mCherry – H2B-H1299 over H1299. G0/G1 and S phase cell synchronization The final condition I developed to arrest H1299 cells in G0/G1 was to first plate 700 000 cells in 10mm dishes and serum starve cells in 0.01% Fetal Bovine Serum over 4 hours. To synchronize cells in S phase H1299 cells were incubated in (2mM) hydroxyurea (HU) over 24 hours. Release of Synchronized Cells Medium was removed from each of the 6 wells along with any floating cells and placed into sterile Eppendorf vials. Samples were centrifuged at 1200 rpm and the media was aspirated. Any remaining pellet was resuspended in 4ml of 20% FBS media. Adenovirus Vectors Adenovirus vectors lacking the viral E1 region whilst expressing single gene products such as E4orf4 or the rtTA inducer (control) under the cytomegalovirus promoter previously described in [126] were used. The control vector used was AdrtTA, expressing the rtTA inducer, while E4orf4 was overexpressed using AdE4orf4 (expressing hemagglutinin [HA]-tagged adenovirus type 2 [Ad2] E4orf4 protein). Initial stocks of these viral vectors were amplified in 293 cells, and titered using both plaque formation assays and fluorescence formation assays. Neera Sriskandarajah 38 Infection In all experiments, old media was removed and cells were infected for 1h at a multiplicity of infection (MOI) of 50 PFU per cell in a low volume of the serum-free culture medium. Under normal cell conditions, H1299 cells were infected at a stage of 80% confluence. For cell synchronization followed by infection, cells were infected at 24 and 48 hours, for G0/G1 and S phase respectively. After 1 hour of infection the serum free media was replaced with either 10% serum complimented media, or 0.01% serum for normal conditions and S-phase arrest as well as G0/G1 phase arrest respectively. S phase arrested cells post infection were then retreated with 2mM HU to prolong arrest post infection. Cell Cycle Analysis by Flow Cytometry Adherent and non-adherent H1299 cells grown in 6 well plates were harvested using low volumes (200ul) of trypsin. Cells were washed twice with phosphate-buffered saline (PBS) and pelleted by centrifugation at 1200 rpm for 5 minutes. Cells were then resuspended in 500ul of PBS and under a vortex, the samples were fixed and permeabilized by the slow addition of ice-cold 95% ethanol. Samples were either stored at -20°C until cell cycle analysis was performed or immediately pelleted by centrifugation at 1200rpm. Cells were then resuspended in 500ul of PBS and treated with RNAse A (400 _g/ml, RNA 675.50; Bioshop Canada) for 40 minutes at 37°C. Lastly cells were treated with Propidium Iodide (PI) 1ul (50mg/ml, PPI777; Bioshop Canada), or 4’,6-diamidino-2-phenylindole (DAPI) kept on ice (4°C) in the dark until data acquisition by either model: LSRII (Becton Dickinson Inc.), Guava technologies (Millipore), or Cell Lab Quanta Sc Flow cytometer(Beckman Coulter). Results from the first two cytometers were analyzed by FlowJo. For each sample, 20,000 events were collected and measured for relative size (FSC) and granularity (SSC), and aggregated cells were gated out. Acquisition of data from the Beckman coulter was analysed in conjunction with Quanta collection and Quanta analysis software (Beckman Coulter). For 2n/4n and live/dead sort experiments, sorted cells were further analyzed for cell cycle DNA content as described in the previous section except that the cytoplasm Neera Sriskandarajah 39 was first removed by incubation with Vindalov’s buffer consisting of 10 mM Tris base, 10 mM NaCl, and 0.1% Nonidet P-40 described in reference (41). This was performed in order read the DNA content specifically of the nuclei passing through the flow cytometer and to minimize debris post sorting. BrdU assay To evaluate the incorporation of BrdU and establish the proportion of G0/G1 synchronized cells that have been released into S-phase, cells were pulse-labeled with 10 uM BrdU (BRU222.250; Bioshop Canada) for 40 min. Cells were fixed for 24 hours, and washed in PBS containing 0.5% BSA. DNA denaturation was performed with the addition of 1.5M HCl for 20 min at room temperature, and neutralized by adding 0.1 M sodium borate for 4 min. Cells were then centrifuged and washed, and fluorescein isothiocyanate (FITC)-conjugated anti-BrdU antibody was added for 1 h at room temperature. DNA was stained with propidium iodide (PI) before analysis by flow cytometry. Time Lapse Microscopy To study the cellular morphology of H1299 cells overexpressing E4orf4, mCherry-H2B H1299 cells were synchronized in either G0/G1 or S phase and infected with AdE4orf4. Cells were then released as described earlier with the addition of 20% v/v Fetal Bovine Serum (HyClone). Time lapse microscopy was performed using AxioVision 3 Microscope equipped with Axiocam HR (Zeiss, Thornwood, NY) Digital camera. Pictures were taken at 15 minute intervals over 24 hours. Photos were taken using both phase contrast (to observe whole cell morphology) and fluorescence microscopy (looking at Histone 2B and hence the nuclei of the cells) at 10X magnification. S phase arrested cells were released and photos were taken immediately after the addition of new medium. G0/G1 arrested cells were released for 18 hours to provide sufficient time for cell attachment prior to photo acquisition for another 24 hours from this time point. Analysis of the time-lapse images was performed using Image J and Adobe Illustrator. Neera Sriskandarajah 40 Cell Sorting For the 2n/4n experiments, Vybrant DyeCycle violet stain (V35003; Invitrogen Molecular Probes) was used to stain the DNA of live H1299 cells overexpressing control vector AdrtTA and AdE4orf4. Samples were harvested using trypsin, rinsed with PBS and pelleted by centrifugation at 1200rpm. Pellets were each resuspended in 100ul of PBS and treated with the Vybrant DyeCycle violet stain at 10 uM for 30 minutes at 37°C. Samples were then maintained at room temperature prior to sorting by flow cytometry. Using a FACSAria sorter (Becton Dickinson), cells were sorted into two distinct populations according to their DNA content (2n versus 4n). For the live/dead sort experiments, cells were harvested as explained earlier, and resuspended in PBS. And treated with PI without fixation. Using the same FACS Aria sorter (Becton Dickinson), live cells and dying cells were sorted for, denoted by PI+(dying cells) and PI- (living cells). Neera Sriskandarajah 41 III. Results 3.1 G0G1 Synchronization A method for synchronizing human lung carcinoma H1299 cells in G0/G1 needed to be developed in order to test E4orf4’s effect on the cell cycle linked to the induction of cell death. Sugar, nucleotides and sufficient amino acids are required for cells to enter G1 and further progress into S phase. Thus, impeding the production or deceasing the supply of these factors should cause the arrest of cells in G0/G1. Low serum conditions are known to be able to arrest cells like mesenchymal stem cells among others in G0/G1 [127]. However, I have attempted many different serum conditions such as 1% 0.5% 0.1%, none of which appeared to work with H1299 cells; however, 1% FBS treated H1299 cells were studied (see Figure III-1). The control cells treated with 10% serum showed a normal cell cycle profile with approximately 64% of the cells in G0/G1. However, H1299 cells treated with 1% FBS medium exhibited a much lower percentage of cells in G0/G1 and little indication of arrest. Figure III-1. G0/G1 Synchronization by 1% (low) serum conditions Seven hundred thousand H1299 cells were seeded into 6cm plates in either 10% or 1% FBS media for 48 hours. Cells were harvested and fixed/permeabilized with ethanol and cell cycle analysis was performed as described in Materials and Methods. Neera Sriskandarajah 42 To achieve better cell cycle arrest several other approaches were tried (summarized in Table 1). The next procedure was a method devised in our lab called nutrient depleted serum starvation. This method consisted seeding 3 million H1299 cells, plating them in 10 cm dishes, and growing them past full confluence for 5 days. The medium was then harvested from each of the plates, and centrifuged at 1200 rpm to remove any remaining cells. To arrest cells for experiments, cells were plated in the overgrown media (Figure III-2). Unfortunately this method in two experiments gave variable results, yielding 73% and 81% G0/G1 populations. This difference perhaps occurred because of variations from experiment to experiment in the level of nutrient depletion. Thus although this method was somewhat effective I sought a more consistent protocol to achieve G0/G1 arrest. Figure III-2. G0/G1 Synchronization by Nutrient depleted serum starvation H1299 cells were treated with either 10% FBS media (control) or with two batches of nutrient depleted serum media (protocol described earlier), over 72 hours to arrest cells in G0/G1. The second and third panels show a variation in G0/G1 arrest. Neera Sriskandarajah 43 Next amino acid free (aa-) medium was tested to arrest the cells. However, with this approach most cells died as shown by the significant loss in cell numbers compared to the controls. In addition it was very difficult to obtain a clear cell cycle profile from the H1299 cells treated with aa- media. In an attempt to improve the arrest, reduce cell death, and achieve a better profile, 1% and 10% serum was added to the aa- free media and the results are shown in Figure III-3. The results showed that treating H1299 cells with aamedia regardless of serum level did not induce sufficient arrest of the cells in G0/G1. Figure III-3. G0/G1 Synchronization by Amino Acid (aa) free media+/- serum H1299 cells treated with either 10% FBS serum (control), aa- media, aa- media complimented with 1% FBS or aa- media complimented with 10% FBS. Cells treated with aa- media show a decrease in the amount of cells in G0/G1 (43.3%), and an even larger decrease with the addition of serum (33%). Neera Sriskandarajah 44 In a further attempt to arrest cells in G0/G1, I returned to the original idea of low serum, however, I tried 0.01% serum. Figure III-4 shows the G0/G1 arrest obtained by seeding 1 million H1299 cells in 6 cm plates in 0.01% FBS media. Using this method, 91.42 % of the cells were arrested in G0/G1 with the majority of the remaining cells (7.21%) in SubG0. Figure III-4: H1299 G0/G1 synchronization using 0.01% FBS media. One million cells were seeded in 6cm dishes and incubated with 0.01% serum media over 48 hours. This resulted in a 91% G0/G1 synchronization. Neera Sriskandarajah 45 To optimize this condition further I varied the seeding amounts of H1299 for both 48 and 72 hours to find the best conditions for G0G1 synchronization. Figure III-5: Varying seeding amounts with 0.01% serum Five, seven and nine hundred thousand H1299 cells were seeded into 6cm dishes, and incubated in 0.01% FBS media for either 48 hours or 72 hours. Cell cycle analysis was then performed post harvesting, ethanol fixation/permeabilization, and treatment with propidium iodide. Neera Sriskandarajah 46 It was found that seeding 700 000 cells and incubating them in 0.01% FBS medium provided the best condition for the highest G0/G1 synchronization with minimal cell death. For example, at 48 hours, cells had a 73.2 % G0/G1 arrest with 4.4% of the cells in Sub G0, whereas at 72 hours 81% arrest with 6.47 % SubG0 cells. For future experiments this seeding amount and either 48 hours or 72 hours were employed. The following table summarizes some of the methods attempted to achieve a strong G0/G1 H1299 cell synchronization. Table 1: Methods employed to synchronize H1299 cells in G0/G1. Method 1% Serum Nutrient depleted serum starvation Amino acid free media Amino acid free media +low serum (1%) 0.01% Serum Neera Sriskandarajah %Sub G0 0.09 0 %G0/G1 47.46 80.8 %S 31.7 8.9 %G2M 18.1 9.6 0 43.3 9.5 6.3 0 33.6 15.7 12.5 7.21 91.42 1.22 0.15 47 3.2. G0/G1 Cell Synchronization and Release Once the conditions for G0/G1.H1299 cell synchronization were obtained, I determined if the arrested cells were able to be released as a synchronized population. In the first panel (left) of Figure III-6, control (untreated) H1299 cells demonstrated a regular cell cycle profile. In panel 2 (center), H1299 cells exhibited a good arrest in 0.01% medium, for 72 hours and in panel 3 (right) 15 hours following addition of 20% serum a well synchronized population of cells in S phase indicating a good response to serum addition. Figure III-6: G0/G1 Cell Synchronization and Release Control (untreated) H1299 cells were grown for 48 hours in 10% FBS media. Cells were arrested with 0.01% FBS media for 72 hours. Following the addition of 20% FBS medium, cells were grown for 15 additional hours. All samples were harvested, fixed, permeabilized and treated with RNAse A as well as propidium iodide (PI), as described in Materials and Methods for cell cycle analysis. Neera Sriskandarajah 48 To further confirm the release of cells from G1 into S phase a BrDU assay was performed. BrDU is a thymidine analog that is incorporated into newly synthesized DNA during S phase in replicating cells. Five cell samples were examined including: control (untreated), G1S arrested (Hydroxyurea (HU) treated), G0/G1 arrested, as well as G0/G1 arrested and released for either 18 hours and 24 hours, and further incubated with BrdU for 40 min. Cells then were subjected to acid DNA denaturation, treated with anti-BrdU antibody tagged with fluorescein, as well as propidium iodide for flow cytometry analysis. Figure III-9 shows that the BrDU incorporation as demonstrated by the 2dimentional horseshoe plot was very minimal (as expected) for both the G0/G1 arrest and G1S arrest with 1.3 and 4.5 % of the cells in S phase respectively. Once G0/G1 synchronized cells were released (with 20% FBS media), there was a significant rise in the population of cells in S phase, at approximately 18 hours, to 13.1%. Finally by 24 hours of release from G0/G1 synchronization, the cohort of cells in each phase of the cell cycle looked similar to the control, with 29.1% of control cells in S phase and 28.1% in cells that have been released 24 hours post G0/G1 arrest. Therefore the system I created to synchronize H1229 in G0/G1 with 0.01% FBS media worked effectively both in terms of arrest and cell synchronization. Thus this procedure can be employed for further studies of the effects of E4orf4 on the cell cycle. Neera Sriskandarajah 49 Neera Sriskandarajah 50 Figure III-7: Validation of G0/G1 release by a BrdU assay Evaluation of the BrdU-incorporation into S-phase cells. Cells were arrested in G1S (control), and G0/G1, and further released by the addition of 20% FBS media. Cells were harvested and incubated with 10M BrdU for 40 min and further fixed in 90% methanol. Partial DNA denaturation was performed with a low concentration of HCl. The output of BrdU incorporated into the DNA, was observed by a Fluorescein-conjugated anti-BrdU antibody. In addition, PI was added for the DNA content determination. Twodimensional flow cytometry results are presented with BrdU-positive population and the respective cell cycle encircled boxes with their corresponding percentages beside them. Neera Sriskandarajah 51 3.3. Time-Lapse Microscopy of H1299 cells expressing E4orf4 In order to observe the morphological changes as well as alterations of the cell cycle as a result of high level E4orf4 expression in H1299 cells, time-lapse microscopy was performed. Specifically, mCherry-H2B H1299 cells, which are red-fluorescent for histone H2B were employed to monitor the nuclear state of DNA during the cell cycle, by fluorescence microscopy. At precisely the same time points as the fluorescent photos, the cellular morphology was observed by phase contrast. To thoroughly study the effects of E4orf4 on the cell cycle, six videos were made over the course of 18 hours post release from three different conditions: asynchronously growing cells; G0/G1 arrested cells following an 18 hour release in 20% v/v FBS medium; and G1/S arrested cells following removal of HU (Supplemental video 1 and 2). First, in the case of asynchronously growing cells, mCherry-H2B H1299 cells were seeded onto 6 well plates with 10% FBS media. Cells were infected with either the control vector AdrtTA or AdE4orf4 vector expressing E4orf4, for 18 hours. Fresh medium (20% serum v/v) was then added to the cells at the zero time point, and a series of photos were taken over the course of 18 hours. The results of the first two videos are shown in Figure III-8, in which cells were blindly chosen, tracked, and assessed for M1: timing to round up, M2: rounded 2 nuclei, M3: maintenance of cell rounding, cytokinesis, failure of cytokinesis, time to re-adhere to the plate, as well as failure to enter mitosis. Method of time lapse analysis adapted from Gascoigne, et al, 2008 [128]. Neera Sriskandarajah 52 Figure III-8: Cell cycle fate of Asynchronous mCherry-H2B H1299 cells in the presence of E4orf4 Graphical representation of time-lapse microscopy of asynchronous mCherryH2B H1299 cells +/- E4orf4. In each case 50 cells were selected at random and tracked individually. Neera Sriskandarajah 53 H1299 cells expressing the control rtTA (left) demonstrated a quick transit through mitosis. In contrast, H1299 cells expressing E4orf4 as shown by the right panel exhibited increased cell rounding, and a higher percentage of cells exhibited a failure in cytokinesis, rendering a rounded ‘two nuclei’ phenotype which may represent the G1 tetraploid population of cells described previously. In addition, E4orf4 expressing cells that were able to complete cytokinesis successfully took a prolonged time to re-adhere to the plate and enter a new round of the cell cycle. Although at an MOI of 50 most cells should be infected with AdE4orf4 and express E4orf4, there were a few cells that appeared to undergo regular transit through mitosis. However, it may be that these few cells were uninfected or poorly expressed E4orf4. Second, the effect of E4orf4 expression on the release of G0/G1 arrested cells by addition of serum was studied. Cells were arrested in G0/G1 using 0.01% serum as described earlier, infected for the last 18 hours of the arrest, and released with 20% FBS serum. To assess this process further I first examined the morphology of these cells. It was clear that from the moment of serum addition that the cells aggregated and looked abnormal. However, over time the cells slowly started to adhere to the plate and began growing as shown in Figure III-9. At time 0 when fresh medium was added, cells infected with either control vector or vector expressing E4orf4 both exhibited some aggregation. However, by 8 hours the cells started to adhere and by 18 hours most cells had settled onto the plate. Thus an analysis of individual cells of these cultures by time-lapse photography was performed as above (Figure III-10). Neera Sriskandarajah 54 Figure III-9: Aggregation of G0/G1 arrested mCherry-H2B H1299 cells H1299 cells arrested in G0/G1 by 0.01% serum, and infected with AdrtTA or AdE4orf4. Under phase contrast, photos were taken at 15 minute intervals for 24 hours. The aggregated arrested cells begin slowly adhering to the plate. Neera Sriskandarajah 55 Figure III-10: Cell cycle fate of mCherry-H2B H1299 cells post G0/G1 Synchronization and 18 Hour release in the presence of E4orf4 Graphical representation of time-lapse microscopy of G0/G1 arrested mCherry-H2B H1299 cells +/- E4orf4. In each case 50 cells were selected at random and tracked individually. Neera Sriskandarajah 56 Cells expressing the control vector seemed to progress very quickly through mitosis. A reasonable range of time in M1 or time to round up was evident, which we define here as the beginning of mitosis. This variation may be perhaps because of differences in the arrest in G0/G1 such that some cells were in G0 and some in G1. In any case, G0/G1 arrested cells expressing E4orf4 exhibited a significant number of single cells that were rounded up and unable to initiate DNA synthesis. There were a few cells that underwent DNA replication but failed to undergo cytokinesis. This result obtained by time-lapse, confirmed the flow cytometry studies that our group had recently published (Cabon, Sriskandarajah et al, 2013) [125]. Finally, time-lapse microscopy was then performed on mCherry-H2B H1299 cells following G1/S arrest with hydroxyurea (HU) and infection with AdrtTA control or AdE4orf4. Figure III-10 shows that, cells expressing the control rtTA showed a rapid transit through mitosis, with some variation in the time to round up (Red: begin mitosis). Cells expressing E4orf4 at S phase however, contained a large proportion that either remained 4N with two rounded up nuclei, delayed cytokinesis, or even 4N cells that appeared to fail to complete cytokinesis and regressed into a single tetraploid cells that re-adhered to the plate. Once again, these observations confirmed previous flow cytometric results that suggested that G1/S phase arrested cells expressing E4orf4 could complete DNA synthesis, but exhibited complications in completing cytokinesis, such that there is a significant population of cells that remained tetraploid. Neera Sriskandarajah 57 Figure III-11: Cell cycle fate of mCherry-H2B H1299 cells post G1/S Synchronization by Hydroxyurea (HU) in the presence of E4orf4 Graphical representation of time-lapse microscopy of G1/S arrested mCherry-H2B H1299 cells +/- E4orf4. In each case 55 cells were selected at random and tracked individually. Thus all the video profiles demonstrated that cells expressing E4orf4 exhibited Neera Sriskandarajah 58 increased mobility, and rounding up (Supplemental videos 2, 4, 6 ). The following table summarizes some of the findings from the time-lapse videos (Supplemental videos 1 to 6). It is important to note that there is up to a 15 minute time variable for each point. Table 2: Comparison of E4orf4 expressing mCherry-H2B H1299 cell mitosis transit compared to vector control AdrtTA Average Time in Mitosis Condition: Asynchronous G0/G1 synchronization G1/S synchronization Range: Time in Mitosis [min, max] Condition: Asynchronous G0/G1 synchronization G1/S synchronization % of E4orf4 expressing cells within range of Control [min, max] Condition: Asynchronous G0/G1 synchronization G1/S synchronization Ratio of Transit Through Mitosis Condition: Asynchronous G0/G1 synchronization G1/S synchronization % Cells that Cytokinese Condition: Asynchronous G0/G1 synchronization G1/S synchronization AdrtTA AdE4orf4 87.61 min 114.6 min 186.3 min 843.9 min 1042.3 min 628.6 min [30,120 ] min [90,150] min [105, 1050* ] min N/A N/A N/A N/A N/A N/A 2% 0% 100%** 1 1 1 9.63X Slower 9.09 X Slower 3.37 X Slower 88% (44/50) 100%(50/50) 94% (47/50) 10% (5/50) 4% (2/50) 16% (8/50) *1050 is a maximum value, in this case because there were a few cells in the control (rtTA) G1/S synchronization condition that remain rounded, mitotic, and lifted for the whole course of the experiment. Otherwise the maximum would be 165 min and the % of E4orf4 cells within the range of control for G1/S condition would then be 16% (8/50)**. Cells expressing E4orf4 in any of the three conditions showed a significant Neera Sriskandarajah 59 increase in average time in mitosis. This effect is depicted as well by the ratio of transit through mitosis, with E4orf4 expressing cells being between 3 and 10 times slower than the control rtTA expressing cells. Only between 0 and 16% of E4orf4 expressing cells are within the range of the control minimum and maximum time in mitosis. In addition, E4orf4 expressing cells showed a clear failure in cytokinesis with respect to each condition ranging from 4 to 16% of cytokinesing cells compared to controls, which range from 88 to 100%. Therefore cells expressing E4orf4 exhibited an aberrant delay in mitosis as well as a delay/failure in cytokinesis. Neera Sriskandarajah 60 3.4. E4orf4 induces cell death at both G1 diploid and G1 tetraploid states. In our previous studies it was not clear if cells were killed by E4orf4 only when they were induced to form G1 tetraploids or if in fact both 2N and 4N populations arrested in G1 went on to die. To determine the fate of G1-arrested diploid and tetraploid cells expressing E4orf4, two types of experiments were performed. In the first, H1299 cells were infected with either the control vector AdrtTA or vector expressing E4orf4, AdE4orf4 for 3 days. At the 72hpi time point cells were harvested and treated with propidium iodide without fixation and further separated by FACS. Propidium Iodide is a DNA stain that is cell permeable only when cells are fixed or if the cells have started to die. Thus this method was employed to sort specifically dying cells that were not yet at the stage of full DNA fragmentation DNA. The top panels of Figure III-13A demonstrate two cohort of cells classified as PI+ (dying) and PI- (living). The control AdrtTA infected cells showed a 5.9% uptake of dye, whereas cells infected with AdE4orf4 had a significantly increased percentage of 43.8% of cells that were PI+. Nuclei from these dying cells were then analyzed by flow cytometry to determine DNA content. The bottom two panels in Figure III-13A show that the dying nuclei from AdrtTA-infected control cultures were largely 2n in DNA content (85.7%), with a small amount of 4n nuclei (3.8%). The sorted PI + dying cells from the AdE4orf4 infected H1229 cohort of cells, showed a larger proportion of 4n nuclei (10.8%) compared to the control and a decreased percentage of 2n nuclei with 38.6% in keeping with the accumulation of both G1 diploid and G1 tetraploid cells. These results suggested that both 2n and 4n cells probably die following expression of E4orf4. Neera Sriskandarajah 61 Figure III-12A: Analysis of E4orf4 induced cell death in 2n and 4n cell populations. H1299 cells were infected with either AdrtTA or AdE4orf4 vectors for 72 h and sorted for the uptake of PI (dead cells; top panels). The nuclei of these cells were then analyzed by flow cytometry for DNA content (bottom panels). Neera Sriskandarajah 62 To confirm that both 2n and 4n cells die following expression of E4orf4 a second approach was taken. Cells were again infected with either the control vector AdrtTA, or vector expressing E4orf4 (AdE4orf4). At the 48hpi time point, unfixed live cells were stained with Vybrant DyeCycle violet stain and subjected to separation by FACS to isolate 2n and 4n cell populations. The sorted cells were then grown separately in culture for an additional 48 hours, after which the unfixed cells were stained with propidium iodide in order to determine the populations of dead/dying cells, given by high PI stain shown in Figure III-13B. It was observed that AdrtTA-infected control cells, shared similar levels of dead cells in both 2n and 4n recultured cells (14.2% and 11.9%, respectively). Such was also the case with AdE4orf4- infected cells (18.4% and 19.7%, respectively). Thus, these results also indicated that both G1 diploid and tetraploid cells undergo E4orf4-induced cell death. Neera Sriskandarajah 63 Figure III-12B: Analysis of E4orf4 induced cell death in 2n and 4n cell populations. H1299 cells were again infected with either AdrtTA or AdE4orf4 vectors for 48 h and sorted for 2n and 4n live cell populations using the Vybrant DyeCycle violet stain. Cells were then returned to culture for an additional 48 h and further analyzed for live/dead cell populations. Neera Sriskandarajah 64 IV. Discussion 4.1. Development of a novel method of G0/G1 synchronization in p53-/- H1299 Human Lung Carcinoma cells. The G1 phase restriction point of the mammalian cell cycle is the oldest, recognized, and widely accepted control point regulating division cycle in mammalian cells [129]. It is widely known that most cancer cells exhibit reduced sensitivity to G0 arrest. Cell lines such as HEK293 and U2OS cells may be G0/G1 arrested in DMEM medium without FBS for 48 to 72 hours [130]. Interestingly, cells derived from the lung appear to have a more difficult time arresting in G0/G1 using serum deprivation, as for example, pulmonary microvascular endothelial cells (PMVEC’s) [131] and p53-/- human lung carcinoma cells (H1299’s). However, in this thesis I have developed a protocol for H1299 cells using 0.01% FBS media over 48 to 72 hours, which can yield up to 90% G0/G1 synchronization. When seeding the cells, the cells must be washed with PBS to remove any additional serum that may increase the percentage of serum in the media. This low percentage is the right critical balance between G0/G1 arrest and minimization of cell death often seen with full serum deprivation. Although the phenotype of the cells appeared aggregated and senescent, I have shown by flow cytometry, cell cycle analysis as well as time-lapse microscopy that H1299 cells arrested in G0/G1 may be released in a synchronized manner with the addition of 20% v/v FBS media. Neera Sriskandarajah 65 4.2. Cellular morphology of E4orf4 expressing cells Although many studies with respect to cell death induced by E4orf4 have been carried out throughout the years, the cellular morphological changes and cell cycle patterns induced by E4orf4 had yet to be observed in real-time. Through synchronization experiments and cell cycle analysis by flow cytometry, expression of E4orf4 at the G1/S boundary resulted in G1 tetraploid (4n) cells as cells in G1/S had passed the restriction point and continued to replicate DNA but exhibited a defect in mitosis/cytokinesis. Expression of E4orf4 at G0/G1 resulted in G1 diploid (2n) cells that failed to initiate DNA synthesis. By comparison using time-lapse microscopy, mCherry-H2B H1299 expressing E4orf4 at G0G1 and following release demonstrated a failure of the nuclei to replicate and split into two daughter cells. When E4orf4 was introduced into cells at the G1/S boundary and then released, cells continued DNA synthesis and remain G1 tetraploid as seen by the replicated 4n cell forming a cleavage furrow, furrow retraction, and failure of cytokinesis during the time-lapse (Supplementary Figure 6). In addition to confirming the cell cycle effects of E4orf4 depicted in the flow cytometry results (Cabon, Sriskandarajah et al. 2013) [125], E4orf4 expressing cells showed an overall increased mobility, and a major delay in mitosis (from approximately 4 to 10X slower compared to controls). In addition, a significant proportion of cells that failed in cytokinesis. The increased mobility may be linked to the effects E4orf4 previously reported on the Src-pathway [77,101]. There were also some aberrant cells that showed blebbing, which is also characterisic of E4orf4 expression [77,101]. Further studies will be needed to identify the major E4orf4 targets in the mitotic phase of the cell cycle that may be responsible for the G1 diploid and tetraploid states and the eventual death of the E4orf4 expressing cells. One such target discovered in previous studies by our group in yeast had suggested that APC may be a target for E4orf4 [111]. In addition our group has shown in yeast that E4orf4 prematurely activates the early mitotic form APCCdc20 as early as S phase, while inhibiting the activation of late mitotic/early G1 form APCCdh1 (111). APCCdh1 is known to be involved in the regulation of DNA synthesis through the programmed degradation of its corresponding substrates such as Cdc6, Cdt1, and Geminin. As such, it would be of great interest to pursue studies on the relationship Neera Sriskandarajah 66 between E4orf4 and APCCdh1, the link with E4orf4’s inhibition of DNA synthesis as well as E4orf4 induced cell death [132-134]. 4.3. G1 arrest is the critical event leading to E4orf4 induced cell toxicity It has been known for over a decade that the human adenovirus E4orf4 protein, when expressed alone at high levels, effectively kills human cancer cells. There continues to exist considerable ambiguity about the events that induce cell death. Several studies indicated that at least some cell types die by apoptosis [91, 93,134-139]; however, in many cases clear apoptotic hallmarks are absent [93,123,135, 137, 140]. The mechanism of these effects still remains uncertain. Earlier work indicated that a major target responsible for E4orf4 toxicity in both human and yeast cells is the B55/Cdc55 regulatory subunit of PP2A [78, 81,117, 134, 136, 139, 141, 143-146). Thus effects of E4orf4 on PP2A or some as yet unidentified target may be responsible for the mitotic effects found here and in earlier work. Several reports suggested that E4orf4 induces a form of apparent mitotic arrest in both human tumor cells and in yeast [78,81, 123, 142, 144-145). Our group has suggested that effects of E4orf4 on mitotic progression might result in mitotic catastrophe, a phenomenon known to lead to subsequent cell death by a variety of mechanisms, depending on of the cell type [123]. From the synchronization experiments our group has recently published (Cabon, Sriskandarajah et al, 2013) [125], it was suggested that two populations of cells in the G1 phase of the cell cycle accumulate; G1 diploid as well as G1 tetraploid. These two states appear to be conditional upon where in the cell cycle E4orf4 levels become sufficient to induce effects. As such, I have further studied fate of G1-arrested diploid and tetraploid cells expressing E4orf4, to determine whether the cell death as a result of E4orf4 overexpression is due to the accumulation of cells in mitosis and the failure of cytokinesis, or rather the dual G1 arrested diploid and tetraploid states. By sorting first for living and dying cells by PI staining without fixation, cells expressing E4orf4 as expected showed more than a seven fold increase in dying cells compared to the control. Cell cycle Neera Sriskandarajah 67 analysis of these two cohorts determined that there was a large proportion of 4n nuclei and a decreased percentage of 2n nuclei, yet maintaining two population G1 diploid and G1 tetraploid cells (Fig III-12A). To confirm these results, I performed another experiment where cells infected with either control vector or vector expressing E4orf4 were sorted for 2n and 4n populations using live cell Vybrant dye cycle stain. Cells were recultured for 48 hours and stained with PI in their unfixed state, for live/dead analysis. It was observed that both controls and cells expressing E4orf4 shared similar levels of dead cells in both 2n and 4n recultured cells. Thus, these results indicated that both G1 diploid (2n) and G1 tetraploid (4n) cells undergo E4orf4-induced cell death. This suggests that it is the arrest itself and not the formation of tetraploids that is the critical event leading to death. Neera Sriskandarajah 68 4.4. Concluding Remarks The work presented in this thesis provides new insights into the ongoing investigation into the mechanism of action of the adenoviral protein E4orf4. A new useful model for G0/G1 synchronization of p53-/- H1299 human lung carcinoma cells was developed and employed in time-lapse microscopy. The results of this project also demonstrated that E4orf4 expressing cells exhibit a major delay in mitosis and failure in cytokinesis and that the induction of tumor cell death at both G1 diploid and G1 tetraploid states. As such, E4orf4 induced cell toxicity, which if exploited, makes it an attractive target for future novel anti-tumor therapy. V. Supplementary Videos 5.1. Asynchronous mCherry-H2B H1299 cells infected with AdrtTA over 18 hours 5.2. Asynchronous mCherry-H2B H1299 cells infected with AdE4orf4 over 18 hours 5.3. mCherry-H2B H1299 cells post G0/G1 Synchronization infected with AdrtTA +18 Hour release over 18 hours 5.4. mCherry-H2B H1299 cells post G0/G1 Synchronization infected with AdE4orf4 +18 Hour release over 18 hours 5.5. mCherry-H2B H1299 cells post G1/S Synchronization by Hydroxyurea (HU) infected with AdrtTA 5.6 mCherry-H2B H1299 cells post G1/S Synchronization by Hydroxyurea (HU) infected with AdE4orf4 All videos are available at: http://teodorolab.mcgill.ca/Movies.html Neera Sriskandarajah 69 VI. References 1. Fields, B.N., D.M. Knipe, and P.M. Howley, Fields' virology, 2007, Wolters Kluwer Health/Lippincott Williams & Wilkins: Philadelphia. 2. Dingle, J.H. and Langmuir, AD. 1968. Epidemiology of acute, respiratory disease in military recruits. Am Rev Respir Dis, 97, Suppl: 1-65. 3. Nemerow, G.R., et al. 2009. Insights into adenovirus host cell interactions from structural studies. Virology. 384(2): 380-8. 4. Ginsberg, H.S., et al. 1955. Relation of the new respiratory agents to acute respiratory diseases. Am J Public Health. 45: 915-922. 5. Yolken, RH., et al. 1982. Gastroenteritis associated with enteric type adenovirus in hospitalized Infants. J Pediatr. 101, 21-26. 6. Jawetz, E. 1959. The story of shipyard eye. Br Med J. 873-876. 7. Trentin, J.J., Yabe, Y. and Taylor, G. (1962) The quest for human cancer viruses. Science, 137, 835-841. 8. Endter, C. T. Dobner. 2004. Cell transformation by human adenoviruses. Curr Top Microbiol Immunol. 273:163-214. 9. Norrby, E., Bartha, et al. 1976. Adenoviridae. Intervirology, 7, 117-125. 10. Acheson, N.H., Fundamentals of Molecular Virology 2nd edition, 2011, John Wiley & Sons, Inc,: Massachusetts. 11. Rosen,L. 1960. A hemagglutination-inhibition technique for typing adenoviruses. Am J Public Health, 71, 120-8. 12. Berk, A. J. 2007. Adenoviridae: The Viruses and Their Replication, Lippincott Williams & Wilkins. 13. Russell, W. C. 2009. Adenoviruses: update on structure and function." The Journal of General Virology. 90(1): 1-20. 14. Smith, J. G., C. M. Wiethoff, et al. 2010. "Adenovirus." Current Topics in Microbiology and Immunology 343: 195-224. 15. Estmer Nilsson, C., et al. 2001. The adenovirus E4-ORF4 splicing enhancer protein interacts with a subset of phosphorylated SR proteins. EMBO J, 2001. 20(4): p. 864-71. 16. Boulanger, P. A., Blair, G.E. 1991. Expression and interactions of human adenovirus oncoproteins. Biochem J. 275 (2): 281-299. 17. Perricaudet, M., et al., 1979. Structure of two spliced mRNAs from the transforming region of human subgroup C adenoviruses. Nature. 281(5733): 6946. 18. Akusjarvi, G et al. 1979. Sequence analysis of adenovirus DNA.IV. The genomic sequences encoding the common tripartitie leader of later adenovirus messenger RNA. J Mol Biol. 134(1):143-58. 19. Ulfendahl, P.J., et al. 1987. A novel adenovirus-2 E1A mRNA encoding a protein with transcription activation properties. EMBO J. 6(7): 2037-44. 20. Stephens, C. E. Harlow. 1987. Differential splicing yields novel adenovirus 5 E1A mRNAs that encode 30 kd and 35 kd proteins. EMBO J. 6(7): 2027-35. 21. van Ormondt, H., J. Maat, et al. 1980. Comparison of nucleotide sequences of the early E1a regions for subgroups A, B and C of human adenoviruses. Gene 12 (12): 63-76. 22. Howe, J.A., et al. 1990. Retinoblastoma growth suppressor and a 300-kDa protein Neera Sriskandarajah 70 appear to regulate cellular DNA synthesis. Proc Natl Acad Sci USA. 87, 58835887. 23. Stein, R.W., et al. 1990. Analysis of E1A-mediated growth regulation functions: binding of the 300- kilodalton cellular product correlates with ElA enhancer repression function and DNA synthesis-inducing activity. J Virol, 64,4421-4427. 24. Braithwaite, A.W., et al. 1983. Adenovirus-induced alterations of the cell growth cycle: a requirement for expression of ElA but not of E1B. J Virol, 45, 192-199. 25. Zerler, B., et al. 1987. Different functional domains of the adenovirus ElA gene are involved in regulation ofhost cell cycle products. Mol Cell Biol. 7,821-829. 26. Spindler, K.R., et al. 1985. An adenovirus early region lA protein is required for maximal viral DNA replication in growth-arrested human cells. J Virol. 53, 742750. 27. Fuchs, M., et al. 2001. The p400 complex is an essential ElA transformation target. CeZZ, 106, 297-307. 28. Dyson, N. and Harlow, E. 1992. Adenovirus ElA targets key regulators of cell proliferation. Cancer Surv. 12, 161-195. 29. Barbeau, D., et al. 1992. Quantitative analysis of regions of adenovirus ElA products involved in interactions with cellular proteins. Biochem Cell Biol. 70, 1123-1134. 30. Nevins, J.R., et al. 1997. Role of the RblE2F pathway in cell growth control. J Cell Physiol. 173,233-236. 31. Bandara, L.R. N.B. La Thangue. 1991. Adenovirus E1a prevents the retinoblastoma gene product from complexing with a cellular transcription factor. Nature. 351(6326): 494-7. 32. Bagchi, S., et al. 1990. Adenovirus E1A proteins can dissociate heteromeric complexes involving the E2F transcription factor: a novel mechanism for E1A trans-activation. Cell. 62(4): 659-69. 33. Whyte, P., et al., 1988. Association between an oncogene and an anti-oncogene: the adenovirus E1A proteins bind to the retinoblastoma gene product. Nature. 334(6178): 124-9. 34. Bondesson, M., et al., 1996. Adenovirus E4 open reading frame 4 protein autoregulates E4 transcription by inhibiting E1A transactivation of the E4 promoter. J Virol. 70(6): 3844-51. 35. Webster, L.Cc, Ricciardi, R.P. 1991. trans-dominant mutants ofE1A provide genetic evidence that the zinc fmger of the trans-activating domain binds a transcription factor. Mol Cell Biol. 11, 4287-4296. 36. Berk, A.J., et al., 1979. Pre-early adenovirus 5 gene product regulates synthesis of early viral messenger RNAs. Cell. 17(4): 935-44. 37. Liu, F., Green, M.R. 1994. Promoter targeting by adenovirus Ela through interaction with different cellular DNA-binding domains. Nature. 368, 520-525. 38. Jones, N. and Shenk, T. 1979. An adenovirus type 5 early gene function regulates expression of other early viral genes. Proc Natl Acad Sei USA. 76, 3665-3669. 39. Gallimore, P.H., A.S. Turnell, 2001. Adenovirus E1A: remodelling the host cell, a life or death experience. Oncogene. 20(54): 7824-35. 40. Virtanen, A., et al. 1984. rnRNAs from human adenovirus 2 early region 4. J Virol, 51, 822-831. Neera Sriskandarajah 71 41. Tauber, B. T. Dobner, 2001. Molecular regulation and biological function of adenovirus early genes: the E4 ORFs. Gene. 278(1-2):1-23. 42. Imperiale, M.J., et al. 1995. Post-transcriptional control of adenovirus gene expression. Curr Top Microbiol Immunol. 199 (2): 139-71. 43. Leppard, K.N., 1997. E4 gene function in adenovirus, adenovirus vector and adeno-associated virus infections. J Gen Virol. 78 (9): 2131-8. 44. Dobner, T. et al. 2001. Nuclear export of adenovirus RNA. Curr Top Microbiol Immunol. 259: 25-54. 45. Weinberg, D.H. Ketner, G. 1986. Adenoviral early region 4 is required for efficient viral DNA replication and for late gene expression. J Virol. 57(3): 833-8. 46. Nevins, J.R., et al. 1979. Regulation of the primary expression of the early adenovirus transcription units. J Virol. 32(3): 727-33. 47. Akusjarvi, G., et al. 1993. Proteins with transcription regulatory properties encoded by human adenoviruses. Trends Microbiol. 1(5): 163-70. 48. Whalen, S.G., et al. 1997. Phosphorylation within the transactivation domain of adenovirus E1A protein by mitogen-activated protein kinase regulates expression of early region 4. J Virol. 71(5): 3545-53. 49. Kleinberger, T. et al. 1993. Adenovirus E4orf4 protein binds to protein phosphatase 2A, and the complex down regulates E1A-enhanced junB transcription. J Virol. 67(12): 7556-60. 50. Leppard, K. N. et al. 1999. The adenovirus type 5 E1b 55K and E4 Orf3 proteins associate in infected cells and affect ND10 components. The Journal of General Virology. 80 (4): 997-1008. 51. Carvalho, T., et al. 1995. Targeting of adenovirus E1A and E4-ORF3 proteins to nuclear matrix-associated PML bodies. The Journal of Cell Biology. 131(1): 4556. 52. Stracker, T. H., et al. 2002. Adenovirus oncoproteins inactivate the Mre11Rad50-NBS1 DNA repair complex. Nature. 418(6895): 348-352. 53. Doucas, V., A. M. Ishov, et al. 1996. Adenovirus replication is coupled with the dynamic properties of the PML nuclear structure. Genes & Development. 10(2): 196-207. 54. Blanchette, P., et al. 2004. Both BC-box motifs of adenovirus protein E4orf6 are required to efficiently assemble an E3 ligase complex that degrades p53. Mol Cell Biol. 24(21): 9619-9629. 55. Querido, E., et al. 2001. Degradation of p53 by adenovirus E4orf6 and E1B55K proteins occurs via a novel mechanism involving a Cullin-containing complex. Genes & Development .15(23): 3104-3117. 56. Querido, E., et al. 1997. Regulation of p53 levels by the E1B 55-kilodalton protein and E4orf6 in adenovirus-infected cells. Journal Of Virology. 71(5): 37883798. 57. Steegenga, W. T., et al. 1998. The large E1B protein together with the E4orf6 protein target p53 for active degradation in adenovirus infected cells. Oncogene. 16(3): 349-357. 58. Baker, A., K. J. Rohleder, et al. 2007. Adenovirus E4 34k and E1b 55k oncoproteins target host DNA ligase IV for proteasomal degradation. Journal of Virology. 81(13): 7034-7040. Neera Sriskandarajah 72 59. Orazio, N. I., et al. 2011. The adenovirus E1b55K/E4orf6 complex induces degradation of the Bloom helicase during infection. Journal of Virology. 85(4): 1887-1892. 60. Cheng, C. Y., et al. 2011. The E4orf6/E1B55K E3 ubiquitin ligase complexes of human adenoviruses exhibit heterogeneity in composition and substrate specificity. Journal of Virology. 85(2): 765-775. 61. Dallaire, F., et al. 2009. Identification of integrin alpha3 as a new substrate of the adenovirus E4orf6/E1B 55-kilodalton E3 ubiquitin ligase complex. Journal of Virology. 83(11): 5329-5338. 62. Halbert, D. N., J. R. Cutt, et al. 1985. Adenovirus early region 4 encodes functions required for efficient DNA replication, late gene expression, and host cell shutoff. Journal of Virology. 56(1): 250-257. 63. Bridge, E., Ketner, G.1990. Interaction of adenoviral E4 and E1b products in late gene expression. Virology. 174(2): 345-353. 64. Weiden, M. D., Ginsberg, H. S. 1994. Deletion of the E4 region of the genome produces adenovirus DNA concatemers. Proceedings of the National Academy of Sciences of the United States of America. 91(1): 153-157. 65. Huang, M. M., Hearing, P. 1989. Adenovirus early region 4 encodes two gene products with redundant effects in lytic infection. Journal of Virology. 63(6): 2605-2615. 66. Weinberg, D. H., Ketner, G. 1986. Adenoviral early region 4 is required for efficient viral DNA replication and for late gene expression. Journal of Virology. 57(3): 833-838 67. Farley, D. C., et al. 2004. Activation of the early-late switch in adenovirus type 5 major late transcription unit expression by L4 gene products. Journal of Virology. 78(4): 1782-1791. 68. Logan, J. and T. Shenk. 1984. Adenovirus tripartite leader sequence enhances translation of mRNAs late after infection. Proc Natl Acad Sci U S A. 81(12): 3655-9. 69. Babiss, L.E., et al. 1985. Adenovirus E1B proteins are required for accumulation of late viral mRNA and for effects on cellular mRNA translation and transport. Mol Cell Biol. 5(10): 2552-8. 70. Halbert, D.N., et al. 1985. Adenovirus early region 4 encodes functions required for efficient DNA replication, late gene expression, and host cell shutoff. J Virol. 56(1): 250-7. 71. Bridge, E., Ketner, G. 1990. Interaction of adenoviral E4 and E1b products in late gene expression. Virology. 174(2):345-53. 72. Pilder, S., et al. 1986. The adenovirus E1B-55K transforming polypeptide modulates transport or cytoplasmic stabilization of viral and host cell mRNAs. Mol Cell Biol. 6(2): 470-6. 73. Cuesta, R., et al. 2001. Preferential translation of adenovirus mRNAs in infected cells. Cold Spring Harb Symp Quant Biol. 66: 259-67. 74. Enders, J.F. 1954. Cytopathology of virus infections: particular reference to tissue culture studies. Annu Rev Microbiol. 8: 473-502. 75. Kanopka, A., et al. 1996. Inhibition by SR proteins of splicing of a regulated adenovirus pre-mRNA. Nature. 381(6582): 535-8. Neera Sriskandarajah 73 76. Shtrichman, R., et al. 1999. Induction of apoptosis by adenovirus E4orf4 protein isspecific to transformed cells and requires an interaction with protein phosphatase 2A. Proc Natl Acad Sci U S A. 96(18):10080-5. 77. Lavoie, J.N., et al. 1998. E4orf4, a novel adenovirus death factor that induces p53-independent apoptosis by a pathway that is not inhibited by zVAD-fmk. J Cell Biol. 140(3): 637-45. 78. Marcellus, R.C., et al. 2000. Induction of p53-independent apoptosis by the adenovirus E4orf4 protein requires binding to the Balpha subunit of protein phosphatase 2A. J Virol. 74(17): 7869-77. 79. Miron, M. J., P. Blanchette, et al. 2009. Localization and importance of the adenovirus E4orf4 protein during lytic infection. Journal of Virology. 83(4): 1689-1699. 80. Branton, P. E., Roopchand, D.E. (2001). The role of adenovirus E4orf4 protein in viral replication and cell killing. Oncogene. 20(54): 7855-65. 81. Miron, M.J., et al. 2004. Nuclear localization of the adenovirus E4orf4 protein ismediated through an arginine-rich motif and correlates with cell death. Oncogene. 23(45): 7458-68. 82. Heilman, D.W., et al. 2006. Apoptin nucleocytoplasmic shuttling is required for cell type-specific localization, apoptosis, and recruitment of the anaphasepromoting complex/cyclosome to PML bodies. J Virol. 80(15): p. 7535-45. 83. Teodoro, J.G., et al. 2004. The viral protein Apoptin associates with the anaphasepromoting complex to induce G2/M arrest and apoptosis in the absence of p53. Genes Dev. 18(16):1952-7. 84. Hauber, J., M. H. Malim, et al. 1989. Mutational analysis of the conserved basic domain of human immunodeficiency virus tat protein. Journal of Virology. 63(3): 1181-1187. 85. Kubota, S., H. Siomi, et al. 1989. Functional similarity of HIV-I rev and HTLV-I rex proteins: identification of a new nucleolar-targeting signal in rev protein. Biochemical and biophysical research communications. 162(3): 963-970. 86. Kuppuswamy, M., T. Subramanian, et al. 1989. Multiple functional domains of Tat, the trans-activator of HIV-1, defined by mutational analysis. Nucleic Acids. Research. 17(9): 3551-3561. 87. Malim, M. H., S. Bohnlein, et al. 1989. Functional dissection of the HIV-1 Rev trans-activator--derivation of a trans-dominant repressor of Rev function. Cell. 58(1): 205-214. 88. Ruben, S., A. Perkins, et al. 1989. Structural and functional characterization of human immunodeficiency virus tat protein." Journal of Virology. 63(1): 1-8. 89. Siomi, H., H. Shida, et al. 1990. Effects of a highly basic region of human immunodeficiency virus Tat protein on nucleolar localization. Journal of Virology. 64(4): 1803-1807. 90. Miron, M. J., I. E. Gallouzi, et al. 2004. Nuclear localization of the adenovirus E4orf4 protein is mediated through an arginine-rich motif and correlates with cell death. Oncogene. 23(45): 7458-7468. 91. Gingras, M.C., et al. 2002. Cytoplasmic death signal triggered by SRC-mediated phosphorylation of the adenovirus E4orf4 protein. Mol Cell Biol. 22(1): p. 41-56. 92. Kleinberger, T. 2000. Induction of apoptosis by adenovirus E4orf4 protein. Neera Sriskandarajah 74 Apoptosis. 5(3): 211-5. 93. Roopchand, D.E., et al. 2001. Toxicity of human adenovirus E4orf4 protein in Saccharomyces cerevisiae results from interactions with the Cdc55 regulatory B subunit of PP2A. Oncogene. 20(38): 5279-90. 94. Marcellus, R.C., et al., 1996. Adenovirus type 5 early region 4 is responsible for E1A-induced p53-independent apoptosis. J Virol. 70(9): 6207-15. 95. Shtrichman, R. and T. Kleinberger. 1998. Adenovirus type 5 E4 open reading frame 4 protein induces apoptosis in transformed cells. J Virol. 72(4): p. 2975-82. 96. Brestovitsky, A., et al. 2011. The adenovirus E4orf4 protein targets PP2A to the ACF chromatin-remodeling factor and induces cell death through regulation of SNF2h-containing complexes. Nucleic Acids Res. 39(15): 6414-27. 97. Zhang, Z., et al. 2011. Genetic analysis of B55alpha/Cdc55 protein phosphatase 2A subunits: association with the adenovirus E4orf4 protein. J Virol. 85(1): 28695. 98. Ben-Israel, H., et al. 2008. Adenovirus E4orf4 protein downregulates MYC expression through interaction with the PP2A-B55 subunit. J Virol. 82(19): p. 9381-8. 99. Shtrichman, R. et al. 2000. Adenovirus E4orf4 protein interacts with both Balpha and B' subunits of protein phosphatase 2A, but E4orf4-induced apoptosis is mediated only by the interaction with B alpha. Oncogene. 19(33): 3757-65. 100. Muller, U., et al. 1992. Adenovirus E4orf4 protein reduces phosphorylation of cFos and E1A proteins while simultaneously reducing the level of AP-1. J Virol. 66(10): 5867-78. 101. Lavoie, J.N., et al. 2000. Adenovirus E4 open reading frame 4-induced apoptosis involves dysregulation of Src family kinases. J Cell Biol. 150(5): 1037-56. 102. Xu, Y., et al. 2008. Structure of a protein phosphatase 2A holoenzyme: insights into B55-mediated Tau dephosphorylation. Mol Cell, 2008. 31(6): p. 873-85. 103. Hunter, T., 1995. Protein kinases and phosphatases: the yin and yang of protein phosphorylation and signaling. Cell. 80(2): 225-36. 104. Virshup, D.M., 2000. Protein phosphatase 2A: a panoply of enzymes. Curr Opin Cell Biol. 12(2): 180-5. 105. Janssens, V., Goris, J. 2001. Protein phosphatase 2A: a highly regulated family of serine/threonine phosphatases implicated in cell growth and signalling. Biochem J. 353(Pt 3): p. 417-39. 106. Lechward, K., et al. 2001. Protein phosphatase 2A: variety of forms and diversity of functions. Acta Biochim Pol. 48(4): 921-33. 107. Zolnierowicz, S., et al. 1994. Diversity in the regulatory B-subunits of protein phosphatase 2A: identification of a novel isoform highly expressed in brain. Biochemistry. 33(39): 11858-67. 108. Janssens, V., J. Goris, et al. 2005. PP2A: the expected tumor suppressor. Curr Opin Genet Dev. 15(1): 34-41. 109. Morgan, D.O., 2007. The cell cycle: principles of control. Primers in biology, London; Sunderland, MA: Published by New Science Press in association with Oxford University Press; Distributed inside North America by Sinauer Associates, Publishers. xxvii, 297 p. Neera Sriskandarajah 75 110. Jeong, A.L., Yang Y. 2013. PP2A function toward mitotic kinases and substrates during the cell cycle. BMB Rep. 46(6):289-94. 111. Mui, M., Z., et al. 2010. “Adenovirus Protein E4orf4 Induces Premature APCCdc20 Activation in Saccharomyces cerevisiae by a Protein Phosphatase 2A-Dependent Mechanism.” J Virol. 84(9): 4798–4809. 112. Kolupaeva V1, Janssens V. 2013. PP1 and PP2A phosphatases--cooperating partners in modulating retinoblastoma protein activation. FEBS J. 280(2):62743. 113. Boke, E. and I.M. Hagan. 2011. Polo, greatwall, and protein phosphatase PP2A Jostle for pole position. PLoS Genet. 7(8): p. e1002213. 114. Alvarez-Fernandez, M., et al. 2011. "Protein phosphatase 2A (B55alpha) prevents premature activation of forkhead transcription factor FoxM1 by antagonizing cyclin A/cyclin-dependent kinase-mediated phosphorylation." J Biol Chem. 286(38): 33029-33036. 115. Shtrichman, R., et al. 1999. Induction of apoptosis by adenovirus E4orf4 protein is specific to transformed cells and requires an interaction with protein phosphatase 2A. Proc Natl Acad Sci U S A. 96(18): p. 10080-5. 116. Cohen, P., et al. 1990. Okadaic acid: a new probe for the study of cellular regulation. Trends Biochem Sci. 15(3): p. 98-102. 117. Li, S., et al. 2009. The adenovirus E4orf4 protein induces G2/M arrest and cell death by blocking protein phosphatase 2A activity regulated by the B55 subunit. J Virol, 2009. 83(17): p. 8340-52. 118. Horowitz, B. et al. 2013. “Structure- and modeling-based identification of the adenovirus E4orf4 binding site in the protein phosphatase 2A B55α subunit.” J Biol Chem. 288(19):13718-27 119. Mui, M., Z., et al. 2013. Identification of the adenovirus E4orf4 protein binding site on the B55α and Cdc55 regulatory subunits of PP2A: Implications for PP2A function, tumor cell killing and viral replication. PLoS Pathog. 9(11): e1003742. 120. Xu, Y., et al. 2006. “Structure of the protein phosphatase 2A holoenzyme.” Cell. 127(6):1239-51. 121. Kanopka, A., et al. 1998. Regulation of adenovirus alternative RNA splicing by dephosphorylation of SR proteins. Nature, 1998. 393(6681): p. 185-7 122. Branton, P.E. and D.E. Roopchand. 2001. The role of adenovirus E4orf4 protein in viral replication and cell killing. Oncogene. 20(54): p. 7855-65. 123. Kornitzer, et al. 2001. Adenovirus E4orf4 protein induces PP2A-dependent growth arrest in Saccharomyces cerevisiae and interacts with the anaphasepromoting complex/cyclosome. J Cell Biol. 154(2): p. 331-44. 124. Li, S., et al. 2009. The adenovirus E4orf4 protein induces growth arrest and mitotic catastrophe in H1299 human lung carcinoma cells. Oncogene. 28(3): p. 390-400. 125. Cabon L, Sriskandarajah N., et al. 2013 Adenovirus E4orf4 Protein-Induced Death of p53-/- H1299 Human Cancer Cells Follows a G1 Arrest of Both Tetraploid and Diploid Cells Due to a Failure to Initiate DNA Synthesis. JVI. 87(24): p. 13168-78. 126. Li S, et al. 2009. The adenovirus E4orf4 protein induces growth arrest and mitotic catastrophe in H1299 human lung carcinoma cells. Oncogene 28:390Neera Sriskandarajah 76 400 127. Zhou ZJ1, Zheng J. 2007. “Effect of low serum culture on the synchrony of cell cycle of mesenchymal stem cells” Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 23(4): 369-71. 128. Gascoigne, K. E., Taylor, S. S. 2008. Cancer Cells Display Profound Intra- and Interline Variation following Prolonged Exposure to Antimitotic Drugs Cancer Cell. 14:111–122. 129. Cooper, S. et al. 2003. “Reappraisal of serum starvation, the restriction point, G0, and G1 phase arrest points” The FASEB Journal. 17(3): 333-340. 130. Navjotsingh, P. et al. 2012. “Checkpoint kinase 1 (Chk1)-short is a splice variant and endogenous inhibitor of Chk1 that regulates cell cycle and DNA damage checkpoints.” PNAS. 109(1):197–202. 131. Norbert Voelkel, Sharon Rounds The Pulmonary Endothelium: Function in Health and Disease.2009. John Wiley & Sons, Inc: Oxford 132. Landry MC, et al. 2009. Regulation of cell death by recycling endosomes and golgi membrane dynamics via a pathway involving Src-family kinases, Cdc42 and Rab11a. Mol. Biol. Cell 20:4091–4106 133. Barford D. 2011. Structure, function and mechanism of the anaphase promoting complex (APC/C). Q. Rev. Biophys. 44:153–190 134. Lin XH, et al. 1998. Protein phosphatase 2A is required for the initiation of chromosomal DNA replication. Proc. Natl. Acad. Sci. U. S. A. 95:14693–14698. 135. Afifi R, et al. 2001. Selection of apoptosis-deficient adenovirus E4orf4 mutants in Saccharomyces cerevisiae. J. Virol. 75:4444–4447 136. Champagne C, et al. 2004. Activation of adenovirus type 2 early region 4 ORF4 cytoplasmic death function by direct binding to Src kinase domain. J. Biol. Chem. 279:25905–25915. 137. Bacchetti S, Graham F. 1993. Inhibition of cell-proliferation by an adenovirus vector expressing the human wild type-p53 protein. Int. J. Oncol. 3:781–788 138. Petersen BO, et al. 2000. Cell cycle- and cell growth-regulated proteolysis of mammalian CDC6 is dependent on APC-CDH1. Genes Dev. 14:2330–2343 139. Petersen P, et al. 2006. Protein phosphatase 2A antagonizes ATM and ATR in a Cdk2- and Cdc7-independent DNA damage checkpoint. Mol. Cell. Biol. 26:1997–2011 140. Pechkovsky A, et al. 2013. E4orf4 induces PP2A- and Src-dependent cell death in Drosophila melanogaster and at the same time inhibits classic apoptosis pathways. Proc. Natl. Acad. Sci. U. S. A. 110:E1724–E1733. 141. Robert A, et al. 2002. Distinct cell death pathways triggered by the adenovirus early region 4 ORF 4 protein. J. Cell Biol. 158:519–528 142. Koren R, et al. 2004. The scaffolding A/Tpd3 subunit and high phosphatase activity are dispensable for Cdc55 function in the Saccharomyces cerevisiae spindle checkpoint and in cytokinesis. J. Biol. Chem. 279:48598–48606 143. Li Y, et al. 2008. Cdc55p-mediated E4orf4 growth inhibition in Saccharomyces cerevisiae is mediated only in part via the catalytic subunit of protein phosphatase 2A. J. Virol. 82:3612–3623 144. Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, Blagosklonny MV, El-Deiry WS, Golstein P, Green DR, Hengartner M, Knight Neera Sriskandarajah 77 RA, Kumar S, Lipton SA, Malorni W, Nunez G, Peter ME, Tschopp J, Yuan J, Piacentini M, Zhivotovsky B, Melino G, Nomenclature Committee on Cell Death 2009. 2009. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 16:3–11 145. Noton E, Diffley JF. 2000. CDK inactivation is the only essential function of the APC/C and the mitotic exit network proteins for origin resetting during mitosis. Mol Cell. 5:85–95 146. Mui MZ, et al. 2013. Identification of the adenovirus E4orf4 protein binding site on the B55α and Cdc55 regulatory subunits of PP2A: implications for PP2A function, tumor cell killing and viral replication. PLoS Pathog. 9(11):e1003742. Neera Sriskandarajah 78
© Copyright 2025 Paperzz