Telomeres, Telomerase and Disease
Transcription
Telomeres, Telomerase and Disease
An EMBO Conference Series Telomeres, Telomerase and Disease 30 April - 4 May 2014 Brussels, Belgium Organizers Peter Baumann HHMI, Stowers Institute for Medical Research Anabelle Decottignies de Duve Institute Jan Karlseder The Salk Institue for Biological Studies Telomeres, Telomerase and Disease Schedule Outline Wednesday 30th April 15:00 17:30 19:30 19:45 21:00 - 18:00 19:30 19:45 20:45 22:00 Arrival and registration Cocktails and dinner Opening remarks Keynote Session: Titia de Lange Session 1: Telomere dynamics Thursday 1st May 09:00 12:30 14:00 16:00 18:30 20:00 - 12:30 13:30 16:00 18:00 19:45 21:30 Session 2: Telomere dysfunction and disease Lunch Session 3: Telomerase assembly and activation Poster Session 1 Dinner Session 4: Telomere replication Friday 2nd May 09:00 12:45 14:00 16:00 18:00 - 12:45 14:00 16:00 18:00 Session 5: Alternative mechanisms of telomere maintenance Lunch Poster Session 2 Free time Belgium beer tasting and off-site dinner Saturday 3rd May 09:00 12:30 13:45 16:00 18:00 19:00 19:30 - 12:30 13:30 15:30 17:45 19:00 19:30 22:00 Session 6: DNA damage response at telomeres Lunch Session 7: Telomeres and non-coding RNAs Session 8: Structure and function Keynote Session: Elizabeth Blackburn Meeting discussion and closing remarks Banquet dinner Sunday 4th May Breakfast and departure 1 Table of Contents Schedule Summary 1 Sponsors 3 Meeting Details Arrival Information Departure Information Breakfast Social Event 4 5 5 6 Meeting Policy 7 Complete Schedule 8 Abstracts Speaker Abstracts Poster Abstracts 23 73 Participant List 205 Reference Maps 217 2 Sponsors We would like to thank the following organizations for their generous support of this conference: We would also like to thank the following institutions for lending administrative time and support: de Duve Institute Salk Institute for Biological Studies Stowers Institute for Medical Research 3 Arrival Information Husa President Park Hotel Blvd du Roi Albert II 44-1000 Brussels, Belgium Tel: +32 (2) – 203 20 20 Fax: +32 (2) – 203 24 40 [email protected] For those arriving at Brussels International Airport: th We are offering bus transportation between Brussels International Airport and Husa President Park on April 30 during peak arrival times. To take the bus from the airport to the hotel, you will meet near the Java Café on Level 2. A map of the airport is shown below for your convenience. Someone holding a sign reading “Telomeres, Telomerase & Disease” near the café will be able to direct you from there. The first bus will depart for Husa around 10:30. Buses will also leave the airport around 12:30, 14:30, and 16:30, but exact departure times will depend on when a bus is full. Note that the last bus is scheduled to depart around 16:30. If you arrive later than this, you will need to use public transportation (see below) or a taxi. If you are lost in the airport, you can call Florian Poulain (member of the Decottignies lab): 32-497-795930. 4 Additional Transportation Information: If you need to arrange your own transportation, you can find additional information here: http://www.brusselsairport.be/en/passngr/to_from_brussels_airport/train/ The airport train station is located below the terminal (basement level-1). Up to four trains an hour connect the airport to Brussels North, Brussels Central and Brussels Midi stations. You should take the train to Brussels North. Then, the hotel is only a 8-9 minute walk away. Please refer to the ‘Reference Maps’ section at the end of the abstract book for a map. Departure Information Buses will also be provided to shuttle participants between Husa President Park Hotel and Brussels International th Airport on Sunday, May 4 during peak departure times. The first bus will depart for the airport around 5:30. Buses will also leave for the airport around 7:30, 9:30, and 11:30. Breakfast Breakfast will be offered each morning of the conference at Husa President Park. There are two restaurants serving breakfast at Husa President Park, Maison Blanche and St-Roch. Both restaurants will offer the same breakfast. 5 Social Event nd Friday, 2 May | Visit of the Grand-Place, Beer-tasting session, and Dinner Transportation There will be 2 bus transfers between Husa President Park and the Grand-Place. These will occur at 16:30 and 17:00. Room is limited to 50 people per bus. We expect that many participants will want to walk the short distance (about 2 km) or take the metro (from “Gare du Nord” to “Rogier” – Line 3). Beer-tasting sessions There will be 5 consecutive beer-tasting sessions, at the times listed below. There is a maximum of 50 people at each session. 17:30 18:00 18:30 19:00 19:30 The beer-tasting session will take place on the Grand-Place itself, in a place called the “Museum of the Beer”. This is indicated as Arbre d’Or on the picture below. Dinner The off-site dinner will be at La Manufacture, located near the Grand-Place. Restaurant La Manufacture Rue Notre Dame du Sommeil 12 1000 Bruxelles Tel: +32 (0)2 502 25 25 There will be bus transfers between La Manufacture and Husa President Park at the following times: 22:30 23:00 23:30 Participants may also like to have an after-dinner drink in one of the many nearby bars and return to the hotel on foot or by taxi in time for the morning session. Please refer to the ‘Reference Maps’ section at the end of the abstract book for a map. 6 Meeting Policy Information presented during the meeting is considered private information. Therefore, the use of audio, photographic, or video recording equipment in any of the sessions is strictly prohibited. Please make sure that cell phones, computers, and PDAs taken into the meeting room are kept in ‘silent’ mode during the sessions. Meeting participants will be given name badges at the start of the conference. These must be worn to all sessions, meals, and social activities. Participants not wearing name badges may not be allowed into the meeting room. Accompanying guests will not be allowed into the meeting room. However, guests registered with the conference are welcome to attend the meals and social events. 7 Wednesday, April 30th 15:00 - 18:00 Lobby Registration 17:30 - 19:30 Mathilde Cocktails and Dinner 19:30 - 19:45 Fabiola Opening Remarks 19:45 - 20:45 Fabiola Keynote Session, Titia de Lange The repression of PARP1 at telomeres Lecture sponsored by Abcam 21:00 - 22:00 Fabiola Session 1: Telomere dynamics Chair: Anabelle Decottignies Julie Cooper Riff’ing on the final step of telomere segregation at anaphase Susan Smith The role of telomere cohesion in aging and ALT cells Woody Wright Telomere looping: A new paradigm for the regulation of gene expression with progressive telomere shortening Thursday, May 1st 09:00 - 10:30 Fabiola Session 2a: Telomere dysfunction and disease Chairs: Sara Selig and Martin Kupiec Steven Artandi A TERT promoter gradient enables the isolation and purification of spermatogonial stem cells Duncan Baird Telomere dysfunction accurately predicts clinical outcome in two common cancers Nya Nelson The complex and differing roles of TIN2 isoforms in telomere regulation Simon Boulton The function and execution of RTEL1 activities at vertebrate telomeres 10:30 - 11:00 Mat, Fab, Fab II Coffee break 8 11:00 - 12:30 Fabiola Session 2b: Telomere dysfunction and disease Sharon Savage Dyskeratosis congenita caused by an OB fold mutation in TPP1 Maria Blasco Role of telomeres in disease and longevity Mary Armanios Disease-specific manifestations of the telomere syndromes 12:30 - 13:30 Mathilde Lunch 14:00 - 16:00 Fabiola Session 3: Telomerase assembly and activation Chairs: Tracy Bryan and Daniela Rhodes Vicky Lundblad Regulated assembly and disassembly of the yeast telomerase quaternary complex Kazunori Tomita Two-step mechanism of telomerase recruitment and activation in fission yeast Kathleen Collins Assembly, Architecture and Function of Human Telomerase Dirk Hockemeyer The essential role of TPP1 in telomerase dependent telomere maintenance Jens Schmidt Identification of the site on telomerase that binds TPP1 to promote recruitment to telomeres Julian Chen A self-regulating template in human telomerase 16:00 - 18:00 VIP, Fab II Poster Session 1, odd numbered posters, refreshments will be served 18:30 - 19:45 Mathilde Dinner 20:00 - 21:30 Fabiola Session 4: Telomere replication Chairs: Tracy Bryan and Daniela Rhodes Toru Nakamura Negative regulation of telomerase by SUMOylation-dependent Tpz1(TPP1) – Stn1 interaction in fission yeast Carolyn Price Roles of human CST in telomere replication and genome-wide replication rescue David Shore Rif1 controls DNA replication timing and anti-checkpoint functions through recruitment of the PP1 phosphatase, Glc7 9 Friday, May 2nd 09:00 - 10:30 Fabiola Session 5a: Alternative mechanisms of telomere maintenance Chairs: Alessandro Bianchi and Lee Wong Roger Reddel Long-term proliferation of human cancer cells in the absence of telomere length maintenance Miguel Godinho Ferreira Absence of telomerase causes the early onset of cancer in zebrafish Vincent Géli The role of SUMO pathway in telomere recombination Roger Greenberg Interchromosomal homology searches drive directional ALT telomere movement and synapsis 10:30 - 11:00 11:00 - 12:45 Mathilde, Fab, Fab II Fabiola Coffee break Session 5b: Alternative mechanisms of telomere maintenance Paulina Marzec Genome instability driven by targeted telomere insertion in ALT cells Harikleia Episkopou Alternative Lengthening of Telomeres is characterized by reduced compaction of telomeric chromatin Rajika Arora A complex interplay between telomere transcription, stability and recombination in human ALT cells Hilda Pickett NuRD-ZNF827 recruitment to telomeres creates a molecular scaffold for homologous recombination Aaron Mendez Bermudez TRF2 is required to protect heterochromatic pericentromeric regions from topological and replicative DNA damage Geneviève Almouzni The multifaces of chromatin assembly, a recipe that mixes new with old partners 12:45 - 14:00 Mathilde Lunch 14:00 - 16:00 VIP, Fab II Poster Session 2, even numbered posters, refreshments will be served 16:00 18:00 Free time Belgian beer tasting and off-site dinner 10 Saturday, May 3rd 09:00 - 10:30 Fabiola Session 6a: DNA damage response at telomeres Chairs: Maria Teresa Teixeira and Ted Weinert Eros Lazzerini Denchi Characterization of the changes in telomeric chromatin upon DNA damage induction Makoto Hayashi Mitotic telomere deprotection dictates cellular fate upon mitotic arrest Lea Harrington An optimal telomere length protects human tumor cells from ionizing radiation Kate Clark Interactions between CST, DDR, and NMD genes show that Stn1 and Ten1 are the key components of the CST complex 10:30 - 11:00 11:00 - 12:30 Mathilde, Fab, Fab II Fabiola Coffee break Session 6b: DNA damage response at telomeres Karl Lenhard Rudolph Delineating the role of upstream DNA damage responses in cell and tissue aging induced by telomere shortening Jacqueline Jacobs Identification of a critical regulator of DNA repair activity at mammalian telomeres Agnel Sfeir DNA polymerase theta promotes alternative-NHEJ at dysfunctional telomeres Brian Luke Rapamycin prevents checkpoint adaptation via autophagy and Cdc5 (PLK1), which increases proliferative potential following telomere dysfunction Francesca Rossiello A role for DNA damage response RNAs (DDRNA) at dysfunctional telomeres 12:30 - 13:30 Mathilde Lunch 13:45 - 15:30 Fabiola Session 7: Telomeres and non-coding RNAs Chairs: Jiri Fajkus and Eric Gilson Paul Lieberman Telomere Response to Viral Infection Joachim Lingner Regulation and Functions of TERRA ncRNA 11 Antonis Kirmizis Identification of novel telomere regulators through systematic genetic screens in yeast Arturo Londono-Vallejo Human RTEL1 regulates the export and cytoplasmic trafficking of ncRNAs, including telomerase RNA Mattia la Torre AKTIP, an E2 variant enzyme that interacts with lamin, is required for correct telomere maintenance 15:30 - 16:00 16:00 - 17:45 Mathilde, Fab, Fab II Fabiola Coffee break Session 8: Structure and function Chairs: Jiri Fajkus and Eric Gilson Lili Pan A billion years of shelterin – conservation of interaction modules and the potential to form higher order structures Nicolas Thomä Examining telomere architecture in yeast Peter Lansdorp Guanine quadruplex structures revisited Omesha Perera A telomere protective function of human telomerase reverse transcriptase (hTERT), which is independent of catalytic activity Ming Lei Structural basis for protein-RNA recognition in telomerase 18:00 - 19:00 Fabiola Keynote Session, Elizabeth Blackburn Regulation of human immune system CD4+ T cell apoptosis specifically by the RNA component of telomerase Lecture sponsored by Active Motif 19:00 - 19:30 Fabiola Meeting discussion and closing remarks 19:30 - 22:00 Mathilde Banquet dinner Sunday, May 4th Breakfast and departure 12 Poster Session 1 | Thursday, May 1st, 16:00 – 18:00 [1] Detection of DNA damage response RNAs (DDRNAs) at dysfunctional telomeres Julio Aguado, Francesca Rossiello, Fabrizio d'Adda di Fagagna [3] A simple model for tandem telomeric G-quadruplexes Patrizia Alberti [5] Identification of telomere dysfunction in Friedreich ataxia Sara Anjomani Virmouni, Sahar Al-Mahdawi, Chiranjeevi Sandi, Hemad Yasaei, Predrag Slijepcevic, Mark Pook [7] Retroviral activation of TERT expression in chicken B-cell lymphomas Karen Beemon, James Justice [9] Impact of cancer treatments in telomere length: A prospective study Carlos Benitez-Buelga, Lara Sanchez-Barroso, Maria Mercedes Gallardo, Maria Apellaniz, Lucia Inglada Perez, K. Yanowski, Miguel Urioste, Ana Osorio, Maria Antonia Blasco, Cristina Rodriguez-Antona, Javier Benitez [11] Control of telomere replication by phosphorylation Carol Cooley, Anoushka Dave, Mansi Garg, Resham Lal Gurung, Aziz Muhamad, Alessandro Bianchi [13] Mutations in Telomerase Reverse Transcriptase Promoter Contribute to Urothelial Cancer: Mechanism and Therapeutic Implications Sumit Borah, Arthur Zaug, Linghe Xi, Natasha Powell, James Costello, Dan Theodorescu, Thomas Cech [15] Beneficial testosterone treatment in a mouse model of aplastic anaemia Christian Bӓr, Fabian Beier, Miguel Foronda, Maria Blasco [17] The Effect of Polymerase-alpha on Telomerase Processivity Chris Caridi, Connie Nugent [19] Identification and validation of genome instability drivers in a panel of human cancers Maria Antonietta Cerone, Nnenna Kanu, Mcgranahan Nicholas, Pierre Martinez, Charles Swanton [21] Telomere and mitochondrial dysfunction in Duchenne Muscular Dystrophy Alex AC Chang, Foteini Mourkioti, Sang-Ging Ong, Joseph Wu, Helen M Blau [23] Anti-metastatic factor inhibits telomerase activity in aggressive cancer cells - emerging connection between telomerase activity and metastasis suppression Vivek Srivastava, Anirban Kar, Maneesh Kumar, Dhurjhoti Saha, Ankita Singh, Vinod Kumar Yadav, Shantanu Chowdhury [25] Separase is required for telomere protection Francesca Cipressa, Patrizia Morciano, Giuseppe Bosso, Linda Mannini, Grazia Daniela Raffa, Antonio Musio, Giovanni Cenci 13 [27] ALT-specific recruitment of NuRD-ZNF827 to telomeres promotes homologous recombination and protects against senescence and apoptosis Dimitri Conomos, Roger Reddel, Hilda Pickett [29] RPA and Pfh1(Pif1) helicase prevent secondary structures formation during telomere replication Julien Audry, Pierre Luciano, Toru Nakamura, Vincent Geli, Stephane Coulon [31] Dissecting the link between the long non-coding RNA TERRA and the alternative lengthening of telomeres mechanism Katharina Deeg, Karsten Rippe [33] Pervasive hallmarks of Alternative Lengthening of Telomeres (ALT) in diploid Sorex granarius (Soricidae, Eulipotyphla) fibroblast cells Irena Draskovic, Natalia Zhdanova, Julia Minina, Tatiana Karamysheva, Clara Novo-Lopes, Win-Yan Liu, Maria Zvereva, Andrey Skvo -Vallejo [35] Molecular Dynamics of Telomeric Heterochromatin Formation Yi-Min Duan, Yang Zhang, Ting Gong, Jin-Qiu Zhou [37] Ssu72 phosphatase regulates telomere length in S. pombe Jose Miguel Escandell, Clara Reis, Maria Gallo, Miguel Godinho-Ferreira [39] HSF1 regulates TERRA transcription upon heat shock Sivan Koskas, Claire Vourc'h, Virginie Faure [41] CBX1 buffers DNA damage responses and senescence induction in response to oncogene activation, γirradiation and telomere dysfunction T. K. Leucht, S. Foersch, S. Tao, A. Lechel, K. L. Rudolph [43] A high content screen for telomerase trafficking regulators uncovers the cytosolic chaperonin TRiC as essential for TCAB1 folding and telomerase function Adam Freund, Steven Artandi [45] Chromosomal instability and cancer cell stemness in the alternative lengthening of telomeres Fani-Marlen Roumelioti, Maria Chiourea, Christina Raftopoulou, Sarantis Gagos [47] Telomere uncapping impairs K-RasG12V-induced lung carcinogenesis Maria Garcia-Beccaria, Paula Martinez, Marta Cañamero, Francisca Mulero, Maria Blasco [49] Telomeric Protein TRF2 Is an Angiogenic Target of WT1 That Regulates the Expression of PDFGRβ Mounir El Mai, Kay Wagner, Jean-François Michiel, Valerie Renault, Nicole Wagner, Eric Gilson [51] RTEL1 mutations cause Hoyeraal-Hreidarsson syndrome by disrupting an essential function of RTEL1 in telomerase-dependent telomere maintenance Galina Glousker, Zhong Deng, Arturo Londoño-Vallejo, Paul M. Lieberman, Yehuda Tzfati [53] PARP1 and ATRX are Required for Proper Telomere Length Maintenance Adam Harvey, Duncan Baird, Eric Hendrickson 14 [55] Which domains are critical for TRF2-REST interaction? Milan Hluchý J P eče C H f [57] Telomere transcripts without subtelomere sequences improve targeting of telomerase in cancer cells Sandra Sampl, Christian Stern, Doris Mejri, Klaus Holzmann [59] Human Rap1 Induces TRF2 Release from Telomeric DNA E š J š á I eč á M ch Z mme m M H chý C H f [61] Sequence variants of the telomerase reverse transcriptase (TERT) gene in Nicotiana tabacum Jana Jureckova, Eva Sykorova, Miloslava Fojtova [63] Multiple interactions within a primer influences Saccharomyces castellii telomerase activity in vitro Cecilia Gustafsson, Ahu Karademir, Roopesh Krishnankutty, Marita Cohn [65] Interfacial inhibition of human telomerase reconstitution Guillaume Kellermann, Markus Kaiser, Florent Dingli, Olivier Lahuna, Florence Mahuteau-Betzer, Damarys Loewd, Evelyne Ségal-Bendirdjian, Marie-Paule Teulade-Fichou, Sophie Bombard [67] Telomerase RNP complex assembly in disease background Elzbieta Kowalska, Georgeta Zemora, Michael Wildauer, Christina Waldsich [69] Telomere Length Kinetics Assay (TELKA) sorts the Telomere Length Maintenance (tlm) mutants into functional groups Martin Kupiec, Linda Rubinstein, Yaniv Harari, Vera Babin [71] Anticancer mechanism of TMPyP4 in breast cancer cell lines MCF7 and MDA-MB-231 Natalia Lipinska, Hanna Holysz, Aleksandra Romaniuk, Mariusz Kaczmarek, Blazej Rubis [73] Identification of the subtelomeric sequence of the long non-coding RNA TERRA unveils its role at telomeres Isabel López de Silanes, Osvaldo Graña, Maria Luigia De Bonis, Orlando Domínguez, David G. Pisano, Maria Antonia Blasco [75] Unraveling the changes in telomere composition during senescence, immortalization, and tumorigenic conversion of normal human fibroblasts Jana Majerská, Joachim Lingner [77] Bridging ageing and metabolism through RAP1? Paula Martinez, David Pisano, Maria A. Blasco [79] HOMOLOGY-DEPENDENT REPAIR IS INVOLVED IN 45S rDNA LOSS IN FAS MUTANTS Veronika Muchova S m m I M zg á M D řáč á M E. G eg Fajkus Ch e Wh e J ří 15 [81] Expression of ATRX represses the Alternative Lengthening of Telomeres phenotype Christine Napier, Jane Noble, Roger Reddel [83] TRF2 Overexpression Induces Telomeric Ultrafine Anaphase Bridges Bernadette Nera, Hui-Shun Huang, Thao Lai, Lifeng Xu [85] Selective Bioluminogenic HDAC Activity Assays for Profiling HDAC Inhibitors Andrew L. Niles, Nathan J. Evans, Kevin R. Kupcho, Thomas A. Kirkland, and Dan F. Lazar [87] Androgenetic fish as models in studies concerning healing of chromosome breaks by de novo telomere addition Konrad Ocalewicz [89] Unusual telomeres in plants Vratislav Peska, Jiri Fajkus [91] Long-term particulate air pollution in association with the mitochondrial-telomere axis of ageing Nicky Pieters, Bram G. Janssen, Karen Smeets, Harrie Dewitte, Bianca Cox, Michelle Plusquin, Ann Cuypers, Tim S. Nawrot [93] Preclinical assessment of the telomerase inhibitor Imetelstat combined with radiotherapy in an orthotopic model of human glioblastoma Sylvain Ferrandon, Céline Malleval, Priscillia Battiston-Montagne, Badia El Hamdani, Radu Bolbos, Patrick Manas, Sergei M Gryaznov, Claire Rodriguez-Lafrasse, Jérôme Honnorat, Delphine Poncet, et al. [95] Telomere Repeat Binding (TRB) proteins serve as functional components of telomeres and interact with telomerase (TERT) Petra Prochazkova Schrumpfova, Ivona Vychodilova, Martina Dvorackova, Jana Majerska, Ladislav Dokladal, Sarka Schorova, Jiri Fajkus [97] Effects of dyskeratosis congenita patient mutations in hTERT on telomerase dimerization and activity Alix Christen, Sophie Redon, Sascha Feuerhahn, Joachim Lingner [99] MTV, a telomeric ssDNA-binding complex, protects Drosophila telomeres and recruits retro-transposon to chromosome ends yi zhang, liang zhang, guanjun gao, yikang rong [101] Induced pluripotent stem cells as a model for telomeric abnormalities in ICF type I syndrome Shira Sagie , Erika Ellran, Hagar Katzir, Rony Shaked, Alaa Ghanayim, Maty Tzukerman, Sara Selig [103] The inhibition of protein-protein interactions within telomeres: a route to cancer chemotherapy? Twana Salih, Weng Chan, Lodewijk Dekker, Charles Laughton [105] Telomere maintenance mechanisms without telomerase are of clinical relevance in colorectal carcinoma Sandra Sampl, Stefan Stättner, Fabienne Bastian, Friedrich Wrba, Brigitte Wolf, Jeremy Henson, Loretta Lau, Roger Reddel, Klaus Holzmann [107] A Shelterin-RTEL1 interaction required for maintaining telomere integrity Grzegorz Sarek, Stephanie Panier, Jean-Baptiste Vannier, Simon Boulton 16 [109] A mouse model for ICF syndrome does not recapitulate the telomeric phenotype of human ICF syndrome patients Gal Larom, Shiran Yehezkel, Robert J. Duszynski, Lucy A. Godley, Claire Francastel, Guillaume Velasco, Sara Selig [111] Subtelomeric elements of the shortest telomere are required for TERRA expression and buffer senescence onset Kamar al zaman Serhal, Marco Graf, Pascale Jolivet, Maria Teresa Teixeira, Brian Luke [113] Development of a high-throughput, fluorescence-based, PCR-free method of C-circle detection to screen drugs and genes targeting the alternative lengthening of telomeres (ALT) pathway in human cancers David Halvorsen, Haroldo Silva [115] The role of DAXX and ATRX in telomere maintenance Zhou Songyang, Quanyuan He, Mengfan Tang [117] Evidence for telomerase- and Rad52-independent sequence alterations at yeast telomeres Clémence Claussin, Michael Chang, Sonia Stinus [119] in vivo isolation of telomeric proteins in the nematode Caenorhabditis elegans Sanghyun Sung, Beomseok Seo, Junho Lee [121] DNA DAMAGE SIGNALLING IN THE RECRUITMENT OF TELOMERASE TO TELOMERES Adrian Tong, Josh Stern, Scott Cohen, Xu-Dong Zhu, Tracy Bryan [123] Single-molecule imaging of RTEL1 in embryonic stem cells Evert-Jan Uringa, Andrew Robinson, Karl Duderstadt, Antoine Van Oijen, Peter Lansdorp [125] Depression and shortened telomeres: implication on shelterin and telomerase Yabin Wei, Lena Backlund, Lina Martinsson, Aleksander A. Mathé, Gregers Wegener, Martin Schalling, Catharina Lavebratt [127] Structure-Function studies on Cdc13 Sofiane Y. Mersaoui, Raymund J. Wellinger [129] Roles of ATRX and H3.3 to assemble a repressive chromatin state at telomeres Maheshi Udugama, Fiona Chang, Lyn Chan, James McGhie, Philippe Collas, Jeffrey Mann, Lee Wong [131] TPP1 is a substrate for the deubiquitinating enzyme USP7 Ivo Zemp, Patrick Reichenbach, Joachim Lingner 17 Poster Session 2 | Friday, May 2nd, 14:00 – 16:00 [2] RTEL-1 promotes telomere-ITS recombination events Megan Brady, Shawn Ahmed [4] Severe liver fibrosis caused by S. mansoni infection in Dkc1m hypomorphic mice Raquel Alves-Paiva, Leandra Ramalho, Cleide Araujo-Silva, Olinda Mara, Vanderlei Rodrigues, Eduardo Rego, Rodrigo Calado [6] Longer telomeres lead to Type II survivors in the absence of telomerase in yeast Usha Aryal, Phillipa Muston, Edward J Louis [8] A novel method of analysis of TANK1 enzymatic activity influence on TRF1 DNA-binding Petra Bencúrová P cj Kł I eč á C H f [10] Implication of INT6 in telomere stability Maname Benyelles, Vincent Mocquet, Serge Bauwens, Pierre Jalinot [12] PLATINATION OF TELOMERES BY CIS-PLATINE IS NOT AT THE ORIGINE OF TRF2 DISPLACEMENT Lina Saker, Samar Ali, Guillaume Kellermann, Evelyne Ségal-Bendirdjian, Joël Poupon, Sophie Bombard [14] Regulation of human telomere transcription Joanna Boros, Anabelle Decottignies [16] The putative Leishmania spp. telomerase RNA (TER) undergoes trans-splicing and contains a conserved template sequence Elton José Vasconcelos, Vinicius Santana Nunes, Marcelo Santos da Silva, Marcela Segatto, Peter Myler, Maria Isabel Nogueira Cano [18] EXPRESSION OF A SMALL INTERNAL FRAGMENT OF DYSKERIN (GSE24.2), DECREASES DNA DAMAGE, OXIDATIVE STRESS AND SENESCENCE IN ATAXIA TELANGIECTASIA CELLS Jaime Carrillo, Laura Pintado Berninches, Cristina Manguán García, Laura Iarriccio, Guillermo Güenechea Amurrio, Juan A. Bueren, Leandro Sastre, Rosario Perona [20] The Ctc1/Stn1/Ten1 complex localizes in APB and regulates C-circle formation in ALT cells Chenhui Huang, Xueyu Dai, Weihang Chai [22] CTC1 Mutations in Telomere Syndromes Impair Telomere Replication Liuh-yow Chen, Jana Majerská, Joachim Lingner [24] Characterization of the binding of the Drosophila telomeric protein Verrocchio to single-stranded DNA Alessandro Cicconi, Emanuela Micheli, Domenico Raimondo, Giovanni Cenci, Maurizio Gatti, Stefano Cacchione, Grazia D. Raffa [26] TELOMERE DAMAGE AND CHROMOSOME INSTABILITY INDUCED BY ACUTE OXIDATIVE STRESS Elisa Coluzzi M c C m e C zz S ef e e h O’C gh e Sgura 18 [28] Shading the shelterin TRFH TRF2 domain through cell-permeable chemical tools: rational design, synthesis and biological effects at telomeres Salvatore Di Maro, Pasquale Zizza, Erica Salvati, Bruno Pagano, Clemente Capasso, Luciana Marinelli, Ettore Novellino, Annamaria Biroccio, Sandro Cosconati [30] Cross talk between EBV and telomerase: the role of TERT in the switch of latent/lytic cycle of the virus Silvia Giunco, Andrea Celeghin, Stefano Indraccolo, Riccardo Dolcetti, Anita De Rossi [32] Influence of telomere dynamics on disease progression and response to therapeutics in bone marrow failure syndromes Erin Degelman, Nicholas Ting, Tara Beattie [34] Telomere Maintenance in Pediatric Medulloblastoma Matthew Sobo, Kathleen Dorris, Ashley Margol, Charles Stevenson, Shahab Asgharzadeh, Stewart Goldman, Lili Miles, Arzu Onar-Thomas, Maryam Fouladi, Rachid Drissi [36] Role of DNA end-binding in Ku heterodimer function Charlene Emerson, Christopher Lopez, Albert Ribes-Zamora, Alison Bertuch [38] Roles of DNA repair factors in plant telomere maintenance Lucie Najdekrova, Miloslava Fojtova, Dagmar Zachova, Karel J. Angelis, Jiri Fajkus [40] Telomerase is essential for zebrafish heart regeneration Dorota Bednarek, Juan Manuel González-Rosa, Oscar Gutiérrez-Gutiérrez, Tania Aguado, Gabriela Guzmán, Alfonso Cortés, Agustín Zapata, Jesús Jiménez-Borreguero, Nadia Mercader, Ignacio Flores [42] Epigenetic regulation of telomeres and telomerases in different model plants Miloslava Fojtova, Pavla Polanska, Eva Majerova, Jana Jureckova, Anna Ogrocka, Jiri Fajkus [44] The Diversity and Evolution of Telomeres in Algae Jana Fulneckova, Tereza Sevcikova, Jiri Fajkus, Alena Lukesova, Marek Elias, Eva Sykorova [46] Yeast Rap1 binds dsDNA in multiple binding modes Erik Feldmann, Roberto Galletto [48] Interactions between the Elg1 clamp-unloader and the DNA damage checkpoint kinase Dun1 Inbal Gazy, Martin Kupiec [50] Keeping telomere under wraps to avoid DNA damage: a job for TRF2 Delphine Bennaroch-Popivker, Sabrina Pisano, Alexandre Gay, Aaron Mendez-Bermudez, Chrysa Latrick, Bei Pei, Simona Miron, Marie-Hélène Le Du, Eric Gilson, Marie-Josèphe Giraud-Panis [52] Development of a high-content, automated platform for rapid analysis of alternative lengthening of telomeres (ALT)-associated promyelocytic leukemia nuclear bodies (APBs) in human cancer cells David Halvorsen, Thomas Hunt, Manali Aggrawal, Haroldo Silva [54] Local telomerase over-expression promotes cancer in a tissue-dependent manner Catarina M. Henriques, Miguel Godinho Ferreira 19 [56] A rabbit ear mechanism by which human Rap1 modulates TRF2 attraction to telomeric DNA E š J š á I eč á M ch Z mme m M H chý Ctirad Hofr [58] Telomerase modulates cyclinD1 gene in concert with NOL1 Juyeong Hong, Jihoon Lee, In Kwon Chung [60] Influence of environmental factors on telomerase-independent survivor formation in fission yeast Kristi Jensen, Baumann Peter [62] Fission yeast without subtelomeres Sanki Tashiro, Yuki Nishihara, Junko Kanoh [64] Dissecting the telomeric and non-telomeric roles of human CTC1 through gene disruption Christopher Kasbek, Jason Stewart, Mary Chaiken, Caitlyn Goodwin, Carolyn Price [66] Therapeutic agents influence the TRF2-TIN2 interactions and their cellular distribution: study in human cancer cell lines Patrycja Klos, Ctirad Hofr, Ivona Necasova, Jiri Fajkus [68] Alternative lengthening of telomeres detected in telomerase-positive canine histiocytic sarcoma Theresa Kreilmeier, Sandra Sampl, Marlene Hauck, Ingrid Walter, Klaus Holzmann, Miriam Kleiter [70] Involvement of SRSF11 in cell cycle-specific recruitment of active telomerase to telomeres at nuclear speckles Jihoon Lee, Sun Ah Jeong, Prabhat Khadka, Juyeong Hong, In Kwon Chung [72] The Mechanism of Epigenetic Behavior at the Mre11/Rad50 Interface Danielle Tatum, In-Joon Baek, Heewon Park, Arthur Lustig, et al. [74] Epigenetic landscape of terminal and internal telomeric DNA using different plant models Eva Majerova, Terezie Mandakova, Miloslava Fojtova, Jiri Fajkus [76] Differential processing of double strand breaks and telomeres at the nuclear envelope in S. cerevisiae Isabella Marcomini, Chihiro Horigome, Susan M. Gasser [78] Telomere shortening induces skeletal and cardiac muscle failure in Duchenne Muscular Dystrophy Foteini Mourkioti, Alex Chang, Alan Meeker, Helen Blau [80] The effects of lifestyle factors on leukocyte telomere length dynamics: Results from the ESTHER Cohort Aysel Muezzinler, Aida Karina Dieffenbach, Katja Butterbach, Hermann Brenner [82] In vitro assessment of the protective role of Cdc13 against degradation of telomeric single-stranded 3’ overhangs Saishyam Narayanan, Cecilia Gustafsson, Marita Cohn [84] How specific is TRF2 binding to telomeric DNA? I eč á, Michal Zimmermann, Ctirad Hofr [86] IDENTIFICATION OF NEW TERT GENE MUTATIONS IN FAMILIAL STUDIES OF PATIENTS PRESENTING WITH APLASTIC ANEMIA/HYPOCELLULAR MYELODYSPLASTIC SYNDROME Valeria Nofrini, Valentina Pierini, Francesco Berardinelli, Caterina Matteucci, Valeria Di Battista, Antonella Sgura, Tamara Iannotti, Alessandro Brozzi, Roberta La Starza, Cristina Mecucci 20 [88] A new role for histone deacetylase 5 in the maintenance of long telomeres Catherine Polese, Clara Lopes Novo, Nicolas Matheus, Anabelle Decottignies, Arturo Londono-Vallejo, Vincent Castronovo, and Denis Mottet [90] A study of telomere-related biomarkers in an arsenic-exposed Bangladeshi population Brandon Pierce, Shantanu Roy, Farzana Jasmine, Muhammad Kibriya, Habibul Ahsan [92] Mitochondria dysfunction in telomerase zebrafish mutants Inês Pimenta de Castro, Madalena Carneiro, Miguel Godinho Ferreira [94] Human RTEL1 is required for the maintenance of long telomeres Rosa Maria Porreca, Christian Naucke, Mike Schertzer, Mylène Perderiset, Irena Draskovic, Arturo Londoño-Vallejo [96] In vivo selection for new telomerase RNA template regions in S. pombe Margaret Pruitt, Richard Dannebaum, Peter Baumann [98] Quantile regression for flow-FISH telomere length measurement as a diagnostic tool for telomere diseases Fernanda Gutierrez Rodrigues, Priscila Santos Scheucher, Edson Zangiacomi Martinez, Bárbara Amélia Aparecida Santana, Rodrigo Tocantins Calado Telomere length dynamics and hematopoietic differentiation in human DKC1-mutant iPSCs Flavia Sacilotto Donaires, Thomas Winkler, Cynthia Dunbar, Rodrigo Tocantins Calado [100] [102] Studies of G quadruplex-hRPA interactions in the telomere maintenance context Layal Safa, Jean-François Riou, Carole Saintomé [104] Uncapped telomeres as determinant for cell sensitivity to G-quadruplex ligands Erica Salvati, Angela Rizzo, Sara Iachettini, Pasquale Zizza, Chiara Cingolani, Carmen D'Angelo, Manuela Porru, Annamaria Biroccio [106] Telomere Structure and Maintenance in Trypanosoma brucei Ranjodh Sandhu, Bibo Li [108] Associations between five-factor model of personality and leukocyte telomere length in elderly men and women – the Helsinki Birth Cohort Study (HBCS) Katri Savolainen, Johan Eriksson, Eero Kajantie, Anu-Katriina Pesonen, Katri Räikkönen [110] Dissecting Alternative Lengthening of Telomeres in the Nematode Caenorhabditis elegans Beomseok Seo, Chuna Kim, Mark Hills, Sung Sanghyun, Stephane Flibotte, Donald Moerman, Lee Junho [112] Telomere alterations and chromosome segregation defects induced by prolonged oxidative stress treatment Elisa Coluzzi, Rossella Buonsante, Anthony J. Asmar, Kelley L. Miller, Daniela Cimini, Antonella Sgura 21 [114] TERT Promoter Mutations Are a Major Indicator of Poor Outcome in Differentiated Thyroid Carcinomas Paula Soares, Miguel Melo, Adriana Gaspar da Rocha, João Vinagre, et al. [116] Choice of processing dysfunctional telomeres influences stem cell aging and survival Tobias Sperka, Satjavani Ravipati, Omid Omrani, Lenhard Rudolph [118] In vivo Interaction of Telomerase, Telomere homeostasis and the Human Papillomavirus Oncogenes Charis Achilleos, Stella Michael, Katerina Strati [120] The DNA-end-replication problem and the establishment of replicative senescence Julien Soudet, Emilie Fallet, Pascale Jolivet, Michael Lisby, Eric Gilson, Maria Teresa Teixeira [122] Effects of bilirubin on telomere integrity and dynamics Anela Tosevska, Christine Moelzer, Karl-Heinz Wagner Insights into telomere protection by Ku heterodimer Sona Valuchova, Eliska Janouskova, Ctirad Hofr, Karel Riha [124] [126] Chromosome Instability: Major Events Likely Start in the Telomeres, Stupid Ted Weinert, Tracey Beyer, Pete Vinton, Rachel Langston, Margherita Paschini, Vicki Lundblad [128] The structural organization of the human telomerase Michael Wildauer, Christina Waldsich [130] A cryptic route to senescence reveals a constitutive protective function of telomerase Zhou Xu, Emilie Fallet, Camille Paoletti, Steffen Fehrmann, Gilles Charvin, Maria Teresa Teixeira [132] SUMOylation of Tpz1 controls telomerase action in fission yeast Mansi Garg, Sahar Mansoubi, Resham Lal Gurung, Steve Plumb, Felicity Watts, Alessandro Bianchi 22 Keynote Session 1 The repression of PARP1 at telomeres Titia de Lange, Isabelle Schmutz The Rockefeller University; New York, USA Mammalian telomeres have structural features that could potentially result in the binding and activation of PARP1 and PARP2. Presumably, activation of these PARPs would be deleterious and would require repression. Our prior work with shelterin-free telomeres had revealed the activation of alternative non-homologous end-joining (alt-NHEJ), a pathway that was shown to be promoted by PARP1 and DNA ligase 3 and inhibited by Ku70/80 (Sfeir and de Lange, Science 2012). Initial data showed that while TRF2 is one of the shelterin subunits required for the repression of alt-NHEJ, it is not the only shelterin factor with this function. We were able to directly examine the localization and activation of PARP1 at telomeres. With these assays for PARP1 activation, we queried conditional mouse embryo fibroblasts to determine which components of shelterin contribute to the repression of PARP1 signaling at chromosome ends. The data indicate that TRF2 and TIN2 act independently to minimize the localization of PARP1 to telomeres and reduce its activation by dysfunctional telomeres. In contrast, TRF1, Rap1, TPP1, or POT1a/b were not needed for the control of PARP1. We will present data on the mechanism by which TRF2 represses PARP1 signaling. Presented by: de Lange, Titia 23 Session 1-1 Riff’ing on the final step of telomere segregation at anaphase Sophie Zaaijer, Nadeem Shaikh, Julia Promisel Cooper National Cancer Institute, NIH, Bethesda, MD and Cancer Research UK, London Rif1 has been implicated as a regulator of several processes fundamental to telomere maintenance – chromosome replication, DSB resection and telomerase-mediated telomere extension. We have uncovered an additional telomeric role for Rif1, later in the cell cycle, that prompts an assessment of Rif1’s fundamental mode of action and suggests a final, regulated step in chromosome segregation. Cells lacking Taz1 suffer stalled telomeric replication forks that result in telomere entanglements that fail to be resolved at mitosis at cold temperatures. Hence, taz1 cells are cold sensitive. Entangled telomeres become apparent as DNA masses persisting between the separating sister chromatids during M-phase. These masses are associated with persistent DNA polymerase alpha foci as well as Rad11RPA and Rad22Rad52, indicating ongoing attempts to deal with entanglements as mitosis progresses. Rif1 sparked our interest when we observed that rif1+ deletion suppresses taz1 cold sensitivity. We have found that Rif1 plays a role in removal of telomeric entanglements rather than the fork-stalling events that generate them, and while Rif1’s telomere binding is reported to depend on Taz1, we find that Rif1 co-localizes with telomeres in the anaphase mid-zone in both wild type and taz1∆ cells. This localization is independent of the presence of spindle microtubules. We present a model in which Rif1 regulates the fate of ssDNA overhangs generated by prolonged replication fork stalling and in turn, the final steps of sister chromatid resolution. Presented by: Cooper, Julia Promisel 24 Session 1-2 The role of telomere cohesion in aging and ALT cells Susan Smith, Mahesh Ramamoorthy The Skirball Institute, NYU School of Medicine, New York, USA Resolution of telomere cohesion in human cells relies on the poly(ADP-ribose) polymerase tankyrase1 that localizes to telomeres in late G2/early mitosis to resolve cohesion and allow progression through mitosis. We found that this “normal” system of telomere resolution is corrupted in aging and ALT cells; they display excess persistent cohesion in mitosis and subsequent anaphase delay. Since a hallmark of ALT is loss of the chromatin remodeler ATRX, we assessed its role in telomere cohesion. Reintroduction of ATRX expression in ALT cells restored normal resolution of telomere cohesion. Conversely, ATRX depletion (like that of tankyrase1) in telomerase positive cancer cells induced persistent telomere cohesion. ATRX is in complex with the free pool of macroH2A histone variants. We show that, dependent on its catalytic PARP activity, tankyrase1 binds specifically to macroH2A1.1, the poly(ADPribose) polymer-binding isoform. In ALT cells (in the absence of ATRX) the free pool of macroH2A1.1 sequesters tankyrase1, rendering it unable to resolve cohesion. Overexpression of tankyrase1 overrides the macroH2A1.1 inhibition and rescues persistent telomere cohesion in ALT cells, but with dire consequences: a dramatic and immediate increase in telomere copying to other chromosomes and impaired cell growth. A similar effect on cell growth was observed in aging fibroblasts. Our studies suggest that keeping sister telomeres in close proximity into mitosis, not only promotes recombination between sister telomeres, but also prevents excessive illegitimate recombination between nonhomologous chromosomes. This may be particularly important in the recombination permissive ALT cell setting and in aging cells as telomeres become critically short. Presented by: Smith, Susan 25 Session 1-3 Telomere Looping: A new paradigm for the regulation of gene expression with progressive telomere shortening Jérôme D. Robin1, Andrew T. Ludlow1, Min Chen2, Frédérique Magdinier3, Kimberly Batten1, Brody Holohan1, Guido Stadler1, Kathyrin R. Wagner4, Jean-Marie Rouillard5, Jerry W. Shay1, Woodring E Wright1 1 Dept. Cell Biology, UT Southwestern Medical Center, Dallas TX 75390 USA Dept. Molecular Biophysics, UT Southwestern Medical Center, Dallas TX 75390 USA 3 Aix Marseille University, INSERM, UMRS 910. Epigenetics, Chromatin and Diseases team, 27 Bd Jean Moulin, Marseille 13385 cedex 05 France 4 Center for Genetic Muscle Disorders, Kennedy Krieger Institute, Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205 USA 5 Biodiscovery, LLC, Ann Arbor, MI 48105 2 Studies of human telomeres have focused on their role as a tumor-suppressor dependent on DNA damage signals from too-short telomeres. We here describe a new paradigm for how telomere shortening can progressively affect gene expression during the human lifespan, long before telomeres become short enough to generate damage signals. ISG15 (Interferon Stimulated Gene 15 kda) is 1 Mb from the telomere and its expression is regulated by telomere length. Many genes between it and the telomere are not affected by telomere shortening. We here demonstrate that the distance between the telomere and ISG15 is small when telomeres are long but the loci separate when telomeres are short. We call this phenomenon TPE-OLD (Telomere Position Effects-Over Long Distances). We next developed high-resolution Hi-C to directly map TPE-OLD interactions in a disease whose locus is adjacent to the telomere of 4q and which exhibits an age-associated phenotype (FacioScapuloHumeral muscular Dystrophy: FSHD) and show multiple interactions, which can extend at least 10 Mb into the subtelomeric region. Global analysis of expression then established that 100s of changes can be regulated by telomere length. Our results suggest new mechanisms for how telomere shortening could influence human aging and disease initiation/progression long before shortening produced DNA damage signals. These changes could be local, affecting stem cell behavior, inflammation and repair. They could also be global, for example where secretory signals from glial cells dividing on a regular basis every seven years could influence hormonal signals governing energy distributions throughout the body. We have found TPE-OLD effects in both myoblasts and fibroblasts, and are investigating its manifestations in epithelial cells. Length- regulated long-range (Mb) telomere chromatin conformation changes may alter gene expression to optimize fitness in species such as humans that live for many decades, and where the optimization of energy utilization may differ significantly between newly mature and aged individuals. These changes may profoundly affect human physiology, aging and disease. Presented by: Wright, Woodring E. 26 Session 2-1 A TERT promoter gradient enables the isolation and purification of spermatogonial stem cells Steven Artandia,*, Matthew Pecha, Alina Garbuzova, Bérénice Benayouna, Meena Sukhwania, Shengda Lina, Stephanie Brockmana, Anne Bruneta, Kyle Orwigb, Jenny Zhanga a b Stanford University University of Pittsburgh Germline mutations in the telomerase pathway cause a diverse set of diseases, including dyskeratosis congenita, pulmonary fibrosis, aplastic anemia and liver cirrhosis. Telomerase expression is controlled in part at the level of transcription of TERT. The ability to identify and isolate TERT-positive cells in vivo would significantly advance our understanding of telomerase function. To address these issues, we have created knock-in transcriptional reporters of TERT expression by replacing the TERT open reading frame with genes encoding fluorescent proteins. Among mouse tissues, telomerase activity is most strongly expressed in testis, a tissue in which resident stem cells fuel the continuous generation of transit amplifying cells, which in turn enter meiosis to produce the male gametes. In the human testis, telomere lengths are preserved with age, although how this is achieved, in contrast to the aging-dependent telomere shortening seen in somatic tissues, remains unresolved. We determined the TERT-reporter expression in the stem cell, transit-amplifying cell and meiotic populations of the testis by wholemount and FACS analysis. The combination of TERT-Reporter activity together with other surface markers allows for purification of each of these cell populations in spermatogenesis by FACS, enabling each of these populations to be isolated for molecular analysis. Our data provide important new insights into how telomeres are maintained in the mammalian germline, preventing telomere attrition that might otherwise precipitate telomere shortening disease phenotypes. This approach may enable a greater understanding of telomerase regulation during carcinogenesis and in normal tissues during aging. Presented by: Artandi, Steven 27 Session 2-2 Telomere dysfunction accurately predicts clinical outcome in two common cancers Duncan Baird, Thet Lin, Kevin Norris, Nicole Heppel, Kate Simpson, Guy Pratt, Robert Hills, Rhiannon Jones, Julia Grimstead, Bethan Britt-Compton, et al. Cardiff University Defining the prognosis of individual cancer sufferers remains a significant clinical challenge. Telomere length has been identified as a prognostic marker in a number of human cancers but it is not routinely used in clinical practice. Here we provide a definition of the threshold length at which telomeres become dysfunctional and show that this transforms telomere analysis into the most prognostic marker of survival in patients with two common malignancies. Using a cohort of chronic lymphocytic leukaemia (CLL) patients we undertook telomere fusion analysis to define the upper telomere length limit at which fusion could be detected. We found that CLL patients with telomeres within the fusogenic range had a significantly shorter overall survival (P<0·0001; HR=13.2 (11.6-106.4)) and importantly this was particularly prognostic in early-stage disease patients (HR=19.3 (17.8-802.5), P<0·0001). Remarkably, the same threshold was also highly prognostic in a cohort of breast cancer patients (HR=18.2 (13.9-242.0), P<0·0001). In both cohorts the fusogenic mean was the dominant variable in multivariate analysis. Finally we investigated whether telomere dysfunction could predict response to therapy. Using the fludarabine/cyclophosphamide arm of the LRF CLL4 clinical trial cohort we showed that the upper limit of telomere dysfunction strongly predicted for shorter progression-free survival and overall survival. Our findings identify the loss of telomeric functional integrity as a critical factor in the pathology of two unrelated malignancies. Routine assessment of this parameter should provide more reliable prognostic information for individual patients suffering from these diseases. Presented by: Baird, Duncan 28 Session 2-3 The complex and differing roles of TIN2 isoforms in telomere regulation Nya Nelson, Alison Bertuch Baylor College of Medicine, Department of Molecular and Human Genetics TIN2 is a central component of the shelterin complex, linking the telomeric DNA binding proteins TRF1 and TRF2 with TPP1/POT1. Heterozygous mutations in TINF2, which encodes TIN2, are found in the telomere disorder dyskeratosis congenita (DC). The mutations, including truncations, cluster in a region shared by both the short (TIN2S) and long (TIN2L) TIN2 isoforms. We hypothesized that TIN2L has functions not shared with TIN2S that could be impacted by DC-cluster mutations. We found that, in contrast to TIN2S, TIN2L was predominantly phosphorylated in vivo and was phosphorylated in vitro by CK2, at a conserved residue present only in TIN2L. While telomeres progressively elongated in HT1080 cells overexpressing TIN2L or TIN2L with a phosphomimetic mutation, telomere length remained static with overexpression of TIN2S or TIN2L with a phosphodead mutation. In co-immunoprecipitation assays, we found that TIN2L had increased interaction with TRF2 compared to TIN2S, and the DCcluster and TIN2L phosphorylation cooperated to promote TRF2 interaction. In contrast to TIN2S, TIN2L bound TRF2 via residue F120, which is also required for TRF2-Apollo interaction. This suggests TIN2L may regulate Apollo recruitment. Conversely, TRF1 interacted more robustly with TIN2S than TIN2L. DC-cluster mutations had no effect on TIN2S-TRF1 interaction, but further reduced TIN2L-TRF1 interaction. Both isoforms interacted similarly with TPP1. These results indicate that the composition of shelterin is likely complex, not only because of stoichiometric differences as previously described, but also due to differences in TIN2S and TIN2L activity, and that this composition may be fundamentally different in patients with TINF2 mutations. Presented by: Nelson, Nya 29 Session 2-4 The function and execution of RTEL1 activities at vertebrate telomeres Grzegorz Sarek, Jean-Baptiste Vannier, Steffi Panier, Simon Boulton London Research Institute, Clare Hall Regulator of telomere length 1 (RTEL1) is an essential DNA helicase that disassembles T-loops and suppresses telomere fragility to maintain the integrity of chromosome ends. The emergence of RTEL1 variants that confer increased susceptibility to high-grade glioma, astrocytomas and glioblastomas has highlighted the importance of RTEL1 for genome stability. Mutations in RTEL1 have also been implicated in Hoyerall-Hreidarsson syndrome (HHS), a severe form of the bone marrow failure and cancer predisposition disorder, Dyskeratosis Congenita. We previously reported that RTEL1 binds to the replisome via a PIP-box dependent interaction with PCNA. Disruption of the RTEL1-PCNA interaction in mice compromised genome-wide and telomere replication, and accelerated tumourigenesis. Unexpectedly, the RTEL1-PCNA interaction was found to be dispensable for T-loop disassembly, which suggested the existence of a distinct mechanism for recruiting RTEL1 to telomeres. Indeed, we have discovered that RTEL1 is recruited to telomeres to promote T-loop disassembly via an S-phase specific interaction with Shelterin. We identify a single point mutation within Shelterin that abrogates the interaction with RTEL1 and phenocopies the Rtel1 null phenotype. Finally, we show that the RTEL1Shelterin interaction is compromised by a mutation in RTEL1 present in a subset of HHS patients. These results establish a dynamic interplay between RTEL1 and Shelterin, which is critical for the timely disassembly of T-loops during S-phase of the cell cycle. Presented by: Boulton, Simon 30 Session 2-5 Dyskeratosis congenita caused by an OB fold mutation in TPP1 Hande Kocaka, Bari Ballewb, Kamlesh Bishta, Joseph Bolandc, Belynda Hicksc, Neelam Girib, Blanche b, Alterb, Catherine Keegana, Jayakrishnan Nandakumara, Sharon Savage , et al. * a University of Michigan National Cancer Institute c Cancer Genomics Research Laboratory, NCI-Frederick b Dyskeratosis congenita (DC), the prototypic telomere biology disorder, is diagnosed by the triad of dysplastic nails, skin pigmentation, and oral leukoplakia, or by leukocyte telomere lengths <1st percentile for age. Patients are at high risk of bone marrow failure, cancer, pulmonary fibrosis, liver disease, and other complications. Causative germline mutations in one of nine telomere biology genes (DKC1, TERC, TERT, TINF2, NOP10, NHP2, WRAP53, CTC1, or RTEL1) are present in ~70% of DC. We performed whole exome sequencing on mutation-negative DC families participating in our IRBapproved longitudinal cohort study using the Illumina HiSeqTM platform. We focused on low frequency, high quality coding variants, and explored all inheritance patterns. The likelihood of a nonsynonymous variant being deleterious was predicted using information from multiple algorithms. The proband of family NCI-275 is a 5 year-old boy with Hoyeraal Hreidarsson syndrome, a severe variant of DC that includes cerebellar hypoplasia, immunodeficiency and intra-uterine growth retardation in addition to DC features. The proband, his older sister, and father have telomeres significantly <1st percentile; all three also harbor a single-amino acid deletion within the TEL patch of the shelterin component TPP1. His healthy twin sisters and mother have telomeres at the 1st percentile and 10th to 50th percentiles, respectively, and do not have the TEL patch mutation. This deletion severely compromises the processivity of telomerase and its recruitment to telomeres. Structural modeling of the mutation using the crystal structure of TPP1-OB as a template reveals disruptive conformational changes, supporting a causative role of this mutation in DC. Presented by: Savage, Sharon 31 Session 2-6 Role of telomeres in disease and longevity Maria A. Blasco Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain Over the past years our laboratory has contributed to dissect the role of telomerase and telomere length as key molecular pathways underlying cancer and aging, as well as has addressed the potential use of telomerase activation as a therapeutic strategy for telomere syndromes and age-related diseases (Blasco et al., Cell, 1997; Tomás-Loba, Cell, 2008). More recently, we have developed a telomerase-based gene therapy strategy that allows telomerase activation in adult organisms and that has shown beneficial effects in a variety of age-related pathologies in mice (Bernardes de Jesus et al., EMBO Molecular Medicine, 2012). I will present recent findings showing the efficacy of this telomerase gene therapy in mouse models of disease. Finally, I will discuss recent findings from our group regarding generation of mice with long telomeres in the absence of genetic modifications. Presented by: Blasco, Maria A. 32 Session 2-7 Disease-specific manifestations of the telomere syndromes Mary Armanios Department of Oncology and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore MD USA The telomere syndromes encompass a group of inherited degenerative disease states that share short telomere length as a defect. These disorders affect both children and adults and have overlapping phenotypes. In children, the predominant manifestation of telomere syndromes is bone marrow failure which is at times seen in the context of features of dyskeratosis congenita. In adults, lung disease usually manifests alone as idiopathic pulmonary fibrosis and, because of its prevalence in the population, it is estimated to account for at least 90% of telomere syndrome cases. The biology that underlies these differential presentations and the mechanisms that underlie the telomere-mediated pulmonary phenotype will be the topic of this presentation. Presented by: Armanios, Mary 33 Session 3-1 Regulated assembly and disassembly of the yeast telomerase quaternary complex Timothy Tucey, Vicki Lundblad Salk Institute for Biological Studies Telomerase from budding yeast consists of the catalytic Est2 protein and two regulatory subunits (Est1 and Est3) in association with the TLC1 RNA, with each of the four subunits essential for in vivo function. Using a biochemical approach that permits quantitative assessment of each of the protein subunits of telomerase, we have monitored the holoenzyme, as well as sub-complexes, through the cell cycle. This has revealed a hierarchy of assembly and disassembly, which results in limiting amounts of the telomerase quaternary complex late in the cell cycle. Formation of the quaternary complex is dependent on interaction of Est3 in late S phase/early G2 with a previously formed Est1-TLC1-Est2 sub-complex. Est3 association is dependent on separate binding sites on both Est1 and Est2, suggesting that Est1 and Est2 form a single interface that is bound by Est3, as well as a regulatory surface on Est3 itself that functions as an apparent toggle switch. The involvement of multiple surfaces that regulate Est3 binding suggests that it sits at a regulatory node for assembly. Surprisingly, once formed, the quaternary complex disassociates through loss of the catalytic Est2 subunit, indicating that assembly and disassembly of the telomerase complex proceed by different pathways. These studies reveal a previously unappreciated means by which telomere homeostasis can be modulated, through a complex and apparently highly regulated process of telomerase assembly and disassembly. Presented by: Lundblad, Vicki 34 Session 3-2 Two-step mechanism of telomerase recruitment and activation in fission yeast Christine Armstrong, Siân Pearson, Kazunori Tomita UCL Cancer Institute, University College London, London, WC1E 6DD, United Kingdom. Current models depict telomerase activation to be achieved by the presence of telomerase at telomeres. In fission yeast, telomerase recruitment is achieved through interaction of the telomerase subunit Est1 with Ccq1, a component of the Pot1 telomere binding complex. Our data show that the association of telomerase with Ccq1 alone is not sufficient to fully activate telomerase. The OB fold domain of Tpz1, a human TPP1 ortholog, positively regulates telomerase activity/processivity after association of telomerase with the Ccq1 complex. In tpz1-K75A mutants, telomeres are critically short even though telomerase is recruited. However, telomerase activity can be restored by fusing Tpz1K75A with the catalytic subunit of telomerase, Trt1. We therefore propose the Ccq1-Est1 interaction to be the recruitment process and the OB-fold of Tpz1 to regulate telomerase activity, presumably through the interaction of Trt1 with Tpz1. These two processes are separable. These findings reveal a layer of telomerase regulation that controls processivity after recruitment. Presented by: Tomita, Kazunori 35 Session 3-3 Assembly, Architecture and Function of Human Telomerase Kathleen Collins University of California, Berkeley Reconstitution of minimal recombinant active telomerase in vitro can require only the templatecontaining telomerase RNA subunit (TER) and the telomerase reverse transcriptase protein (TERT). In cells, additional chaperone and telomerase holoenzyme proteins direct the process of TER biogenesis and folding, catalytic activity and telomere elongation. Dissecting the cellular requirements for TER and TERT structure and associated factors has been complicated by overlapping and interlaced subunit roles. We are using a wide array of approaches to investigate the biochemical specificity and mechanisms that underlie unique features of the telomerase catalytic cycle, intimate protein and RNA domain co-folding, enzyme activity on telomere substrates, and RNP regulation over the cell cycle. Presented by: Collins, Kathleen 36 Session 3-4 The essential role of TPP1 in telomerase dependent telomere maintenance Alec Sexton, Kathy Collins, Dirk Hockemeyer University of California, Berkeley Department of Molecular & Cell Biology Telomeric binding proteins regulate the recruitment of telomerase to telomeric DNA in human stem and tumor cells, thereby establishing telomere length homeostasis as necessary for their immortal phenotype. TPP1 has been previously implicated in facilitating this interaction. Specifically, a region in the OB-fold of TPP1, called the TEL patch, mediates TPP1’s ability to interact with the telomerase holoenzyme and to stimulate its catalytic processivity in vitro. However, the physiological role of this interaction in telomere maintenance is still unclear. Here, we use genome editing of the endogenous TPP1 locus to demonstrate that the TEL patch of TPP1 is essential for telomere maintenance in pluripotent stem cells (hPSCs). Genetically engineered hPSCs carrying homozygous substitutions in the TEL patch of TPP1 (TPP1ΔL) phenocopy telomerase deficient TERT-/- hESCs, undergoing telomere shortening and eventually cell death. TERT-/- cells can be complemented by ectopic TERT expression but not TERTR132D, an allele which fails to interact with TPP1. Conversely, TPP1ΔL/ΔL hPSCs can be rescued by wild type TPP1. We used this complementation assay to test a panel of TPP1 sequence substitutions and fusion proteins to understand the overall pathway of telomerase function at telomeres. By demonstrating the essential role of TPP1 in the telomere maintenance pathway this data provides a unique target to interfere with telomerase activity in tumor cells. Presented by: Hockemeyer, Dirk 37 Session 3-5 Identification of the site on telomerase that binds TPP1 to promote recruitment to telomeres Jens C Schmidta,*, Andrew B Dalbyb, Thomas R Cechc a BioFrontiers Institute University of Colorado Boulder, Damon Runyon Cancer Research Foundation BioFrontiers Institute University of Colorado Boulder c BioFrontiers Institute University of Colorado Boulder, Howard Hughes Medical Institute b Human chromosomes end in protein-DNA complexes termed telomeres, containing G-rich repetitive DNA sequences, which bind the six protein shelterin complex. The shelterin complex carries out multiple functions: it protects chromosome ends, prevents them from being recognized as sites of DNA damage and recruits telomerase, an RNA-containing reverse transcriptase that synthesizes telomeric repeat DNA. Recently, a group of amino acids on the OB-fold domain of the shelterin component TPP1, termed the TEL patch, was demonstrated to be essential to recruit telomerase to telomeres (Nandakumar et al 2012, Zhong et al 2012). In contrast, the site on telomerase that interacts with the TPP1 OB-fold has remained poorly defined. Here we identify separation-of-function mutations in the telomerase protein component hTERT that disrupt the interaction of telomerase with the OB-fold domain of TPP1 in vivo and in vitro but have no effect on the catalytic activity of telomerase. The mutant telomerases fail to localize to telomeres in vivo, their processivity is not stimulated by POT1/TPP1 in vitro and they are incapable of maintaining telomere length in cultured cells. We will provide evidence that these residues define the site on telomerase that directly binds to the TPP1-OB fold. Presented by: Schmidt, Jens C 38 Session 3-6 A self-regulating template in human telomerase Julian J.-L. Chen, Andrew F. Brown, Xiaodong Qi, Yinnan Chen, Joshua D. Podlevsky, Mingyi Xie Arizona State University, Department of Chemistry and Biochemistry, Tempe, Arizona 85287, USA Telomerase is a specialized reverse transcriptase (RT) containing an intrinsic telomerase RNA (TR). It synthesizes telomeric DNA repeats, (GGTTAG)n in humans, by reiteratively copying a precisely defined, short template sequence from the integral TR component. The specific mechanism of how the telomerase active site utilizes this short template region accurately and efficiently during processive DNA repeat synthesis has remained elusive. Here we report that the human TR template, in addition to specifying the repeat sequence, is embedded with a single-nucleotide signal to pause DNA synthesis. After the addition of a dT residue to the DNA primer, specified by the signaling residue 49 rA in the template, telomerase extends the DNA strand with three additional nucleotides and then pauses DNA synthesis. This sequence-defined pause site coincides precisely with the structurally defined template boundary, and cooperatively precludes incorporation of non-telomeric nucleotides from residues outside the template region. Additionally, this sequence-defined pausing mechanism prevents premature arrest of nucleotide synthesis and is the predominate mechanism for generating the characteristic 6-nt ladder banding-pattern of telomeric DNA products in vitro. In the absence of the pausing signal, telomerase stalls nucleotide addition at multiple sites along the template, generating DNA products with diverse repeat registers at the termini. Our findings demonstrate a unique selfregulating mechanism of the human TR template for high fidelity synthesis of DNA repeats. Presented by: Chen, Julian J.-L. 39 Session 4-1 Negative regulation of telomerase by SUMOylation-dependent Tpz1(TPP1)-Stn1 interaction in fission yeast Ross Lowa, Bettina Mosera, Alice Hoylea, Ya-Ting Changa, Keisuke Miyagawab, Katsunori Tanakab, Toru Nakamura*a, a Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA b Department of Bioscience, Kwansei Gakuin University, Sanda 669-1337, Japan Studies in fission yeast have previously identified conserved shelterin and Stn1-Ten1 complexes, and established Rad3(ATR)/Tel1(ATM)-dependent phosphorylation of the shelterin subunit Ccq1 at Thr93 as the critical modification for telomerase recruitment. The shelterin subunits Poz1 and Rap1 negatively regulate Thr93 phosphorylation and telomerase recruitment by promoting the timely recruitment of the lagging strand DNA polymerase to limit Rad3 checkpoint kinase accumulation. Intriguingly, ChIP analyses have also revealed that Poz1 and Stn1 show nearly identical temporal recruitment patterns with slightly delayed peak binding compared to Tpz1(TPP1)/Ccq1/Trt1(TERT). We have also found that the shelterin subunit Tpz1(TPP1) interacts with Stn1-Ten1, based on yeast threehybrid (Y3H) assay. However, it remained unclear if Tpz1(TPP1)-Stn1 interaction plays a role in telomere length regulation, and if Poz1’s ability to enforce the timely lagging-strand synthesis might be dependent on Tpz1(TPP1)-Stn1 interaction. To address these questions, we have identified a tpz1 mutation (tpz1-K242R) that significantly weakens the Tpz1(TPP1)-Stn1 interaction, and determined that this mutation causes significant telomere elongation, major reduction in late S/G2-phase binding of Stn1, and massive increase in late S/G2- phase binding of Trt1(TERT). We also established that Lysine 242, which occurs within a SUMOylation consensus sequence motif, is indeed SUMOylated in S/G2-phase. Southern blot analysis also found that pmt3D (SUMO deletion mutant), tpz1-K242R, and pmt3D tpz1-K242R cells show similar extent of telomere elongation, indicating that SUMOylation of Tpz1 at K242 negatively regulates telomere extension. Our results thus suggest that SUMOylationdependent Tpz1(TPP1)-Stn1 interaction facilitates Stn1 recruitment to telomeres and timely dissociation of telomerase to limit telomere extension in late S/G2-phase. Presented by: Nakamura, Toru 40 Session 4-2 Roles of human CST in telomere replication and genome-wide replication rescue Jason Stewarta, Feng Wanga, Anukana Bhattacharjeea, Carolyn Priceb,* a b University of Cincinnati Dept. Cancer Biology, University of Cincinnati CST (CTC1-STN1-TEN1) is the mammalian homolog of the Cdc13-Stn1-Ten1 complex from S. cerevisiae. Human CST has evolved various roles in telomere and non-telomeric DNA replication. During telomere replication, CST first facilitates replication of the duplex DNA and then enables fill-in synthesis of the C- strand following telomerase action. CST also aids is replication recovery following genome-wide replication stress. Cells depleted of CST display decreased nucleotide uptake and reduced firing of late or dormant origins after HU-induced replication fork stalling. In the absence of exogenous replication stress, cells show increased anaphase bridges without telomere fusions. We have developed a separation of function mutant that rescues CST-depleted cells from the defects in C-strand fill-in and new origin firing in response to HU but not defects in telomere duplex replication or anaphase bridge formation. Our results demonstrate that CST is needed to rescue cells from multiple forms of replication stress and that the transactions involved in this rescue differ with the type of replication problem. We have also shown that CST helps cells recover after treatment with diverse DNA damaging agents that impair replication. Cells with reduce CST show decreased recovery while cells that overexpress CST have enhanced recovery. Since the agents evoke different repair pathways, our results are consistent with CST acting at a common step during subsequent re-start of DNA replication. Overall, our findings indicate that while CST may use the same final mechanism to rescue replication following diverse challenges to the replication machinery, the actual pathway to this replication rescue varies. Presented by: Price, Carolyn 41 Session 4-3 Rif1 controls DNA replication timing and anti-checkpoint functions through recruitment of the PP1 phosphatase, Glc7 Maksym Shyiana, Stefano Mattaroccia, Laure Lemmensa, Tianlai Shib, Nicolas Thomäb, David Shorea,* a b Department of Molecular Biology, University of Geneva Friedrich Miescher Institute for Biomedical Research, Basel The Rif1 protein, originally identified as a budding yeast telomere-binding factor controlling the telomerase pathway, has more recently been implicated in conventional DNA replication control, and in the DNA damage response, from yeast to mammals. Here we will describe experiments showing that budding yeast Rif1 interacts through two short N-terminal motifs (RVxF and SILK) with the PP1 phosphatase (Glc7 in yeast) to carry out both replication and anti-checkpoint functions. More specifically, we will show how Rif1-Glc7 inhibits the activation of pre-replication complexes (pre-RCs) through regulation of two different Dbf4-dependent kinase (DDK) substrates. In addition, we will describe experiments that implicate Rif1-Glc7 in an anti-checkpoint function at both telomeric and non- telomeric double-strand breaks (DSBs). Given the apparent conservation of the Rif1-PP1 interaction, we propose that these studies in yeast will be highly relevant to Rif1 function in higher eukaryotes. Presented by: Shore, David 42 Session 5-1 Long-term proliferation of human cancer cells in the absence of telomere length maintenance Loretta Laua, Rebecca Dagga, Jeremy Hensonb, Hilda Pickettb, Axel Neumannb, Roger Reddelb,* a b The Children's Hospital at Westmead, Westmead, NSW 2145, Australia Children's Medical Research Institute, Westmead, NSW 2145, Australia Cellular immortalization is a hallmark of cancer, and requires an active telomere length maintenance mechanism (TMM). Every immortalized cancer cell line studied to date has telomerase or Alternative Lengthening of Telomeres (ALT) activity. We have now identified two neuroblastoma (NB) cell lines that proliferate extensively (>300 population doublings [PDs]) despite continuous telomere shortening. Their telomeres start out very long (mean terminal restriction fragment [TRF] length of 31 to 37 kb) and steadily decrease in length at a rate of 50 – 80 base pairs/PD without any evidence of compensatory lengthening events. The telomeres appear very similar to the long and heterogeneous telomeres of ALT cell lines, with the exception that individual telomere bands are visible on Southern blot analysis, in contrast to the typical smeared TRF pattern of ALT cell lines. This unique pattern has only been observed previously in ALT cell lines where ALT activity has been switched off by genetic manipulation. These cells are telomerase-negative, but, unlike ALT cells, they lack mutations in p53, ATRX, DAXX, H3.3 and IDH1/2, lack T-circles and ALT-associated PML nuclear bodies, and have very low C-circle levels. Moreover, we have identified a subset of NB tumours (~10% of high-risk NBs) which have characteristics similar to these cell lines, and found that patients with these tumours have better prognosis than those with telomerase or ALT. These cell lines (and, most likely, cancers) therefore undergo very longterm proliferation despite failing to maintain telomere length. Presented by: Reddel, Roger 43 Session 5-2 Absence of telomerase causes the early onset of cancer in zebrafish Madalena Carneiro, Joana Nabais, Tania Carvalho, Miguel Godinho Ferreira Instituto Gulbenkian de Ciência, Oeiras, Portugal Does telomere-shortening act as a tumour suppressor or an oncogene-type mechanism? In human somatic cells, telomerase expression is restricted and age-progressive telomere attrition occurs with each cell division. As tumour suppressors, critically short telomeres trigger p53 activation and restrain cell proliferation. As “oncogenes”, short telomeres are themselves sources of genome instability and tumorogenesis. Humans with telomerase deficiencies and shorter telomeres are tumour-prone attesting to the possibility that short telomeres may be conducive to cancer. Zebrafish has recently emerged as a complementary cancer model exhibiting increased tumourogenesis in later life. Unlike lab mice, zebrafish have human-size telomeres that decline with age and we have shown that ztert-/- fish die prematurely displaying several ageing phenotypes. We now report that ztert-/- fish exhibit an anticipation of tumourogenesis that mimics both the rates and types of older fish. Spontaneous tumours arise in young ztert-/- fish in a context of an overall decline in cell proliferation, increased cell senescence and inflammation. These tumours exhibit characteristics of ALT. In parallel, we are characterizing the role of telomerase in an established model for melanoma in zebrafish. Transgenic fish expressing human BRAF-V600E in melanocytes develop invasive melanoma in a p53-/- mutant background within the first year of age. Absence of telomerase in this model significantly reduces the incidence of melanoma. However, in contrast to spontaneous tumours, the timing of melanoma formation in this model is unaffected. Our results thus suggest that exceedingly short telomeres provoke the early onset of tumours in zebrafish depending on the status of p53. Presented by: Ferreira, Miguel Godinho 44 Session 5-3 The role of SUMO pathway in telomere recombination Ferose Charifia, Dmitri Churikova, Nadine Eckert-Bouletb, Sonia Silvab, Marie-Noelle Simona, Michael Lisbyb, Vincent Géli*a, a b Cancer Research Center of Marseille University of Coppenhaguen In budding yeast, sumoylation of the telomere-associated and DNA repair proteins is thought to promote sequestration of the telomeres and persistent DNA double-strand breaks to the nuclear periphery, where canonical homologous recombination (HR) is repressed. We have previously shown that eroded telomeres that cannot be extended are shifted from their perinuclear location to the nuclear pore complexes (NPCs). The SUMO protease Ulp1 also localizes at the NPC and may contribute to channelling damage to alternative repair. We have analyzed the dependence of the two HR- dependent survivor pathways in telomerase-negative cells on Rad52 sumoylation and telomere relocalization to NPC. We found that sumoylated Rad52 promotes Rad51-dependent type I, whereas Rad52 desumoylation (presumably by Ulp1 at the NPC) favours type II survivor formation. Interestingly, we found that telomere shift to NPC was dependent on Rad59, whereas Rad51 played no role in this process. To test the importance of telomere relocalization to NPC for the type II pathway, we introduced LexA binding sites in the vicinity of telomere VI-R, and expressed LexA-fused proteins to either target VI- R to the NPC or to locally modulate sumoyalation at VI-R. Our results suggest that NPC targeting affects the dynamics of telomere recombination. Also, targeting Ulp1 catalytic domain to a specific telomere was found to be sufficient to induce its recombination even in the presence of telomerase. We will present our current view of the role of the SUMO pathway in telomere recombination. Presented by: Géli, Vincent 45 Session 5-4 Interchromosomal homology searches drive directional ALT telomere movement and synapsis Roger Greenberg Department of Cancer Biology Perelman School of Medicine, University of Pennsylvania Telomere length maintenance is a requisite feature of cellular immortalization and a hallmark of human cancer. While most human cancers express telomerase activity, approximately 10-15% employ a recombination-dependent telomere maintenance pathway known as Alternative Lengthening of Telomeres (ALT), an incompletely understood process that is characterized by multi-telomere clusters and associated promyelocytic leukemia protein bodies. Here, we show that a DNA double-strand break (DSB) response at ALT telomeres triggers long-range movement and clustering between chromosome termini, resulting in homology-directed telomere synthesis. Damaged telomeres initiate increased random surveillance of nuclear volumes before displaying rapid directional movement and association with recipient telomeres over micron-range distances. This phenomenon required homologous recombination proteins and the Hop2-Mnd1 heterodimer, which is essential for homologous chromosome synapsis during meiosis. These findings implicate a meiotic-like homology searching mechanism in ALT dependent telomere maintenance and provide a molecular basis underlying the preference for recombination between non-sister telomeres during ALT. Presented by: Greenberg, Roger 46 Session 5-5 Genome instability driven by targeted telomere insertion in ALT cells Paulina Marzeca,*, Claudia Armenisea, Gaelle Perotb, Christina Raftopoulouc, Sarantis Gagosc, Frederic Chibonb, Jerome Dejardina a Institute of Human Genetics, CNRS UPR1142,141 rue de la Cardonille,34090 Montpellier, FRANCE Translational Research, Department of Biopathology, Institut Bergonié, 229 cours de l’Argonne, 33076 Bordeaux, FRANCE c Laboratory of Genetics and Gene Therapy, Center of Basic Research II, Biomedical Research Foundation, of the Academy of Athens, GREECE b The Breakage-Fusion Bridge cycle is a classical mechanism of telomere driven genome instability, in which critically short or unprotected telomeres are fused to other chromosomal extremities, creating dicentric chromosomes which eventually break at mitosis. We uncover a new mechanism of telomere driven genome instability, specifically occurring in cells which have activated the Alternative Lengthening of Telomeres pathway. In these cells, we show that telomeric DNA is added to multiple discrete sites throughout the genome. These sites correspond to regions usually regulated by NR2C/F transcription factors, and we show that these factors drive local telomere DNA addition by recruiting telomeric chromatin. This new mechanism, which we name targeted telomere insertion (TTI), generates potential common fragile sites that destabilize the genome. We propose that targeted telomere insertion driven by NRC2/F proteins creates complex karyotypes in ALT tumors. Presented by: Marzec, Paulina 47 Session 5-6 Alternative Lengthening of Telomeres is characterized by reduced compaction of telomeric chromatin Harikleia Episkopoua,*, Irena Draskovicb, Amandine Van Benedena, Gaëlle Tilmana, Marina Mattiussia, Matthieu Gobina, Nausica Arnoulta, Arturo Londoño-Vallejob, Anabelle Decottigniesa a Genetic and Epigenetic Alterations of Genomes, de Duve Institute, Catholic University of Louvain, Brussels 1200, Belgium b Telomeres and Cancer Laboratory, Equipe Labellisée Ligue, UMR3244-UPMC-Institut Curie, Paris 75248, France Proper telomeric chromatin configuration is thought to be essential for telomere homeostasis and stability. Previous studies in mouse suggested that loss of heterochromatin marks at telomeres might favor onset of Alternative Lengthening of Telomeres (ALT) pathway, by promoting homologous recombination. However, analysis of chromatin status at human ALT telomeres has never been reported. Here, using isogenic human cell lines and cellular hybrids, which rely either on telomerase or ALT to maintain telomeres, we show that chromatin compaction is reduced at ALT telomeres and this is associated with a global decrease in telomeric H3K9me3. This, subsequently, leads to upregulation of telomere transcription. Accordingly, restoration of a more condensed telomeric chromatin through telomerase-dependent elongation of short ALT telomeres reduces telomere transcription. We further show that loss of ATRX chromatin remodeler function, a frequent characteristic of ALT cells, is not sufficient to decrease chromatin condensation at telomeres nor to increase the expression of telomeric RNA species. These results offer new insight on telomeric chromatin properties in ALT cells and support the hypothesis that telomeric chromatin decondensation is important for ALT pathway. Presented by: Episkopou, Harikleia 48 Session 5-7 A complex interplay between telomere transcription, stability and recombination in human ALT cells Rajika Aroraa,*, Yongwoo Leea, Harry Wischnewskia, Catherine Brunb, Claus Azzalina a b Eidgenössische Technische Hochschule Zürich (ETHZ), Institute of Biochemistry The Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario We previously generated telomerase-positive HeLa cell lines where transcription of one specific telomere (‘transcriptionally inducible telomere’: tiTEL) can be induced experimentally. Transcription induction did not alter tiTEL length or stability, nor did it affect telomerase-mediated elongation of tiTELs. We have now generated tiTEL-containing U2OS cells, which maintain their telomeres through the ‘alternative lengthening of telomeres’ (ALT) pathway. Transcription induction in U2OS cells leads to increased rates of tiTEL fragility, sister tiTEL exchanges and accumulation of RNA:DNA hybrids at tiTELs. These data suggest that in an ALT background, TERRA transcription compromises telomere replication and promotes homologous recombination between telomeric sequences, possibly by engaging in RNA:DNA hybrids at telomeres (‘telomeric R-loops’; telR-loops). Consistent with this hypothesis, different ALT cell lines exhibit increased rates of TERRA transcription from CpG island subtelomeric promoters and of telomere fragility as compared to telomerase-positive cells. Over-expression of TRF1 in U2OS cells diminishes telR-loops and telomere fragility, while siRNA-mediated depletion of RNaseH1 increases telR-loops levels, telomere fragility, telomere loss, sister telomere exchanges and extrachromosomal telomeric circles. Strikingly, depletion of RNaseH1 in HeLa cells has only a minor impact on telomere homeostasis. We propose that telR-loops are dynamic structures appearing at human telomeres and their improper resolution, possibly during cell cycle progression, might compromise replication of telomeric sequences and trigger homologous recombination or telomere loss. TERRA transcription-mediated telomere recombination may be a mechanism utilized by ALT cells to maintain their telomeres in the face of telomere integrity. Presented by: Arora, Rajika 49 Session 5-8 NuRD-ZNF827 recruitment to telomeres creates a molecular scaffold for homologous recombination Dimitri Conomos1,2, Roger R. Reddel2,3, and Hilda A. Pickett1,2 1 Telomere Length Regulation Group, Children’s Medical Research Institute, Westmead, New South Wales, Australia 2 Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia 3 Cancer Research Unit, Children’s Medical Research Institute, Westmead, New South Wales, Australia Alternative lengthening of telomeres (ALT) is an homologous recombination (HR)-dependent mechanism for de novo synthesis of telomeric DNA. Nuclear receptors are bound to the telomeres of cells that utilize ALT. Here we demonstrate that nuclear receptors recruit ZNF827, a zinc finger protein of unknown function, which recruits the nucleosome remodeling and histone deacetylation (NuRD) complex via binding to an N-terminal RRK motif within ZNF827. This results in hypoacetylation of telomeric chromatin, decreased shelterin binding, a diminished telomeric DNA damage response (DDR) as well as enhanced telomere-telomere interactions and recruitment of HR proteins, and is critically important for cell viability and proliferation. We propose that NuRD-ZNF827 recruitment to telomeres establishes a molecular platform which remodels telomeric chromatin and creates an environment which promotes telomere-telomere recombination, and integrates and controls multiple mechanistic elements of ALT activity. Presented by: Pickett, Hilda 50 Session 5-9 TRF2 is required to protect heterochromatic pericentromeric regions from topological and replicative DNA damage Aaron Mendez Bermudeza,*, Karine Jameta, Jing Yeb, Eric Gilsona a Institute for Research on Cancer and Aging, Nice (IRCAN), CNRS UMR 7284 - INSERM U1081 Medical School, University of Nice, France b Shanghai Ruijin Hospital affiliated to Shanghai Jiaotong University, School of Medicine, China Previous results from our lab showed that TRF2 binds to a subset of interstitial telomeric sequence (ITS) and pericentromeric satellite III (SatIII) DNA (Simonet et al, 2011) and that it is required to protect a stretch of telomeric sequences artificially inserted in the interior of chromosome 4 against replicative DNA damage (Ye et al, 2010). In this work, we investigate whether TRF2 could also be involved in the protection of naturally occurring extratelomeric sequences. First, chromatin immunoprecipitation coupled with deep sequencing (ChIP-seq) analysis revealed that, in addition to telomeric sequences, a reduced expression of TRF2 triggers γ-H2AX accumulation at pericentromeric SatIII regions. In addition, we showed by immunofluorescence that cells suffering from topological stress caused by topoisomerase downregulation (either Top1 or TopIIa or TopIIB) or by treatment with the topoisomerase II inhibitor ICRF193 exhibit an increased binding of TRF2 to the pericentromeric SatIII region of chromosome 9 and also an increased in DNA damage. Conversely, in cells treated with TSA or compromised for the histone methyltransferase responsible for H3K9me3, Suv39H1, the binding of TRF2 to pericentromeric SatIII repeats is reduced. Finally, in support of a role for TRF2 in the protection of pericentromeric chromatin against replicative DNA damage, treatment of cells with hydroxyurea and aphidicolin as well as inhibition of the BLM and WRN helicases increases the binding of TRF2 to SatIII regions. Together, these findings suggest that TRF2 is recruited to heterochromatic pericentromeric regions to alleviate the topological problems occurring during their replication. Presented by: Mendez Bermudez, Aaron 51 Session 5-10 The multifaces of chromatin assembly, a recipe that mixes new with old partners Genevieve ALMOUZNI Institut Curie / CNRS Our dissection of the mechanisms of chromatin assembly, from the basic structural unit, the nucleosome, up to higher-order structures in the nucleus has enabled to characterize key chaperones involved in nucleosome assembly as well as to define the dynamics of new histone incorporation in chromatin. We have examined specific nuclear domains: non-coding centromeric heterochromatic regions, which are of major importance for chromosome segregation.Our findings have shed light on the fundamental issues of the dynamics, fate, and inheritance of histones, with their specific marks typical of particular chromatin domains. Our current hypothesis is that histone chaperones function in an ‘assembly line’ with specificity for individual histone variants to mark defined regions of the genome. Remarkably, we have found that misregulation of specific histone chaperones is a common feature of aggressive breast cancers. Our recent studies show that the depletion of the histone chaperone ASF1 leads to the induction of the characteristics of Alternative Lengthening of Telomeres. We will discuss our most recent advances in the regulatory pathways that target histone chaperones and variants to control the assembly line and its connecting network. Casanova M. et al. (2013) Cell Rep., 4, 1156-1167. Abascal F. et al. (2013) Mol. Biol. Evol., 30, 18531866. Adam S. et al. (2013) Cell, 155, 94-106. Szenker E. et al. (2013) In Fundamentals of Chromatin, J. Workman & Abmayr S. Eds., Springer Verlag, pp. 375-427. O'Sullivan R.J. et al. (2014) Nature Struct. Mol. Biol., 21, 167-174. Lacoste N. et al. (2014) Mol. Cell, 53, 631-644. Presented by: ALMOUZNI, Genevieve 52 Session 6-1 Characterization of the changes in telomeric chromatin upon DNA damage induction Cristina Bartoccia, Jolene Diedrichb, Yates Johnb, Eros Lazzerini Denchia,* a b Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA When telomeres become critically short DNA damage response factors are recruited at chromosome ends initiating a cellular response to DNA damage. Here, we performed Proteomic Isolation of Chromatin fragments (PICh) to define the changes in chromatin composition that occur upon onset of acute telomere dysfunction triggered by depletion of the telomere-associated factor TRF2. This unbiased isolation and purification of telomere-associated proteins in functional or dysfunctional conditions revealed the dynamic changes in chromatin composition that take place at telomeres upon DNA damage induction. Our analysis identified several proteins that are lost from telomeres following DNA damage induction, as well as several proteins that localize to dysfunctional telomeres. Based on our chromatin purification and mass spectrometry analyses, we describe a critical role for the polycomb group protein Ring1b in NHEJ-mediated end-to-end chromosome fusions in response to telomere dysfunction. Our data suggest that cells with reduced levels of Ring1b have a reduced ability to repair uncapped telomeric chromatin. Our data represent the first unbiased isolation of chromatin undergoing DNA damage and represent a valuable resource to map the changes at the level of chromatin composition in response to DNA damage activation. Presented by: Lazzerini Denchi, Eros 53 Session 6-2 Mitotic telomere deprotection dictates cellular fate upon mitotic arrest Makoto T Hayashia,*, Anthony J Cesareb, Jan Karlsedera a b Salk Institute for Biological Studies. La Jolla, California 92037 Children’s Medical Research Institute. Sydney, Australia. 2145 The DNA damage response activated at shortened telomeres avoids tumorigenesis by inducing senescence. Upon the bypass of senescence, cells enter a crisis, during which further telomere shortening acts both tumor suppressive and tumor promoting, by inducing cell death and chromosome instability, respectively. Although telomere fusions and the resulting genome instability are wellcharacterized features of the crisis, the mechanism underlying cell death has not been fully understood. Our finding that prolonged mitotic arrest induces telomere deprotection prompted us to ask whether mitotic telomere deprotection has a physiological function. Here, we found that IMR-90 E6E7 cells that bypassed senescence display a spontaneous mitotic arrest phenotype in the pre-crisis cells. The arrest phenotype was induced by loss of p53, and suppressed by telomerase overexpression, indicating extreme telomere shortening as the underlying cause. However, telomere fusions induced by overexpressing dominant-negative TRF2 did not cause a mitotic arrest phenotype in middle-aged IMR-90 E6E7, suggesting fusions do not directly trigger mitotic arrest. The actual cause of the spontaneous mitotic arrest is under investigation. Pre-crisis cells with premature sister chromatid separation, which is a consequence of mitotic arrest, suffer from increased telomere deprotection, suggesting pre-crisis cells also undergo mitotic telomere deprotection. Exacerbation of mitotic telomere deprotection by partial knockdown of TRF2 increased the cell death ratio during mitosis and sensitized cells to drugs that induce mitotic arrest. We propose that mitotic telomere deprotection functions to promote cell death during mitotic arrest in both cells that bypassed senescence, and cells that are exposed to mitotic drugs. Presented by: Hayashi, Makoto T 54 Session 6-3 An optimal telomere length protects human tumor cells from ionizing radiation Jennifer Fairliea, Lea Harrington*b, a University of Edinburgh, School of Biological Sciences, Wellcome Trust Centre for Cell Biology University of Montreal, Institute for Research in Immunology and Cancer Visiting Professor, University of Edinburgh, School of Biological Sciences b The acquisition of telomerase reverse transcriptase (TERT) expression and its ability, together with the telomerase RNA, to maintain telomeres at chromosome ends is one of the hallmarks of cancer. More than 85% of all human cancers possess telomerase activity, and telomerase inhibition has been established in several model systems to be a tractable target for cancer therapy. As human tumor cells typically maintain short equilibrium telomere lengths, we wondered if excessive telomere elongation would impact the fitness of human tumor cells. Indeed, we found that telomere elongation beyond a certain length significantly decreased cell clonogenic survival after gamma irradiation. Susceptibility to irradiation was dosage-dependent and increased at telomere lengths >17 kbp despite the fact that all chromosome ends retained telomeric DNA. We postulate that human cancer cell survival may be selected around an optimal telomere length that balances the maintenance of telomere integrity and efficient DNA replication. Presented by: Harrington, Lea 55 Session 6-4 Interactions between CST, DDR and NMD genes show that Stn1 and Ten1 are the key components of the CST complex Eva Holstein, Kate Clark, David Lydall Institute for Cell and Molecular Biosciences, Newcastle University Medical School, Newcastle upon Tyne, UK A large and diverse set of proteins, including CST complex, Nonsense Mediated Decay (NMD) and DNA Damage Response (DDR) proteins play important roles at the telomere in mammals and yeast. Here we show that NMD, like the DDR, affects single stranded DNA (ssDNA) production at uncapped telomeres. We also show that the requirement for Cdc13, one of the components of CST, can be efficiently bypassed when aspects of DDR and NMD pathways are inactivated. However, identical genetic interventions do not bypass the need for the other CST components, Stn1 and Ten1. We show that disabling NMD alters the stoichiometry of CST components at telomeres and permits Stn1 to bind telomeres in the absence of Cdc13. Our data supports a model that Stn1 and Ten1 cap the telomere in a Cdc13-independent manner and have implications for the function of CST components across eukaryotes. Presented by: Clark, Kate 56 Session 6-5 Delineating the role of upstream DNA damage responses in cell and tissue aging induced by telomere shortening Karl Lenhard Rudolph Leibniz Institute for Age Research, Fritz-Lipmann-Institute (FLI), Jena, Germany Telomere shortening limits the proliferative capacity of cells and the regenerative capacity of tissues by induction of DNA damage checkpoints. Telomere dysfunction can also contribute to cancer initiation by inducing chromosomal fusions, fusion-bridge-breakage cycles and chromosomal instability (CIN). Late generation telomerase knockout mice (G3 Terc-/-) with shortened telomeres exhibit premature aging and chromosomal instability. In yeast, Exo1-dependent DNA end resection at dysfunctional telomeres mediates induction of DNA damage responses. The deletion of Exo1 improved tissue maintenance and prolonged lifespan of telomere dysfunctional mice. The study indicated that Exo1 dependent end resection contributes to both the induction of DNA damage checkpoints and chromosomal fusions in response to telomere shortening in mammalian cells. It is currently not known which process – endresection induced DNA damage checkpoints or chromosomal fusions - is mediating the adverse effect of telomere shortening on tissue aging. In my talk I will present how specific genes that regulate the upstream induction of DDRs influence the proliferative capacity of cells, tissue maintenance, and oncogenic transformation in response to telomere shortening. Presented by: Rudolph, Karl Lenhard 57 Session 6-6 Identification of a critical regulator of DNA repair activity at mammalian telomeres Vera Boersmaa, Sandra Segura-Bayonaa, Marieke Peuschera, Jaco van der Torrea, Nathalie Moattia, Alexandre Orthweinb, Daniel Durocherb, Jacqueline Jacobsa,* a The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON M5G 1X5, Canada b Appropriate repair of DNA lesions and inhibition of repair activities at telomeres are critical for the maintenance of genome integrity and prevention of genomic instability. Upon loss of telomere protection natural chromosome ends are processed by DNA repair factors that cause chromosome end-to-end fusions. Upon cell division these fusions result in missegregation of chromosomes and unbalanced chromosomal rearrangements that can compromise cell viability and promote cancer development. Similarly, inaccurate or inappropriate repair of DNA lesions can contribute to aging or tumorigenesis by fueling genetic alteration. The mechanisms underlying control of DNA repair activities are not completely understood. Therefore, our laboratory undertakes candidate-driven and unbiased approaches to identify factors with important roles in the responses to telomere dysfunction. In particular, we performed loss-of-function genetic screens in p53-deficient mouse embryo fibroblasts in which we inactivate TRF2 to induce telomere uncapping. While this results in a chromosome fusion phenotype that becomes so severe that cells arrest or die, we identified multiple shRNAs that allow cells to survive and proliferate despite prolonged telomere uncapping. We have focused our validation and follow-up efforts on several screen hits not previously known to act at telomeres and linked to diverse activities, including ubiquitylation, methylation, control of mitosis and DNA damage tolerance. In this process we identified a new factor that promotes NHEJ-mediated chromosome end-to-end ligation and telomere-induced genomic instability by inhibiting 5’ DNA-end-resection. Moreover, this factor is also essential for efficient end-joining of DNA DSBs during immunoglobulin class switch recombination. Presented by: Jacobs, Jacqueline 58 Session 6-7 DNA polymerase theta promotes alternative-NHEJ at dysfunctional telomeres Pedro Mateos-Gomeza, Fade Gongb, Kyle Millerb, Eros Lazzerini-Denchic, Agnel Sfeira,* a Skirball Institute, NYU Langone Medical Center, New York, NY, USA Institute for Cell and Molecular Biology, UT Austin, Austin, Tx, USA c The Scripps Research Institute, San Diego, CA, USA b Mammalian cells have evolved complex mechanisms to repair double strand breaks (DSBs). In addition to the error-free homology-directed repair (HDR), DSBs can be rejoined by the two error-prone pathways of classical Non-Homologous End-Joining (NHEJ), and alternative-NHEJ. The engagement of end-joining pathways at the end of mammalian chromosomes is detrimental, and is inhibited by the six-subunit shelterin complex. While classical-NHEJ is primarily blocked by TRF2, alt-NHEJ is only unleashed when telomeres loose the binding of the entire shelterin complex (shelterin-free), and in cells lacking Ku70/80. To define the molecular differences between the two distinct end-joining pathways, we applied deep sequencing technology on telomeric DNA isolated either from TRF2 null mouse embryonic fibroblasts (MEFs), or shelterin-free/Ku80 deficient MEFs. Telomeres fused by alt-NHEJ were marked by nontelomere insertions that scar the junction between fused ends. Based on this analysis, we tested the function of a number of translesion synthesis DNA polymerases and identified polymerase theta (PolQ) as a key alternative-NHEJ factor. PolQ is a poor fidelity A-family DNA polymerase with an activity that appears to be template independent, and known to be preferentially expressed in cancers cells. Inhibiting PolQ blocked telomere fusions in shelterin-free/Ku80 deficient MEFs, but did no impact classical-NHEJ fusions in cells lacking TRF2. In addition to its role at telomeres, PolQ depletion impaired alternative-NHEJ repair when tested in the context of non-reciprocal chromosomal translocation. In summary, our study underscores a novel function for the highly promiscuous PolQ, potentially accounting for the increased mutagenicity of the alternative-NHEJ pathway of DNA repair. Presented by: Sfeir, Agnel 59 Session 6-8 Rapamycin prevents checkpoint adaptation via autophagy and Cdc5 (PLK1), which increases proliferative potential following telomere dysfunction Julia Klermund, Katharina Bender, Brian Luke University of Heidelberg, ZMBH The DNA damage checkpoint provides a quality control function for the genome by preventing cells with damaged DNA from continuing in the cell cycle. In response to dysfunctional telomeres, yeast cells arrest at the G2/M border due to the activation of a Mec1 (ATR) / Rad53 (Chk2) – mediated arrest, which prevents the onset of mitosis. When damaged telomeres persist for longer periods without repair, the checkpoint is eventually extinguished and cells continue to cycle even in the presence of damage – a phenomenon referred to as checkpoint adaptation. Checkpoint adaptation is typically accompanied by the de-phosphorylation of Rad53 and the progression of cells into the following G1 phase of the cell cycle. Surprisingly, the inhibition of the conserved TOR signaling pathway, through rapamycin addition, is able to prevent cells with dysfunctional telomeres from adapting, thereby establishing crosstalk between the cellular nutritional status and the DNA damage response. Indeed, in the presence of rapamycin Rad53 phosphorylation remains high and cells do not enter the following G1 phase. Importantly, the rapamycin-mediated inhibition of checkpoint adaptation increases cell viability following long term telomere dysfunction from 30% to over 80%. We have found that rapamycin prevents checkpoint adaptation via two parallel pathways 1. Through the induction of autophagy and 2. Via inhibition of Cdc5 (PLK1) – mediated inactivation of Rad53. Finally, we were able to demonstrate that rapamycin extends chronological lifespan, in yeast, in a DNA damage checkpointdependent manner. Therefore nutrient status has a critical influence on cell cycle regulation following telomere dysfunction. Presented by: Luke, Brian 60 Session 6-9 A role for DNA damage response RNAs (DDRNA) at dysfunctional telomeres Francesca Rossielloa,*, Julio Aguadoa, Fabrizio d'Adda di Fagagnab a IFOM – the FIRC Institute for Molecular Oncology Foundation, Milan, Italy IFOM – the FIRC Institute for Molecular Oncology Foundation, Milan, Italy CNR – Consiglio Nazionale delle Ricerche, Pavia, Italy b We have recently shown that small non-coding RNAs are novel components of the DNA damage response (DDR) machinery (Francia et al. Nature 2012). These small RNAs, named DDR RNAs or DDRNAs, have the sequence of the damaged locus and their biogenesis depends on Dicer and Drosha endoribonucleases. DDRNAs are necessary for DDR activation at DNA double-strand breaks (DSBs) following ionizing radiations, endonuclease cleavage or oncogene-induced DNA replication stress. Moreover, a role for Dicer and Drosha has been proposed for DSB repair (Wei et al. Cell 2012). Deprotected telomeres resemble DSBs as they lead to DDR signaling activation and DNA repair events. We investigated the potential role of DDRNAs in DDR activation at telomeres. Upon TRF2 inactivation, we show that transient RNase A treatment of permeabilized mouse cells impairs DDR foci at telomeres and DDR signaling can be restored by reintroducing cellular RNA. Consistently with the DDRNA biogenesis, Dicer or Drosha knock down prevents DDR activation at dysfunctional telomeres. In addition, knock down of either of these two endoribonucleases impairs telomeric fusions upon telomere uncapping. Furthermore the inhibition of telomeric DDRNAs by the use of oligonucleotide molecules with a complementary sequence can prevent accumulation of DDR markers at telomeres and senescent-associated cell cycle arrest in human cells. These data suggest that the full activation of the DDR cascade at uncapped telomeres is RNA-dependent and is controlled by Dicer and Drosha. Presented by: Rossiello, Francesca 61 Session 7-1 Telomere Response to Viral Infection Zhong Denga, Eui Tae Kimb, Nick Stonga, Jayaraju Dheekollua, Zhuo Wanga, Harold Riethmana, Nigel Fraserb, Matthew Weitzmanb, Paul Lieberman*a, a b The Wistar Institute University of Pennsylvania, School of Medicine Telomeres have well-established functions in chromosome-end protection in response to DNA replication stress, but it is not yet known whether telomeres provide protection against other forms of nuclear stress, like viral infection. Viruses typically subvert host-cell regulatory mechanisms that function in anti-viral capacities. Here, we show that acute infection by herpes simplex virus type 1 (HSV-1) causes massive changes in telomere activity and structure. We found that HSV-1 induces a rapid increase in telomere repeat-containing RNA (TERRA) transcripts. This TERRA induction is accompanied by dissociation of telomere repeat binding factors (shelterin), and ultimately the selective loss of telomere repeat DNA. The HSV-1 encoded E3 ubiquitin ligase ICP0 contributes to TERRA induction, telomere repeat factor dissociation, and proteasome-dependent degradation of telomere protection protein TPP1. We also find that the HSV-1 replication factor ICP8, a single-stranded DNA binding protein, colocalizes with telomere DNA foci and binds to subtelomere DNA proximal to telomere repeats. In addition, ICP8-mediates viral replication compartments assembly at sites of telomere DNA foci. Furthermore, shRNA depletion of TPP1 accelerates viral replication. These findings reveal that viral infection can extensively remodel cellular telomeres, and further suggest that telomeric factors can play a broader role in restricting and regulating viral infection in the nucleus. Presented by: Lieberman, Paul 62 Session 7-2 Regulation and Functions of TERRA ncRNA Antonio Porro, Sascha Feuerhahn, Joachim Lingner, et al. EPFL The long noncoding RNA TERRA sustains several telomere functions. In Saccharomyces cerevisiae, TERRA transcription promotes telomere end shortening by exonuclease 1. In this organism, TERRA has also been implicated in seeding the formation of nucleoplasmic telomerase clusters, which precedes association of these clusters with short telomeres for their extension. On the other hand, TERRA is a potent inhibitor of at least human telomerase in vitro but this effect may be prevented in vivo by TERRA binding proteins such as hnRNPA1 or by downregulation of TERRA during S phase. TERRA has also been suspected to mediate the exchange of single strand telomere binding proteins upon telomere replication. Finally, TERRA may help recruiting proteins that modulate telomeric chromatin and DNA processing enzymes. By RNA-IP, we recently identified the LSD1 lysine demethylase as TERRA binder and in ChIP and Q-TIP experiments we see LSD1 at telomeres. We provided evidence that TERRA and LSD1 cooperate with the MRE11 nuclease at telomeres that are uncapped by depletion of TRF2. We propose a neat 3-step mechanism that guides removal of single-stranded terminal telomeric DNA at uncapped telomeres. The mechanism involves telomere transcription into TERRA, TERRA-guided recruitment of the LSD1 demethylase to MRE11 at uncapped telomeres and LSD1-mediated activation of the DNA nuclease activity of MRE11. I will report on the mechanisms that lead to upregulation of the TERRA transcriptome when TRF2 is depleted and TERRA interactions with chromatin modifying enzymes. Presented by: Lingner, Joachim 63 Session 7-3 Identification of novel telomere regulators through systematic genetic screens in yeast Dimitris Kyriakoua, Emmanouil Stavroua, Panayiota Demosthenousa, Bryan-Joseph San Luisb, Charlie Booneb, Antonis Kirmizisa,* a Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada b Pervasive transcription is a feature of eukaryotic genomes resulting in numerous non-coding transcripts. Recent high-throughput studies in S. cerevisiae using hybridization to DNA-tiling arrays have uncovered the expression of several long intergenic non-coding RNAs (lincRNAs). In an attempt to illuminate the biological function of selected lincRNAs we have employed a synthetic genetic array (SGA) technique. Using this assay we expect to identify genetic interactions by generating systematically double mutants of a lincRNA with a genome-wide library of protein-coding genes. Since the SGA procedure has been previously applied only on protein-coding genes we first validated our approach by performing a screen for the yeast telomerase RNA (TLC1) that represents an example of a lincRNA with a known function. Indeed, we identified and validated several synthetic genetic interactions between TLC1 and genes whose biological functions associate with telomere organization. In particular, we identified genetic interactions between TLC1 and the telomeric Ku complex, DNA recombination factors that maintain telomere length and the INO80 nucleosome-remodeling complex, which has been recently implicated in the control of chromatin structure at telomeres. Interestingly, TLC1 also genetically interacts with other chromatin regulators, such as various histone modifiers whose role in telomere organization remains unknown. Among the other lincRNAs screened, one of them (SUT457), genetically interacts with genes involved in telomere maintenance, implicating this lincRNA within this process. Overall, our findings demonstrate that SGA can be utilized to point towards the biological function of lincRNAs in yeast and in the above examples identify new candidates that control telomere structure. Presented by: Kirmizis, Antonis 64 Session 7-4 Human RTEL1 regulates the export and cytoplasmic trafficking of ncRNAs, including telomerase RNA Michael Schertzera, Rosa Maria Porrecaa, Karina Jouravlevaa, Mylène Perderiseta, Florent Dinglib, Damarys Loewb, Tangui Le Guenc, Jean Pierre De Villartayc, Patrick Revyc, Arturo Londono-Vallejoa,* a Telomeres and Cancer lab, Institut Curie Laboratory of Proteomic Mass Spectrometry, Institut Curie c INSERM, U768 b Hoyeraal-Hreidarsson syndrome (HHS) is a severe form of Dyskeratosis congenita characterized by developmental defects, bone marrow failure, and immunodeficiency and has been associated with telomere dysfunction. Recently, mutations in RTEL1, a helicase first identified in Mus musculus as being responsible for the maintenance of long telomeres, have been identified in several HHS patients. We show that RTEL1 interacts with exportins XPO1, XPO5, and XPOT and is required for the export and correct cytoplasmic trafficking of non-coding (nc) RNAs, including pre-U2, a component of the major spliceosome complex, and TERC, the telomerase RNA. While reconstitution of telomerase activity is deficient in RTEL1-HHS cells, these cells show abnormal subcellular partitioning of TERC and pre-U2, defects in the recycling of ribonucleotide proteins (RNP) in the cytoplasm, phenotypes that can be experimentally induced by the expression of RTEL1 variants carrying mutations in the helicase and Cterminal domains. Our work unravels broad and totally unanticipated roles for RTEL1 in RNP export and trafficking, thus leaving open the possibility that general defects in RNP biogenesis contribute to the pathology of HHS. Presented by: Londono-Vallejo, Arturo 65 Session 7-5 AKTIP, an E2 variant enzyme that interacts with lamin, is required for correct telomere maintenance Mattia La Torrea,*, Romina Burlaa, Mariateresa Carcuroa, Grazia Daniela Raffaa, Alessandra Galatia, Domenico Raimondob, Angela Rizzoc, Laura Ciapponia, Giovanni Cencid, Enrico Cundarie, et al. a Dipartimento di Biologia e Biotecnologie, Sapienza, Università di Roma, 00185, Italy Istituto Pasteur Fondazione Cenci Bolognetti, Sapienza, Università di Roma, 00185, Italy b Dipartimento di Fisica, Sapienza, Università di Roma, 00185, Italy c Istituto Nazionale Tumori Regina Elena, Rome 00144, Italy d Dipartimento di Biologia e Biotecnologie, Sapienza, Università di Roma, 00185, Italy e Istituto di Biologia e Patologia Molecolari del CNR, Sapienza, Università di Roma, 00185, Italy Telomeric DNA challenges cell metabolism at multiple levels requiring the shelterin complex and the telomere accessory factors for chromosome end protection and for correct telomere replication. We report here on AKTIP, an ubiquitin E2 variant enzyme, that we have identified as required for mammalian telomere metabolism. AKTIP was investigated on the basis of its homology with the Drosophila telomere-capping protein Pendolino. AKTIP interacts with purified TRF1 and TRF2, and immunoprecipitates telomeric DNA. Loss of AKTIP/Ft1 results in fragile telomeres and sister telomere associations, which, in turn, have been associated to telomere replication problems, as clearly assessed in TRF1-depleted cells. In doubly depleted TRF1/AKTIP cells, the TRF1-induced fragile telomere phenotype was epistatic to that of AKTIP, suggesting that TRF1 and AKTIP are involved in a common molecular pathway. In fact, in AKTIP depleted cells, telomeric replication is impaired, and AKTIP interacts with the DNA replication factors PCNA and RPA70. Intriguingly, AKTIP locates at the nuclear periphery, the site of late replicating heterochromatic DNA, and interacts with lamins. In vivo, the depletion of the mouse homologue of AKTIP, Ft1, causes severe developmental abnormalities and premature death. Our results suggest that AKTIP/Ft1 plays a crucial role in telomere maintenance presumably acting in lamin-associated replication factories. Presented by: La Torre, Mattia 66 Session 8-1 A billion years of shelterin – conservation of interaction modules and the potential to form higher order structures. Lili Pan2,3, Cian Stutz4, Nicolas Thomä4, Peter Baumann1,2,3 1 Howard Hughes Medical Institute Stowers Institute for Medical Research, Kansas City, MO 64110 3 Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66103 4 Friedrich Miescher Institute for Biomedical Research, Basel, 4058, Switzerland 2 In mammalian cells, the shelterin proteins (TRF1, TRF2, RAP1, TIN2, POT1 and TPP1) are central to telomere protection and length maintenance. The fission yeast Schizosaccharomyces pombe telomere maintenance machinery shares fundamental similarities with the mammalian factors. Taz1 (hTRF1/2 ortholog) interacts with double-stranded telomeric repeats; Rap1 binds to Taz1 as hRAP1 binds to hTRF2 and the Pot1-Tpz1 complex (hPOT1-hTPP1 homologs) bind single stranded telomeric DNA. Finally, Poz1, functionally similar to hTIN2, forms a bridge between Taz1/Rap1 and Pot1/Tpz1. All five proteins play critical yet different roles in telomere length regulation. Deletion of Taz1, Rap1 and Poz1 leads to telomere elongation, while deletion of Tpz1 and Pot1 results in rapid telomere loss. How these proteins work together to maintain telomere length equilibrium is not well understood. The Poz1-Tpz1 interface is of particular interest as it forms the boundary between negative and positive regulators of telomere length. To understand the role of this interaction, we solved the crystal structure of Poz1 bound to Tpz1 at 2.4Å resolution. Surprisingly, the Poz1-Tpz1 complex shares remarkable similarities with the docking motifs in human TRF1 and TRF2 that bind TIN2 and Apollo, respectively. Indeed, the corresponding regions of TRF2 and Apollo can functionally substitute for the fission yeast proteins. Structural and biochemical analysis further revealed a homodimerization interface in Poz1. Mutational analysis indicates that Poz1 homodimerization is critical for telomere length regulation. We speculate that the complexity of interactions between telomeric proteins forms the basis for a higher order architecture with implications for telomere function. Presented by: Pan, Lili 67 Session 8-2 Examining telomere architecture in yeast Nicolas Thomä Friedrich Miescher Institute for Biomedical Research Maulbeerstrasse 66 4051 Basel - Switzerland Budding yeast telomeres comprise irregular TG₁₋₃ DNA repeats bound by the general transcription factor Rap1. Rif1 and Rif2, along with Rap1, form the telosome, a protective cap that inhibits telomerase, counteracts SIR-mediated transcriptional silencing, and prevents inadvertent recognition of telomeres as DNA double-strand breaks. We provide a molecular, biochemical, and functional dissection of the protein backbone at the core of the yeast telosome. The X-ray structures of Rif1 and Rif2 bound to the Rap1 C-terminal domain and that of the Rif1 C terminus are presented. Both Rif1 and Rif2 have separable and independent Rap1-binding epitopes, allowing Rap1 binding over large distances (42110 Å). We identify tetramerization (Rif1) and polymerization (Rif2) modules that, in conjunction with the long-range binding, give rise to a higher-order architecture that interlinks Rap1 units. The Rif1 Nterminus exhibits high apparent affinity for DNA, and is expected to engage telomeric single-stranded and double stranded regions once gaps, or overhangs, arise that that are longer than 50nt. This molecular Velcro relies on Rif1 and Rif2 to recruit and stabilize Rap1 on telomeric arrays and is required for telomere homeostasis in vivo. Parallels and differences between the protein architecture of S.cerevisiae and S.pombe telomeres will be discussed. Presented by: Thomä, Nicolas 68 Session 8-3 Guanine quadruplex structures revisited Peter Lansdorp European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands and Terry Fox Laboratory, BC Cancer Research Centre and University of British Columbia, Vancouver, Canada Reduced telomerase levels in humans result in very short telomeres. Other factors that regulate telomere length were studied by crossing mice with long telomeres (Mus musculus) to mice with short telomeres (Mus spretus). A “telomere elongation” trait was mapped to a region at the tip of chromosome 2. One candidate gene in this region, “novel helicase-like” was studied in C.elegans. Animals without this helicase showed a remarkable “signature” phenotype: genome-wide deletions that start at the 3’end of guanine tracks > 18 nucleotides. We dubbed the gene Deletion of G-rich DNA1 (dog-1) and proposed that the DOG-1 helicase could be required to unwind intramolecular guanine quadruplex (G4) DNA structures arising during lagging strand DNA synthesis. More recent studies in yeast have shown that G-rich DNA is unstable if replicated by leading strand DNA synthesis in the absence of PIF1. The human homolog of dog-1 is FANCJ and “novel helicase-like“ was renamed "Regulator of telomere length-1” or RTEL-1. All these studies implicate G4 structures in genome stability and telomere regulation. Both FANCJ and RTEL-1 are FeS helicases that unwind G4 structures. In order to further study the role of guanine quadruplex structures in biology we recently described monoclonal antibodies to synthetic G4 DNA structures. Surprisingly, one of these antibodies, 1H6, shows bright nuclear staining of most cells suggesting that G4 structures are not formed during replication but are already present in some cells prior to replication. The emerging role of G4 structures in nuclear organization and cell biology will be discussed. Presented by: Lansdorp, Peter 69 Session 8-4 A telomere protective function of human telomerase reverse transcriptase (hTERT), which is independent of catalytic activity Omesha N. Pereraa,*, Michelle F. Maritzb, Anthony J. Cesarea, Karen L. MacKenzieb, Tracy M. Bryana a Children’s Medical Research Institute, Westmead, NSW 2145, Australia Children’s Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, Sydney NSW 2031, Australia b The majority (>85%) of cancers rely on the ribonucleoprotein enzyme telomerase to catalyse the addition of telomeric DNA repeats to counteract telomere shortening, thus conferring cells with an unlimited replicative lifespan. There is growing evidence for a number of non-canonical functions of telomerase, and more specifically the protein hTERT, that are independent of telomere maintenance and catalytic activity. One of these includes a protective function for telomerase in protecting telomeres from DNA damage. We have observed that over-expression of a catalytically inactive (D712A) hTERT in immortal GM639 fibroblasts can reduce the number of telomere dysfunction-induced foci (TIF) in comparison to cells transfected with an empty vector control, to a level similar to that observed in cells over-expressing wild-type hTERT. Interestingly, over-expression of a mutant hTERT which does not localise to telomeres also demonstrated a similar rescue of TIF. This suggests that hTERT potentially activates a telomereindependent signalling pathway that provides protection indirectly. A potential mechanism involves the oxidative stress pathway. Upon increasing intracellular reactive oxygen species (ROS) in HT1080 cells, we observed a doubling in the number of TIF, which was rescued upon over-expression of D712A. Furthermore, depletion of endogenous hTERT decreased levels of the antioxidant, superoxide dismutase 2 (SOD2), which corresponded to increased mitochondrial superoxide levels, as well as a further increase in ROS-induced TIF. These data suggest that hTERT may possess a protective function independent of its catalytic activity, to protect immortal cells from telomeric DNA damage through activation of a telomere-independent signalling pathway. Presented by: Perera, Omesha N. 70 Session 8-5 Structural basis for protein-RNA recognition in telomerase Jing Huang1,2,3, Andrew F. Brown4, Jian Wu1, Jing Xue1, Christopher J. Bley4, Dustin P. Rand4, Lijie Wu1, Rongguang Zhang1, Julian J.-L. Chen4, and Ming Lei1,2,3 1 National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China 2 Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI 48109, USA 3 Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109, USA 4 Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ 85287, USA Telomerase is a large ribonucleoprotein complex minimally composed of a catalytic telomerase reverse transcriptase (TERT) and an RNA component (TR) that provides the template for telomeric DNA synthesis. However, it remains unclear how TERT and TR assemble into a functional telomerase. Here we report the first crystal structure of the conserved regions 4 and 5 (CR4/5) of TR in complex with the TR-binding domain (TRBD) of TERT from the teleost fish Oryzias latipes. The structure shows that CR4/5 adopts an L-shaped three-way-junction conformation with its two arms clamping onto TRBD. Both the sequence and conformation of CR4/5 are required for the interaction. Our structural and mutational analyses suggest that the observed CR4/5-TRBD recognition is common to species from yeast to humans, and CR4/5 in vertebrate TR might play a similar role in telomerase regulation as stem-loop IV in Tetrahymena TR. Presented by: Lei, Ming 71 Keynote Session 2 Regulation of human Immune system CD4+ T cell apoptosis specifically by the RNA component of telomerase Elizabeth Blackburn, Francesca Gazzaniga Department of Biochemistry and Biophysics, University of California San Francisco, CA, USA Genetic deficiencies in telomerase RNA (hTR) levels in humans exacerbate telomere shortening and cause a spectrum of diseases. While loss of telomere protective function can account for the pathology of these inherited diseases, no previous studies have identified any other consequences of the diminished levels of hTR in these patients. T cells are differentiated adult human cells that rapidly upregulate telomerase activity from low basal levels upon mitotic stimulation. We tested whether this upregulation is a consequence of the stimulation response or, conversely, is necessary for T cell proliferation and survival. Surprisingly, knocking down telomerase enzymatic activity level in stimulated CD4+ T cells to an equal extent by depleting either hTR or hTERT produced very different short-term responses. First, the hTERT knockdown caused no effect in the short time frame of the experiments. In contrast, the hTR knockdown reproducibly induced Bim-mediated apoptosis. This apoptosis occurred despite the absence throughout the experimental time-course of detectable p53 activation or telomere shortening or damage. Experiments including utilizing hTR mutated in various known functional regions provided evidence that hTR specifically in a telomerase enzymatically inactive form protects CD4+ T cells from apoptosis, including dexamethasone-induced apoptosis. This is the first report suggesting a new protective role for the non-coding telomerase RNA in normal human cells through a telomere- and telomerase enzymatic-independent pathway. We propose that in addition to having long-term effects on telomere shortening, reduced hTR potentially impacts on immune system functionality through a new route: by perturbing short-term survival of differentiated T cells. Presented by: Blackburn, Elizabeth 72 Poster 1 Detection of DNA damage response RNAs (DDRNAs) at dysfunctional telomeres Julio Aguadoa,*, Francesca Rossielloa, Fabrizio d'Adda di Fagagnab a The FIRC Institute of Molecular Oncology Foundation, Via Adamello 16, 20139 Milan, Italy -The FIRC Institute of Molecular Oncology Foundation, Via Adamello 16, 20139 Milan, Italy -Istituto di Genetica Molecolare – Consiglio Nazionale delle Ricerche, Via Abbiategrasso 207, 27100 Pavia, Italy b Our lab has recently shown that small non-coding RNAs are novel components of the DNA damage response (DDR) machinery (Francia et al. Nature 2012). These small RNAs, named DDRNAs, are involved in the control of the DNA damage response (DDR) activation, have the sequence of the damaged locus and their biogenesis depends on Dicer and Drosha endoribonucleases. Upon TRF2 inactivation telomeres are uncapped and a DDR is triggered. Different approaches are being undertaken to detect the potential presence of putative DDRNAs upon telomere uncapping. A method based on qRT-PCR is being developed to detect and characterize DDRNAs at dysfunctional telomeres. We will report on our progresses in our attempts to monitor telomeric DDRNAs accumulation. Presented by: Aguado, Julio 73 Poster 2 RTEL-1 promotes telomere-ITS recombination events Megan Brady, Shawn Ahmed Department of Genetics, Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill NC 27599, USA Telomerase deficiency ultimately results in critically shortened telomeres that are joined to yield endto-end chromosome fusions. Though critically shortened telomeres can be directly ligated via NHEJ or single-strand annealing, they can also recombine with interstitial telomere sequence (ITS) tracts thereby duplicating subtelomeric segments of the genome prior to fusion. We developed a PCR-based assay to study telomere-ITS recombination and found that recombination events occurred within ITS tracts, which are composed of degenerate telomere repeats, irrespective of orientation to the telomere, and at sites of longest perfect homology to the telomere, typically 12 to 30 nt in length. RTEL1 promotes telomere stability, regulates homologous recombination, and is essential for development and cell proliferation in mammals. Deficiency for the C. elegans homolog of RTEL1 helicase, RTEL-1, does not abrogate viability or fertility, allowing us to assess the consequences of RTEL-1 dysfunction in the context of telomerase deficiency. We found that RTEL-1 plays a central role telomere-ITS recombination events, indicating that this helicase not only plays a role in normal telomere homeostasis but also promotes illegitimate recombination events at critically shortened telomeres. Our results are consistent with the proposal that RTEL-1 may promote synthesis-dependent strand annealing events at sites of DNA damage, and imply that telomere-ITS recombination events reflect inappropriate engagement of a major homologous recombination pathway. Presented by: Ahmed, Shawn 74 Poster 3 A simple model for tandem telomeric G-quadruplexes Patrizia Alberti Muséum national d'Histoire naturelle, Paris (France) In human, telomeric DNA is composed of tandem repeats of the GGGTTA motif. The average length of the telomeric 3’-overhang in human chromosomes (100-200 nt) makes this overhang prone to fold into tandem G-quadruplexes (G4), like beads on a string. While the in vivo formation and the role of tandem G4 at telomeres still remain elusive, evidences of the in vitro formation of this kind of structures have been reported in several studies. How stable are these structures? When considering (GGGTTA)7,11,15GGG sequences, able to fold into two, three and four consecutive G4, respectively, I found that the global stability of the formed structures decreased with the number of repeats. This behaviour has already been reported for similar sequences (1), nevertheless it appears to depend on boundary conditions. For exemple, the two- and three-tandem G4 structures formed by the sequences TTA-(GGGTTA)7,11GGG display the same thermal stability (2). Results obtained from an analysis of melting transitions suggested that the G4 units in a tandem structure were not identical nor independend and revealed an unfavourable coupling free energy in the formation of three-tandem G4 structures (2). What makes G4 units in a tandem structure not identical? do the G4 units interact each other? Despite the accumulation of data, a systematic approach to the comprehension of the stability of these kind of structures is still lacking. In this poster, I present a simple model to explain the observed stability of tandem telomeric G4 and its dependence on boundary sequences. (1) Vorlickova et al. (2005) NAR 33, 5851 (2) Petraccone et al. (2011) JACS 133, 20951 Presented by: Alberti, Patrizia 75 Poster 4 Severe liver fibrosis caused by S. mansoni infection in Dkc1m hypomorphic mice Raquel Alves-Paiva, Leandra Ramalho, Cleide Araujo-Silva, Olinda Mara, Vanderlei Rodrigues, Eduardo Rego, Rodrigo Calado University of Sao Paulo Mutations in telomerase complex genes cause telomere shortening and are associated with bone marrow failure, dyskeratosis congenita, aplastic anemia, and liver diseases. Clinically, approximately 7% of DC patients present hepatic disease, such as hepatic fibrosis, cirrhosis, and HCC. We evaluated Dkc1 deficiency, based on Dkc1m hypomorphic mouse model (kindly provided by Davide Ruggero, Sloan-Kettering Institute, USA), to chronic liver damage in a schistosomiasis experimental model. The immune response to eggs of S. mansoni trapped in human tissues causes hepatosplenic disease and liver fibrosis. Telomere length was performed by qPCR and Dkc1m mice had slitghly shorter telomeres in comparison to WT mice. Dkc1m mice WT B6 controls were infected with 30 cercaries by SC injection in the dorsal region. Over the course of infection, Dkc1m mice presented anemia and diarrhea, although gain of body weight was noted. Mice were euthanized 45 days after S. mansoni infection and liver fibrosis intensity was assessed. S. mansoni infection caused liver damage as fibrosis and hepatomegaly and splenomegaly in both infected groups. S. mansoni exposure caused significantly more hepatic fibrosis in Dkc1m mice in comparison to infected WT B6 mice (P<0.0001), as evidenced by Sirius red staining (quantification by ImageJ software). Our findings suggest that Dkc1m mice are more susceptible to liver damage mediated by S. mansoni infection in comparison to WT B6 mice. Mouse model for chronic liver injury using animals with telomerase complex mutations may represent a tool to investigate liver damage in response to chronic liver disease. Presented by: Alves-Paiva, Raquel 76 Poster 5 Identification of telomere dysfunction in Friedreich ataxia Sara Anjomani Virmouni, Sahar Al-Mahdawi, Chiranjeevi Sandi, Hemad Yasaei, Predrag Slijepcevic, Mark Pook Biosciences, SHSSC, Brunel University, Uxbridge, UK In Friedreich ataxia (FRDA), GAA repeat expansion within the first intron of the frataxin (FXN) gene leads to downregulation of FXN, resulting in oxidative stress, mitochondrial iron accumulation and neuronal atrophy. We hypothesised that telomere length might be shortened as a result of oxidative damage in FRDA. To investigate telomere function in FRDA, we initially assessed the telomere length in human and mouse FRDA fibroblasts and we found that both cell types had chromosomes with relatively longer telomeric repeats compared to controls. In contrast, we noted a significant telomere shortening in FRDA leukocyte cells compared to control cells. Consequently, we screened the FRDA fibroblasts for expression of telomerase activity and we identified that the telomerase activity was not present in these cells. We then assessed the co-localisation of PML bodies with telomeres and frequencies of telomere sister chromatid exchange (T-SCE) in these cells. Our results showed significantly higher co-localised PML foci with telomeric DNA and substantially higher T-SCE levels in the FRDA cell lines relative to the controls suggesting activation of an alternative lengthening of telomeres (ALT)-like mechanism. In addition, further analysis revealed an accelerated rate of telomere shortening and a higher frequency of telomere dysfunction-induced γ-H2AX foci in FRDA fibroblasts compared to control cells. Thus, we report the novel finding of telomere dysfunction in FRDA that is characterised by both ALT-like activation and accelerated telomere shortening. Our findings provide novel insights into mechanisms of FRDA molecular disease progression with implications for future FRDA therapy. Presented by: Anjomani Virmouni, Sara 77 Poster 6 Longer telomeres lead to Type II survivors in the absence of telomerase in yeast Usha Aryala,*, Phillipa Mustonb, Edward J Louisa a b University of Leicester University of Nottingham Telomeres in budding yeast Saccharomyces cerevisiae are maintained by telomerase. Loss of telomerase leads to progressive shortening of the telomeres and eventually cellular senescence. However, a few cells adapt and become survivors by lengthening their telomeres via recombination, a process known in humans as the alternative lengthening of telomeres (ALT). Survivors can be of Type I and Type II, characterized by the amplification of the sub-telomeric Y’ repeats or the telomeric TG1-3 repeats respectively. We have shown that starting with longer telomeres at the initiation of telomerase loss leads to a higher proportion of Type II survivors and vice-versa. Here we show that telomerase negative cells with artificially induced long telomeres can produce Type II survivors in the absence of RAD59, a recombination protein thought to be required for Type II survivor pathway and in the absence of the endonuclease SAE2, loss of which predominantly produces Type I survivors from wild type length telomeres. Other gene deletions such as sgs1Δ do not respond in this way. Telomere elongation was achieved by growing RAD59/rad59Δ and SAE2/sae2Δ diploids, heterozygous for telomerase catalytic subunit Est2/est2Δ, in ethanol cultures for a week prior to sporulation for generating telomerase negative survivors with the respective gene knockouts. Cells treated with ethanol display late senescence and a more efficient recovery from crisis. This suggests that longer telomeres favour the Type II survivor pathway, resulting in faster growth recovery after crisis in the absence of telomerase, overcoming some of the previously thought genetic requirements for Type II survival. Presented by: Aryal, Usha 78 Poster 7 Retroviral activation of TERT expression in chicken B-cell lymphomas Karen Beemon, James Justice Johns Hopkins University We are studying the role of TERT at early stages of tumorigenesis in chicken B- cells. Analysis of avian leukosis virus (ALV) proviral integration sites in chicken lymphomas can lead to the discovery of activated oncogenes and inactivated tumor suppressors. Deep sequencing studies of a series of metastatic lymphomas showed that the most common integration sites were in the TERT promoter. Several-fold increases in TERT mRNA levels and telomerase activity were observed in the tumors. In some tumors, these integrations were clonal, suggesting that TERT activation is an early step in tumor induction. Extensive sequencing to identify other ALV integrations in these tumors has been carried out. We are currently asking whether the ALV integration is changing the alternative splicing pattern of TERT in the tumors. We are also asking whether TERT over-expression from a retroviral vector will lead to rapid tumorigenesis in chickens. We will also analyze cooperating genes in these TERT-induced tumors. In the past year, a number of investigators reported point mutations in the human TERT promoter in melanomas, gliomas, and many other types of tumors, especially those derived from cells with low self-renewal (Horn et al, Huang et al, Killela et al., all 2013). These mutations also led to slightly increased expression of TERT mRNA and were thought to be an early event in oncogenesis. Thus, we think our chicken tumor system is a good model for study of the role of TERT in initiation of many types of tumors. Presented by: Beemon, Karen 79 Poster 8 A novel method of analysis of TANK1 enzymatic activity influence on TRF1 DNA-binding Petra Bencúrová, Patrycja Anna Kłos, Ivona Nečasová, Ctirad Hofr Chromatin Molecular Complexes, CEITEC and Faculty of Science, Masaryk University, Brno, CZ-62500, Czech Republic Mammalian telomeres are composed of long TTAGGG tandem repetitive sequences. TRF1 is one of key shelterin proteins involved in telomere maintenance. TRF1 along with TRF2 recruit various proteins to telomeric sites via specific binding to telomeric repeats. Previous works have shown that TRF1 is a strong negative regulator of the length of telomeric DNA. Tankyrase 1 (TANK1) is a protein containing domains with homology to ankyrins and to the catalytic domain of poly (ADP-ribose) polymerase (PARP). Among various functions of TANK1, the best studied is its function at telomeres. TANK1 unprotects telomeres via poly(ADP)-ribosylation of TRF1 which results in an eventual telomeric DNA elongation. Our study is focused on the development of novel fluorescence based method that explores TANK1 influence on protein TRF1 equilibrium DNA-binding and TANK1 reaction kinetics. This work was supported by CEITEC (CZ.1.05/1.1.00/02.0068), the European Social Fund (CZ.1.07/2.3.00/30.0019), GACR (P205/12/0550), and KONTAKT II (LH13054). Presented by: Bencúrová, Petra 80 Poster 9 Impact of cancer treatments in telomere length: A prospective study Carlos Benitez-Buelgaa,*, Lara Sanchez-Barrosob, Maria Mercedes Gallardoc, Maria Apellanizb, Lucia Inglada Perezb,e, K. Yanowskia, Miguel Uriosted,e, Ana Osorioa,e, Maria Antonia Blascoc, Cristina Rodriguez-Antonab,e, Javier Beniteza,e a Human Genetic Group, Spanish National Cancer Research Center (CNIO) Endocrine Cancer Group, Spanish National Cancer Research Center (CNIO) c Telomeres and Telomerase Group, Spanish National Cancer Research Center (CNIO) d Familial Cancer Group, Spanish National Cancer Research Center (CNIO), Network of Research in Rare Diseases (CIBERER) e Network of Research in Rare Diseases (CIBERER), Spanish National Cancer Research Center (CNIO), Madrid 28029, Spain b BRCA1 and BRCA2 play a role in telomere stability. Recently we demonstrated by q-RT-PCR that BRCA1/2 mutation carriers present shorter telomeres in peripheral blood than controls or sporadic breast cancer patients, suggesting that the mutations in these genes are responsible for this event. To test this hypothesis, we are currently performing a prospective study in a series of 241 familial breast and ovarian cancer patients that includes affected and non affected BRCA1/2 mutation carriers, analyzing different parameters (percentage of short telomeres by high throughput quantitative telomere FISH, telomerase activity, oxidative stress, treatment, exogenous factors) that could alter the telomere length. We have observed that, in addition to the BRCA1/2 mutation, telomere shortening is mainly associated with the treatment. To confirm these findings, we have measured telomere length in DNA from peripheral blood of 265 sporadic breasts cancer patients treated since diagnosis with chemotherapy. We have demonstrated that since the beginning there is a telomere shortening that becomes statistically significant around 1216 weeks of treatment. Then we observe that telomeres remain significantly shortened over a period of 1.5 years, after which they start to elongate, back to the telomere length values of the control group with a speed of 0.06 T/S per year. Our results stress the need to perform prospective studies taking in account the treatment status of the patients to get conclusive results about the relation between telomeres and cancer. Presented by: Benitez-Buelga, Carlos 81 Poster 10 Implication of INT6 in telomere stability Maname Benyelles, Vincent Mocquet, Serge Bauwens, Pierre Jalinot Laboratoire de Biologie Moléculaire de la Cellule, Unité Mixte de Recherche 5239, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon, Lyon, France The INT6 protein has been implicated in the breast cancer developpement. As part of the translation initiation factor eiF3, INT6 is necessary for the translation of specific mRNA such as histone mRNA. It has also been involved in the Nonsense Mediated mRNA Decay process. In this latter one, INT6 could be a link between eiF3 and the NMD complex including UPF2 and UPF1. In parallel, INT6 is necessary for a correct replication by favoring the ubiquitination of the MCM7 helicase. Recent data also showed that at the telomeres, UPF1 plays a role in the adequate replication fork progression. Then, UPF1 defects lead to the accumulation of the telomere repeat containing RNA (TERRA) at the telomeres. Based on those data, we hypothesized that INT6 could influence the telomere integrity. Here, by treating U2OS cells with siRNA, we found that INT6 silencing increased total TERRA molecules and that was confirmed by RT-qPCR. These variations were not due to TERRA stability modification. So, we hypothesis that this accumulation could be explained by a telomere instability because INT6 silencing generates telomere dysfunction induced focis (Tifs). Specifically, using a PNA-FISH approach, we observed that the inhibition of INT6 induced telomeres free ends (TFEs) which correlate with telomere length decreasing (Teloblot). In addition, we found fragile telomeres (FTs) which could correspond to replication defects. From all these observations, we propose that the protein INT6 is important for telomere integrity through a replication defect that we have not yet elucidate. Presented by: Benyelles, Maname 82 Poster 11 Control of telomere replication by phosphorylation Carol Cooley, Anoushka Dave, Mansi Garg, Resham Lal Gurung, Aziz Muhamad, Alessandro Bianchi Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK The action of telomerase is strictly linked to conventional DNA replication. We and others have previously shown that telomere length affects replication timing in yeast. In addition, Rif1 is required to specify the replication timing of telomeres. We demonstrate, in both buddying and fission yeast, that the function of Rif1 in DNA replication, at telomeres and elsewhere in the genome, is due to its ability to interact with PP1 phosphatases. In both organisms Rif1 is at least partly responsible for recruiting PP1 to telomeres. We show that, in budding yeast, Rif1-bound PP1 acts to reverse the action of DDK on the MCM helicase. Intriguingly, we find that the early-replication phenotype of short budding yeast telomeres is due to the action of the Tel1 kinase. Several lines of evidence indicate that the role of Tel1 in DNA replication at short telomeres is not to affect Rif1 itself. Because we have yet to identify a role of Mcms in the Tel1 pathway, it remains to be established whether Tel1 and Rif1 opposing activities converge on the same target(s) in affecting replication timing. The PP1-recruiting function of S. pombe Rif1 extends beyond the control of replication timing: we find that Rif1 alleles defective in the ability to interact with PP1 have elongated telomeres, implicating PP1 phosphatases in the control of telomerase action. Presented by: Bianchi, Alessandro 83 Poster 12 PLATINATION OF TELOMERES BY CIS-PLATINE IS NOT AT THE ORIGINE OF TRF2 DISPLACEMENT Lina Sakera, Samar Alia, Guillaume Kellermanna, Evelyne Ségal-Bendirdjiana, Joël Pouponb, Sophie Bombarda,* a b INSERM UMR-S 1007, Université Paris Descartes; Paris, France Laboratoire de Toxicologie-Biologie, Hôpital Lariboisière, Paris, France Telomeres are DNA-protein structures that cap chromosome ends protecting them from recognition by the DNA repair machinery. In mammalians, among the six telomeric proteins belonging to the shelterin, TRF1 and TRF2 are both proteins able to bind directly to the double stranded telomeric DNA and are involved in telomere length regulation and chromosome end protection. Since telomeric DNA consists of highly repetitive short sequences of adjacent guanine residues (TTAGGG)n, it is a potential target for platinum complexes and especially for the anti-tumor drug cis-platin. In vitro, we have shown that adducts of cis-platin on telomeric DNA prevent the fixation of TRF2 and in a lesser extent the one of TRF1 to telomeres (1). Our aim was to determine if it could be the case in cellulo. Telomeres from two cancerous cells lines treated by cis-platin were purified and their platinum content has been quantified using ICP-mass. The amount of remaining telomeric proteins bound to telomeres has been also evaluated. As expected, we found that treatment of cells by cis-platin led to the platination of their telomeres. However, the amount of platinum bound to telomeres cannot explain, alone, the specific displacement of TRF2 from telomeres, suggesting that another mechanism is at the origin of TRF2 displacement. 1. Ourliac-Garnier, I. et al (2010) J. Biol. Inorg. Chem. 15,. 641-654 Presented by: Bombard, Sophie 84 Poster 13 Mutations in Telomerase Reverse Transcriptase Promoter Contribute to Urothelial Cancer: Mechanism and Therapeutic Implications Sumit Boraha,*, Arthur Zauga, Linghe Xia, Natasha Powella, James Costellob, Dan Theodorescuc, Thomas Cecha a University of Colorado Boulder BioFrontiers Institute, Dept of Chemistry and Biochemistry and HHMI University of Colorado Denver Anschutz Medical Campus, Dept of Pharmacology c University of Colorado Denver Anschutz Medical Campus, Cancer Center b Although telomerase activity is upregulated in 85% - 90% of cancer cells, the molecular mechanisms by which this happens are incompletely understood. The recent finding of highly recurrent point mutations in the TERT promoter in several types of cancer suggests their importance for oncogenesis. For some cancers, such as urothelial cancer (UC), these mutations are more frequently observed than any other. Although the promoter mutations were hypothesized to increase transcription by further recruitment of E-twenty-six transcription factors, very mild increases were observed in luciferase reporter assays. We seek to more thoroughly understand the significance of these mutations for oncogenesis by specifically focusing on UC – the fifth most common cancer in the Western world. We have collected 23 UC cell lines, sequenced the TERT promoters of each, and measured the levels of TERT protein, TR RNA, telomere lengths and, using direct assays, endogenous telomerase activity. Although heterogeneous with respect to these parameters, the five cell lines with the greatest telomerase activity harbor the -124 promoter mutation. Furthermore, we have collected genome-wide data to identify exon mutations and to measure mRNA expression levels in each cell line, to identify genes whose altered expression and/or mutation exacerbate or substitute for TERT promoter mutations. Finally, the susceptibility of several of these cell lines to the telomerase inhibitor BIBR1532 may reveal whether the promoter mutations might provide a biomarker for anti-telomerase therapy. Our goal is to elucidate the molecular mechanisms by which these mutations contribute to oncogenesis, and potentially reveal unexplored aspects of telomerase biology. Presented by: Borah, Sumit 85 Poster 14 Regulation of human telomere transcription Joanna Boros, Anabelle Decottignies Genetic and Epigenetic Alterations of Genomes, de Duve Institute, Catholic University of Louvain, Brussels, Belgium Telomeres, despite their highly heterochromatic nature, are transcribed into TElomeric Repeatcontaining RNAs (TERRA). TERRA molecules are transcribed by RNA polymerase II, initiate from subtelomeric promoters and are controlled by cell cycle and telomere length. Ensuring proper levels of TERRA in cells is vital for telomere maintenance, as recent studies suggested an involvement of these non-coding RNAs in various aspects of telomere biology, including heterochromatin formation or Tloop regulation. In addition, a recent quantitative SILAC-based screen identified additional putative TERRA interacting proteins involved in a variety of cellular processes. All these data suggest that generation of TERRA RNA has to be subjected to tight transcriptional regulation. To obtain more insight into TERRA regulation, we have performed an in silico analysis of human subtelomeric promoter regions. Distribution patterns of general core promoter elements within the CpG islands containing promoters imply, that transcription initiates from multiple sites. Interestingly, in promoter regions lacking the CpG islands, we identified TATA-box like elements indicative of single-site transcription initiation. In addition, we also identified putative sites for specific transcription factor and verified its presence on human TERRA promoters by ChIP. Moreover, we demonstrate that TERRA can be a downstream effector of a signaling pathway, which can provide potential link between mitochondria maintenance and telomere integrity. Presented by: Boros, Joanna 86 Poster 15 Beneficial testosterone treatment in a mouse model of aplastic anaemia Christian Bӓr, Fabian Beier, Miguel Foronda, Maria Blasco Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, E-28029, Spain Aplastic anaemia (AA) is a rare but serious disorder which can develop at any stage of life and which is characterised by cytopenia in one or more of the three blood lineages. Most cases of AA are due to autoimmune processes against the hematopoietic stem cell compartment. One general features of AA is the presence of shortened telomeres. In this regard, recent studies demonstrated that a small but significant subset of patients carry mutations in the core telomerase components Tert or Terc. Haploinsufficiencies in components of the telomerase complex lead to accelerated telomere attrition and subsequently to stem cell exhaustion and bone marrow failure. Interestingly, testosterone has been used for over half a century in the treatment of AA. Despite of frequent induction of remission following hormone therapy, the underlying mechanism and the cause for treatment response remained obscure. However, recent studies showed that sex hormones increase telomerase activity in primary human haematopoietic cells in vitro. It is therefore conceivable that testosterone-mediated telomerase activation could attenuate or reverse AA disease progression associated with impaired telomere maintenance. To address this question, here we use a mouse model of aplastic anaemia recently developed in our laboratory in which we can shorten telomeres at will specifically in the bone marrow compartment. We find that testosterone therapy increases blood counts and extends life-span under conditions of continuous telomere erosion. Importantly, longitudinal follow-up studies revealed a positive correlation between telomere length and hormone treatment in peripheral blood. Presented by: Bӓr, Christian 87 Poster 16 The putative Leishmania spp. telomerase RNA (TER) undergoes trans-splicing and contains a conserved template sequence Elton José Vasconcelosa, Vinicius Santana Nunesb, Marcelo Santos da Silvac, Marcela Segattod, Peter Mylera, Maria Isabel Nogueira Canob,* a Seattle Biomedical Research Institute, Seattle, Washington, United States of America Genetics Dept., Bioscience Institute, Universidade Estadual Paulista Júlio de Mesquita Filho, UNESP, Botucatu, Sao Paulo, Brazil c Genetics Dept., Bioscience Institute, Universidade Estadual Paulista Júlio de Mesquita Filho, UNESP, Botucatu,Sao Paulo, Brazil d Genetica Dept., Bioscience Institute, Universidade Estadual Paulista Júlio de Mesquita Filho, UNESP, Botucatu, Sao Paulo, Brazil b Telomerase RNAs (TERs) are highly divergent among different species, varying both in size and sequence composition. Recent studies have unraveled a 993 nt transcript that is part of the trypanosomatid protozoa Trypanosoma brucei TERT holoenzyme (TbTER). Due to the existence of alternative polyA sites, TbTER might also achieve a length of about 1,500 nt. Here we present LeishTER, a candidate to the telomerase RNA component of Leishmania spp., another pathogenic trypanosomatid that causes leishmaniasis, a neglected tropical multi-symptoms disease. Merging a thorough computational screening together with RNA-Seq evidences and target-driven molecular biology assays, we have detected a ~2,150 nt-long transcript assigned as LeishTER, one of the longest TERs described so far. Similarly to TbTER, LeishTER is also trans-spliced, probably polyadenylated and bears a C/D box snoRNA domain. LeishTER was co-purified with the telomerase protein component (TERT) from TERT-IP nuclear extracts. Using RNA FISH coupled with immunofluorescence, it was possible to see that LeishTER co-localizes with TERT in a cell cycle-dependent manner. Our in silico prediction of the LeishTER secondary structure strongly suggests the existence of a bona fide singlestranded template sequence similar to the TbTER template and a C[U/C]GUCA motif-containing Helix II, which is also conserved in Tetrahymena thermophila TER and represents the template boundary element (TBE). This study is the first report on a candidate to Leishmania spp TER component, paving the way to further investigations on the biogenesis of parasite TERT RNP and, consequently, its role in parasite telomere biology and as a future target for chemotherapeutic intervention. Presented by: Cano, Maria Isabel Nogueira 88 Poster 17 The Effect of Polymerase-alpha on Telomerase Processivity Chris Caridi, Connie Nugent University of California, Riverside Telomeres, the ends of linear chromosomes, serve three functions; protecting DNA from nucleolytic degradation, blocking the DNA damage checkpoint and ensuring replication of chromosome ends. Telomerase, a reverse transcriptase that extends the 3’ ends of chromosomes, is important for maintaining sufficient telomere length ensuring genome stability and the replicative lifespan of cells. One question regarding telomere biology is how telomerase mediated extension is terminated. Telomere binding proteins promote the selection of ends for telomerase extension, but once telomerase engages an end, little is known about what limits the number of repeats that it synthesizes. We investigated the role of the polymerase alpha-primase complex (Pol-α) in regulation of telomerase access and repeat addition processivity in S. cerevisiae. To test the effect of Pol-α on telomerase activity an assay developed by Linger et al was used. Using a Two Template-Single Telomere Extension Assay (2T-STEX, Chang et al. 2007) we have determined that a temperature sensitive allele of POL1 (cdc17-2) results in increased telomerase processivity at both the permissive and non-permissive temperatures. In addition, telomerase is more promiscuous in extending ends, lacking a preference for extending shorter ends. Thus, Pol-α may play a role in stopping telomerase extension and allowing for “fill-in” synthesis of the complementary strand. This mis-regulation of telomerase may account for the elongated telomeres observed in cdc17-2 strains. Whether the impact of Pol-α on telomerase is mediated through its contacts with the Cdc13-Stn1-Ten1 proteins is being determined by testing alleles more specifically deficient for interaction among the complexes. Presented by: Caridi, Chris 89 Poster 18 EXPRESSION OF A SMALL INTERNAL FRAGMENT OF DYSKERIN (GSE24.2), DECREASES DNA DAMAGE, OXIDATIVE STRESS AND SENESCENCE IN ATAXIA TELANGIECTASIA CELLS Jaime Carrilloa,*, Laura Pintado Berninchesa, Cristina Manguán Garcíaa, Laura Iarriccioa, Guillermo Güenechea Amurriob, Juan A. Buerenb, Leandro Sastrea, Rosario Peronaa a Instituto de Investigaciones Biomédicas CSIC/UAM, IDIPaz and CIBER de enfermedades raras. Madrid, Spain. b Gene therapy in haematopoyetic system department. CIEMAT and CIBER de enfermedades raras. Madrid, Spain. The ATM gene is mutated in Ataxia telangiectasia (AT), although the mechanisms underlying AT disease are still not fully understood. ATM protein senses and regulates cellular redox status, using a complex network of downstream signaling pathways, and is also a key transducer of DNA damage signals to downstream effectors. ATM deficient cells show increased ROS accumulation, activation of the p38 protein kinase and increased basal levels of DNA damage. GSE24.2 a peptide corresponding to an internal domain of Dyskerin has proved to induce telomerase activity by stabilizing TERC and increasing expression of TERT (Machado-Pinilla, R. et al. (2008) Blood, 111:2606-14). GSE24.2 (Gestelmir) has been recently approved by EMA for the treatment of Dyskeratosis congenita. Due to these findings, GSE24.2 was expressed in AT human primary fibroblasts and lymphoblastoid cell lines, which harbor different mutations in the ATM gene, to study the effect in DNA damage, ROS production, expression of proinflammatory cytokines, survival to radiomimetic drugs, activation of p38 protein kinases and senescence induction. Infection of these cells with a lentiviral vector expressing GSE24.2 showed a reduction in basal levels of DNA damage, decreased levels of ROS, lower levels of p38 phosphorylation and pro-inflammatory cytokines, higher resistance to bleomycin induced DNA damage and lower senescence than cells infected with control lentivirus expressing GFP. Furthermore, treatment of AT fibroblasts with nanoparticles loaded with synthetic GSE24.2 or a shorter derived peptide GSE4 was able to rescue from DNA damage and senescence. Financial Support: (FIS-P11-00949 and INNPACTO IPT-2012-0674-090000 Cristina Manguan is contracted by CIBERER. Presented by: Carrillo, Jaime 90 Poster 19 Identification and validation of genome instability drivers in a panel of human cancers Maria Antonietta Ceronea,*, Nnenna Kanua, Mcgranahan Nicholasb, Pierre Martinezb, Charles Swantonc a UCL Cancer Institute, Paul O'Gorman Building, Huntley St., London WC1E 6BT, UK Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK c Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK UCL Cancer Institute, Paul O'Gorman Building, Huntley St., London WC1E 6BT, UK b Genome instability is a hallmark of all cancers. It is characterized by alterations and/or loss of part or whole chromosomes and often results in aneuploidy. High genome instability has been associated with a more malignant phenotype, resistance to treatment and worse prognosis for the patients. The molecular mechanisms underlying genomic instability are still not completely understood. Several large-scale genomic studies have been performed to identify gene mutations that cause genome instability. The aim of this study was to evaluate the functional impact of a subset of gene mutations on inducing and/or enhancing genome instability. Using the data obtained by exome sequencing of hundreds of human tumors recently published in the literature, we have selected 65 unique genes significantly mutated in four cancer types: breast, colon, lung and kidney. These genes are all potential tumor suppressor genes, whose alterations result in loss of function of the proteins of interest. They include 30 genes mutated in breast cancer, 25 in colon, 12 in kidney and 24 in lung. To analyze their function, we will use three cell lines per tumor type and perform a siRNA screen using as read-out a panel of assays to monitor several aspects of genome stability, including induction of DNA damage, presence of segregation errors, alteration of the cell cycle profile, cytokinesis defects, telomere maintenance, viability and cell death. Preliminary results will be presented at the meeting. Presented by: Cerone, Maria Antonietta 91 Poster 20 The Ctc1/Stn1/Ten1 complex localizes in APB and regulates C-circle formation in ALT cells Chenhui Huang, Xueyu Dai, Weihang Chai Washington State University Telomerase-negative cancer cells counteract telomere loss by activating the telomerase-independent ALT pathway. However, the molecular details of the ALT pathway are largely elusive. The Ctc1/Stn1/Ten1 complex (CST) was previously reported to play an important role in telomere maintenance through promoting efficient telomere replication, mediating C-strand fill-in, and regulating telomerase activity in telomerase-expressing non-ALT cancer cells. In this study, we report that human CST forms a functional complex localizing in the ALT-associated PML bodies (APBs) in ALT cells. Suppression of CST inhibits ALT cell proliferation and significantly diminishes C-circle abundance without affecting the overall telomere length. The impact of CST on C-circle homeostasis is unlikely due to replication defect induced by CST defection. Using the separation-of-function mutations of Ctc1, we also find that CST binding to telomeres is important for maintaining the CC levels in ALT cells, while the CST/polalpha interaction is dispensable for regulating CC production. Our findings therefore reveal a role of CST in regulating C-circle formation that is independent of its function in telomere replication, suggesting that the function of CST at telomeres is multifaceted. Presented by: Chai, Weihang 92 Poster 21 Telomere and mitochondrial dysfunction in Duchenne Muscular Dystrophy Alex AC Changa,*, Foteini Mourkiotia, Sang-Ging Ongb, Joseph Wub, Helen M Blaua a Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, and Stanford Cardiovascular Institute, Stanford, CA, USA b Division of Cardiology, Department of Medicine and Stanford Cardiovascular Institute, Stanford, CA, USA Duchenne muscular dystrophy (DMD) is a lethal X-linked recessive disease that is result of mutations in the dystrophin gene and is the most common myopathic disease in humans with a prevalence of one in every 3500 males. Dystrophin is crucial for the formation of a dystrophin-glycoprotein complex (DGC), which connects the cytoskeleton of a muscle fiber to the surrounding extracellular matrix in both skeletal and cardiac muscles. In the heart, loss of dystrophin leads to increased fibrosis and death in the third decade of life due to dilated cardiomyopathy. A conundrum in studying and developing therapies for DMD has been the lack of a mouse model that fully recapitulates the clinical phenotype, as mice that lack dystrophin (mdx model), unlike patients, exhibit only mild skeletal muscle defects, essentially no cardiac defects and have a relatively normal lifespan. Our lab reasoned that the difference in the manifestation of the disease in mice and humans could be telomere length, as mice have substantially longer telomeres than humans. We created a novel mouse model with shortened telomere lengths (similar to humans) that fully recapitulates the skeletal muscle (Cell. 2010;143:10591071; the mdx/mTRKO model) and cardiac muscle phenotype of DMD (Nat Cell Biol. 2013; 15:895-904; dilated cardiomyopathy). Interestingly, we observed a 45% reduction in cardiomyocyte telomere length in our mdx/mTRKO animals as well as patient samples. Here we present new evidence of mitochondrial dysfunction and telomere dysfunction. Presented by: Chang, Alex AC 93 Poster 22 CTC1 Mutations in Telomere Syndromes Impair Telomere Replication Liuh-yow Chen, Jana Majerská, Joachim Lingner Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Switzerland Mutations in CTC1 lead to the telomere syndromes Coats Plus and Dyskeratosis Congenita, but the involved molecular mechanisms remain unknown. CTC1 forms with STN1 and TEN1 a trimeric complex termed CST, which promotes telomere DNA synthesis and inhibits telomerase mediated telomere elongation. Here we identify CTC1 disease mutations that disrupt CST complex formation, the physical interaction with DNA polymerase α-primase, telomeric ssDNA binding in vitro or/and nuclear localization and telomere association in vivo. While having diverse molecular defects, expression of CTC1 mutant proteins commonly leads to the accumulation of internal single-stranded gaps of telomeric DNA, suggesting telomere DNA replication defects as a primary cause of the disease. Furthermore, cytological analysis indicates that telomerase inhibition enhances telomere structure loss upon expression of CTC1 mutant proteins, suggesting that telomerase may be able compensate the telomere replication defect. Hence the telomere defect initiated by CTC1 mutations is distinct from the telomerase deficiency seen in classical forms of telomere syndromes, which cause short telomeres due to reduced maintenance of distal telomeric ends by telomerase. In addition, our analysis provides direct molecular evidence that CST collaborates with DNA polymerase α-primase to promote faithful telomere DNA replication. *Current Address Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan Presented by: Chen, Liuh-yow 94 Poster 23 Anti-metastatic factor inhibits telomerase activity in aggressive cancer cells - emerging connection between telomerase activity and metastasis suppression Vivek Srivastavaa, Anirban Kara, Maneesh Kumara, Dhurjhoti Sahaa, Ankita Singha, Vinod Kumar Yadavb, Shantanu Chowdhuryc,* a Proteomics and Structural Biology Unit, CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110 020, India b G.N.R. Knowledge Centre for Genome Informatics, CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi 110 020, India c G.N.R. Knowledge Centre for Genome Informatics, Proteomics and Structural Biology Unit, CSIRInstitute of Genomics and Integrative Biology, Mathura Road, New Delhi 110 020, India Anti-metastatic function of NME2 has been demonstrated in several human cancers, however mechanisms of metastasis control by NME2 are poorly understood. In genomewide location (ChIP-seq) experiments we found presence of NME2 on telomeres in multiple human cancer cells. This led to the finding that NME2 associates with telomerase and inhibits telomerase activity in vitro and in cellulo (Kar et al., Nucleic Acids Research). Recent results, using a loss of function mutant, confirm interaction of NME2 with hTERT is through direct protein-protein association in a particular domain of telomerase. Mechanistic evidence supports inhibition of telomerase reverse transcription function in presence of NME2. Furthermore, we found that telomerase-inhibition is dependent on NME2-telomerase association. The impact of NME2-mediated inhibition of telomerase activity on metastatic potential of aggressive cancer cells has also been studied. These results will be presented. Together these novel findings present an unique opportunity to explore telomerase function in metastasis. Presented by: Chowdhury, Shantanu 95 Poster 24 Characterization of the binding of the Drosophila telomeric protein Verrocchio to single-stranded DNA Alessandro Cicconia,*, Emanuela Michelia, Domenico Raimondob, Giovanni Cencia, Maurizio Gattia, Stefano Cacchionea, Grazia D. Raffaa a b Dipartimento di Biologia e Biotecnologie Charles Darwin, Sapienza University, Roma, Italy Dipartimento di Fisica, Sapienza University, Roma, Italy Drosophila telomeres are composed of three specialized non-LTR retrotransposons. These elements are recognized by a specific telomeric complex, called terminin, whose subunits are not conserved in other eukaryotic organisms. The terminin proteins HOAP and HipHop bind double stranded DNA, and are enriched over a large telomeric domain, while Modigliani (Moi) and Verrocchio (Ver) localize only at the very end of the chromosome. Ver has an OB-fold domain, a structural motif present also in other proteins that are capable of binding single-stranded DNA, such as RPA and POT1. Moi is not able to bind single-stranded DNA on its own, but it’s responsible for Ver recruitment at telomeres, through interaction with HOAP. Moi and Ver closely interact with each other and are mutually dependent for their stability, suggesting that their function could be analogous to TPP1-POT1 function. Using EMSA assay, we showed that Ver binds several single-stranded probes, in a sequence-independent manner and with single-stranded DNA specificity. We have further analyzed Ver binding to a telomere model system using AFM imaging. We found that Ver binds DNA in dimeric and tetrameric form, in agreement with the fact that OB-fold containing proteins bind DNA with two or more OB-folds. Our results reinforce the model of telomere protection in Drosophila based on the formation of the terminin complex, that is functionally analogous to the mammalian shelterin complex, but lacks sequence specific binding activity. Presented by: Cicconi, Alessandro 96 Poster 25 Separase is required for telomere protection Francesca Cipressaa,*, Patrizia Morcianoa, Giuseppe Bossoa, Linda Manninib, Grazia Daniela Raffaa, Antonio Musiob, Giovanni Cencia a Department of Biology and Biotechnology "Charles Darwin" Section of Genetics, SAPIENZA University of Rome 00185 Rome, Italy b Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Pisa In most organisms, telomeres are extended by telomerase and contain GC-rich repeats. Drosophila telomeres are elongated by transposition events rather than telomerase activity and are assembled in a sequence independent fashion. Drosophila telomeres are capped by terminin, a complex that includes the non-conserved and fast evolving proteins HOAP, HipHop, Moi and Ver, which localize and function only at telomeres, protecting them from fusion events. The other proteins that protect Drosophila telomeres from fusion (Eff, HP1, ATM, Rad50, Mre11,Nbs, and Woc) are evolutionarily conserved, do not localize only at telomeres and play several functions in addition to telomere protection. Here, we show that mutations in the Drosophila Separase coding gene (Sse) lead to endoreduplication and telomeric fusion (TFs). We also demonstrate that Sse physically binds both the terminin proteins and HP1 and that it is enriched at telomeres. While localization of terminin and Sse are not interdependent, loss of Sse strongly reduces HP1 levels. Overexpression of HP1 in Sse mutants suppresses telomere fusion but does not rescue the endoreduplication phenotype, suggesting that the TFs seen in Sse mutants are due to a reduction of the HP1 level. A catalytically inactive SSE failed to restore the HP1 level and to reduce TF frequency indicating that the SSE endopeptidase activity is ultimately required for telomere protection. Finally, siRNA-induced depletion of ESLP1, the SSE human ortholog, caused dysfunctional telomeres and HP1 reduction in human primary fibroblasts, highlighting and evolutionarily conserved role of separase in telomere protection. Presented by: Cipressa, Francesca 97 Poster 26 TELOMERE DAMAGE AND CHROMOSOME INSTABILITY INDUCED BY ACUTE OXIDATIVE STRESS Elisa Coluzzia,*, Monica Colamartinoa, Renata Cozzia, Stefano Leonea, Nathan O’Callaghanb, Antonella Sguraa a b Dept. of Science, University of “ Roma Tre”, Viale Guglielmo Marconi, 446, 00146 Rome, Italy. CSIRO Food and nutritional Sciences, Nutritional Genomics Laboratory, Adelaide, Australia. One of the main role of telomere is to maintain chromosome and genome stability. In this work, our interest was focused on the role of telomere in chromosome instability induced by oxidative stress. With the aim to evaluate if the chromosome instability induced by oxidative stress is related specifically to telomeric damage, we used human primary fibroblasts (MRC-5) treated with two doses of H2O2 (100 and 200 μM) for 1 hour. The evaluation of the cell cycle by citofluorimetric analysis showed no cell cycle effects after treatments. The analysis of chromosome instability (CIN) demonstrated a significant telomere shortening 48 hours after treatment and an increase of CIN (micronuclei, nuclear buds and nucleoplasmic bridges) at the same time. At the subsequent times (72 and 96 hrs) we can observe a restoring of telomere length and a reduction of CIN, leaving us to suppose a correlation between telomere shortening/dysfunction and chromosome instability. To understand the mechanism leading to chromosome instability, we evaluate the persistence of oxidative stress-induced damage up to 24 hours after treatment analyzing genomic and specifically telomeric FPG-sensitive sites by modified comet assay and qPCR, respectively. The results obtained demonstrated that after 24 hrs genomic damage was completely repaired while telomeric oxidative damage persist up to 24 hrs leading us to hypothesize that telomere dysfunction could be responsible of chromosome instability observed 48 hrs from treatment. Furthermore, the analysis of nucleoplasmic bridges hybridized with telomeric probe will give us additional information on the role of telomere in the observed aberration. Presented by: Coluzzi, Elisa 98 Poster 27 ALT-specific recruitment of NuRD-ZNF827 to telomeres promotes homologous recombination and protects against senescence and apoptosis Dimitri Conomosa,*, Roger Reddelb, Hilda Picketta a Telomere Length Regulation Group, Children’s Medical Research Institute, Westmead, NSW, Australia. b Cancer Research Unit, Children’s Medical Research Institute, Westmead, NSW, Australia. It has recently been shown that nuclear receptors are recruited to the telomeres of cells that utilize ALT (1,2). We proposed that this contributes to remodeling of the telomeric architecture of ALT cells to promote ALT activity (3), but the mechanism remained to be determined. Here we demonstrate that the nucleosome remodeling and histone deacetylation (NuRD) complex is recruited to nuclear receptors by an N-terminal RRK motif within ZNF827, a zinc finger protein of unknown function. We show that the NuRD-ZNF827 complex compacts ALT telomeric chromatin and is required for ALT, and alteration of its expression level affects numerous ALT phenotypic characteristics and recruitment of homologous recombination (HR) proteins. NuRD-ZNF827 binding to telomeres promotes interactions of telomeres with telomeric DNA in ALT-associated PML bodies (APBs). ZNF827 depletion increases the telomeric DNA damage response and results in senescence and/or apoptosis, in an ALT-specific manner. We propose that NuRD-ZNF827 provides an environment that fosters HR and chromatin compaction and thereby inhibits recognition of DNA damage at the telomeres of ALT cells. (1) Dejardin, J. and Kingston, R. E. Purification of proteins associated with specific genomic loci. Cell 136: 175-186, 2009. (2) Conomos, D., et al. Variant repeats are interspersed throughout the telomeres and recruit nuclear receptors in ALT cells. J. Cell Biol. 199: 893-906, 2012. (3) Conomos, D., et al. Alternative lengthening of telomeres: remodeling the telomere architecture. Front. Oncol. 3: 1-7, 2013. Presented by: Conomos, Dimitri 99 Poster 28 Shading the shelterin TRFH TRF2 domain through cell-permeable chemical tools: rational design, synthesis and biological effects at telomeres Salvatore Di Maroa, Pasquale Zizzab, Erica Salvatib, Bruno Paganoa, Clemente Capassoc, Luciana Marinellia, Ettore Novellinoa, Annamaria Birocciob, Sandro Cosconatid,* a Department of Pharmacy, University of Naples “Federico II”, Naples, Italy Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy c Istituto di Biochimica delle Proteine—CNR, Naples, Italy d DiSTABiF, Second University of Naples, Caserta, Italy b In mammalian cells, the telomeric repeat binding factor (TRF) homology (TRFH) domain–containing telomeric protein TRF2 associates with different downstream factors necessary for telomere maintenance and cell-cycle progression. While cogent experimental evidences demonstrate that the TRF2 recruiting activity is a critical arbiter of cell fate decisions, no chemical tools have ever been described to modulate this function. Based upon the crystal structure of TRF2 in complex with the Apollo C-terminal peptide, we have designed and synthesized a series of peptides through mutations and cyclization. Aided by computer simulations we were able to identify a potent TRF2 TRFH binder with a Kd value of 125 nM and two times more affine that the wild-type Apollo peptide (250 nM). By using human transformed BJ fibroblasts (BJ-EHLT), we demonstrate that, differently to the linear Apollo peptide, our chemotype is able to induce marked DNA damage response and deconvolution microscopy analysis showed that the damaged foci colocalized with TRF1, a good marker for interphase telomeres forming the so-called TIF. Future experimental studies will involve the employment of this pharmacological tool to study the role of TRF2 TRFH protein hub in shaping and safeguarding telomeres. Presented by: Cosconati, Sandro 100 Poster 29 RPA and Pfh1(Pif1) helicase prevent secondary structures formation during telomere replication Julien AUDRYa, Pierre LUCIANOa, Toru NAKAMURAb, Vincent GELIa, Stephane COULONa,* a b CRCM-Marseille University of Illinois at Chicago In fission yeast, the telomeric complex, also named shelterin complex, ensures the protection of chromosomes ends and contributes to the recruitment of telomerase. It is established that the nontelomeric factor ATRRad3 kinase phosphorylates Ccq1 to promote recruitment and activation of telomerase when replication is completed. Lately, we have shown that the Replication Proteine A (RPA) participates to telomere maintenance in a telomerase-dependent manner but independently of ATRRad3 pathway. RPA recruitment at telomeres is cell cycle regulated and peaks in late S-phase. A point mutation (D223Y) provokes telomeres shortening. This mutation does not impede RPA recruitment at telomeres, but the presence of RPAD223Y at telomeres is prolonged. ChIP of replicative polymerases have shown that Polε is recruited at telomeres indifferently in wild-type and D223Y strains while Polα remains longer at telomeres after completion of S-phase in RPA1D223Y mutant. These observations suggest that D223Y allele causes defects in replication of the G-rich lagging strand telomere. In agreement, 2D-gel analysis showed that replication of telomeric regions is severely altered in D223Y strain. Our results suggest that D223Y mutation impedes the proper replication of the G-rich telomeric DNA and provokes accumulation of secondary DNA structures like G-quadruplex. DNA helicases are known to unwind these structures. Strikingly, we have shown that over-expression of either Pfh1(Pif1) or S. cerevisae Pif1 are able to rescue the short telomere phenotype of RPAD223Y. We propose that RPA and Pfh1Pif1 act at telomeres to prevent accumulation of DNA secondary structures for efficient telomere elongation. Presented by: COULON, Stephane 101 Poster 30 Cross talk between EBV and telomerase: the role of TERT in the switch of latent/lytic cycle of the virus Silvia Giuncoa, Andrea Celeghinb, Stefano Indraccoloa, Riccardo Dolcettic, Anita De Rossid,* a Istituto Oncologico Veneto-IRCCS, Padova, Italy Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova; Padova; Italy c Cancer Bio-Immunotherapy Unit, CRO-IRCCS, National Cancer Institute, Aviano, Italy d Department of Surgery, Oncology and Gastroenterology, Section of Oncology and Immunology, University of Padova; Padova; Italy; Istituto Oncologico Veneto-IRCCS, Padova, Italy b Epstein-Barr virus (EBV)-associated malignancies in vivo, as well as lymphoblastoid cell lines (LCLs), obtained in vitro by EBV infection of B cells, selectively express latent viral proteins and maintain ability to grow indefinitely through inappropriate activation of TERT, the catalytic component of telomerase. Our studies demonstrated that high level of TERT expression in LCLs prevented the EBV lytic cycle, which is instead triggered by TERT silencing (Giunco et al, Clin Cancer Res 2013;19:2036). As the lytic infection promotes the death of EBV-positive tumor cells, understanding the mechanism(s) by which TERT affects the latent/lytic status of EBV may be important for setting new therapeutic strategies. BATF, a transcription factor activated by NOTCH2, the major NOTCH family member in B cells, negatively affects the expression of BZLF1, the master protein of the viral lytic cycle; therefore we analyzed the interplay between TERT, BATF, and NOTCH2 in LCLs. We found that LCLs with high TERT levels showed high NOTCH2 and BATF expression, while LCLs with low TERT levels did not. Infection of TERT-negative LCLs with a retroviral vector expressing TERT(pBABE-hTERT) increased NOTCH2 and BATF at both mRNA and protein levels. By contrast, infection of TERT-negative LCLs with retroviral vectors expressing NOTCH2(NOTCH2-IC) did not induce TERT transcription. By using luciferase reporter assay we demonstrated that ectopic expression of TERT (pEGFP-hTERT) activated NOTCH2 promoter (pGL3-TATA-N2PR-2327/-99) in a dose-dependent manner; while NOTCH2 did not activate the TERT promoter. Taken together, these results suggest that TERT modulates the lytic/latent status of EBV through the NOTCH2 pathway. Presented by: De Rossi, Anita 102 Poster 31 Dissecting the link between the long non-coding RNA TERRA and the alternative lengthening of telomeres mechanism Katharina Deeg, Karsten Rippe Research Group Genome Organization and Function, German Cancer Research Center (DKFZ) & BioQuant, Heidelberg, Germany To counteract the continuous shortening of the telomeres, cancer cells activate a telomere maintenance mechanism and thereby acquire unlimited proliferative potential. A fraction of human cancers utilizes the alternative lengthening of telomeres (ALT) mechanism that involves recombination and repair processes. Hallmarks of the ALT pathway are colocalizations of telomeres and promyelocytic leukemia protein (PML), termed ALT-associated PML-bodies (APBs) and the presence of extrachromosomal telomere repeats. In addition, ALT-positive cells have elevated levels of the telomeric repeat-containing RNA (TERRA), a long non-coding RNA that is transcribed from the telomeres. Notably, TERRA has been shown to stimulate telomere recombination in yeast cells lacking telomerase. However, it remains elusive whether TERRA is essential for the ALT pathway in cancer cells. We performed a comprehensive analysis of telomere maintenance features in glioblastoma cell lines with different genetic background and reveal a strong correlation between TERRA levels and the presence of ALT in these tumors. Yet, APB depletion by stable knockdown of PML did not alter TERRA levels. To investigate whether TERRA is a functional component of the ALT pathway, we depleted the telomeric part of TERRA by antisense oligonucleotide transfection and analyzed for changes in the number of APBs and C-circle levels. TERRA knockdown in the ALT-positive U2OS cell line did not significantly affect any of the characteristic ALT features tested. This suggests that TERRA is not needed for the maintenance of ALT, but may still be essential for ALT initiation. Presented by: Deeg, Katharina 103 Poster 32 Influence of telomere dynamics on disease progression and response to therapeutics in bone marrow failure syndromes Erin Degelman, Nicholas Ting, Tara Beattie The University of Calgary Aplastic Anemia (AA) is a bone marrow failure disease where insufficient levels of hematopoietic cells are produced. In approximately 10-15% of patients, the AA evolves into myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML). Studies have demonstrated an association between shortened chromosome ends, advanced AA and increased risk of developing blood cancers. Heterozygous mutations in the gene encoding the telomerase protein component hTERT, are seen in approximately 10% of patients, resulting in shortened telomeres. However, how telomerase mutations and shortened telomeres impact disease progression and response to treatment is not well understood. To understand the biochemical properties and cellular consequences of mutant hTERT expression we have generated expression constructs corresponding to four hTERT mutations. These mutations have been identified in patients with familial AA and AML and family history of evolution of disease and are all located in functionally distinct regions of the protein. We have demonstrated that the hTERT mutants retain telomerase activity in vitro and we are investigating the consequences of their expression in leukemic cells lines. Preliminary data suggests that expression of certain mutant hTERT proteins in THP-1 cells, results in a delay of the G1/S transition, which may have profound implications during hematopoiesis. In addition, THP-1 cells expressing mutant hTERT proteins are more resistant to chemotherapeutic agents suggesting that treatment protocols might have differential efficacies depending whether cells express wild-type or mutant hTERT. By defining the role of telomeres in hematological disorders, it may be possible to alter treatment strategies based on predicted outcomes from our investigations. Presented by: Degelman, Erin 104 Poster 33 Pervasive hallmarks of Alternative Lengthening of Telomeres (ALT) in diploid Sorex granarius (Soricidae, Eulipotyphla) fibroblast cells Irena Draskovica,*, Natalia Zhdanovab, Julia Mininab, Tatiana Karamyshevab, Clara Novo-Lopesa, Win-Yan Liua, Maria Zverevac, Andrey Skvortsovc, Nikolay Rubtsovb, Arturo Londo o-Vallejoa a Telomeres Cancer Laboratory, “E uipe Labellis e Ligue Contre le Cancer”, Institut Curie, 26 rue d'Ulm, 75248 Paris, France; UPMC - University Paris 06, 75005 Paris, France. b Institute of Cytology and Genetics of Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia. c Moscow State University, Faculty of Chemistry, Moscow, Russia. The telomere structure in the Iberian shrew Sorex granarius is characterized by unique, striking features. While the closely related S. araneus bears telomeres very similar in structure to those found in humans, the short arms of S. granarius acrocentric chromosomes carry extremely long telomeres (up to 300 kb) with interspersed rDNA repeat blocks. In this work, we investigate the telomere physiology of S. granarius fibroblast cells and found that telomere repeats are transcribed on both strands and that there is no telomere-dependent senescence mechanism, with telomere lengths being maintained in long-term cultures. Although telomerase activity is detectable all along the cell culture, we also discovered that hallmarks of Alternative Lengthening of Telomeres (ALT) mechanism are omnipresent, including telomere length heterogeneity, presence of telomere sister chromatid exchanges (T-SCE), formation of ALT-associated PML bodies (APBs), production of telomere circles (T-circles) and high frequency of telomeres carrying marks of DNA damage response. Our results indicate that both telomerase-dependent and ALT-like mechanisms participate in telomere length homeostasis of normal S. granarius fibroblasts. Presented by: Draskovic, Irena 105 Poster 34 Telomere Maintenance in Pediatric Medulloblastoma Matthew Soboa, Kathleen Dorrisa, Ashley Margolb, Charles Stevensona, Shahab Asgharzadehb, Stewart Goldmanc, Lili Milesa, Arzu Onar-Thomasd, Maryam Fouladia, Rachid Drissia,* a Cincinnati Children's Hospital Medical Center The Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California c Ann Robert H. Lurie Children’s Hospital of Chicago d St. Jude Children’s Research Hospital b The prognostic significance of telomerase and telomere maintenance mechanisms in pediatric embryonal tumors has not been well defined. We, therefore, conducted a multi-institutional retrospective study of telomerase expression and telomere maintenance in 79 newly-diagnosed pediatric medulloblastomas and 15 primitive neuroectodermal tumors (PNET). Telomerase activity, hTERT promoter methylation, expression levels of hTERT, NPR3, and c-MYC mRNA and TERC RNA, and presence of ALT were evaluated. Cox proportional hazard regression analyses evaluated the association of clinical factors, NPR3 and c-MYC expression levels and telomere maintenance variables with progression-free survival (PFS) or overall survival (OS). Median age at diagnosis was 6.5 and 6.0 years for medulloblastoma and PNET cohorts, respectively. Seventy-three percent of medulloblastoma patients and 67% of PNET patients were M0 at diagnosis. hTERT expression levels were increased compared to non-neoplastic brain controls in 91.0% of medulloblastoma and 69.2% PNET samples. Ninety-three percent of medulloblastoma and 50% PNET samples assessed demonstrated telomerase activity. Median telomerase activity was 3-fold higher in anaplastic versus non-anaplastic medulloblastoma cohorts. All medulloblastoma samples that used ALT also exhibited evidence of telomerase use. In multivariable analysis of all medulloblastoma subjects, increased hTERT expression level was associated with worse PFS and OS in models that also included risk status or age. Similarly, in patients ≥ 36 months at diagnosis, increased hTERT expression level was associated with worse PFS and OS when simultaneously modeling for risk status. The relation between hTERT promoter methylation, hTERT expression and outcome will be discussed. M.S. and K.D. contributed equally Presented by: Drissi, Rachid 106 Poster 35 Molecular Dynamics of Telomeric Heterochromatin Formation Yi-Min Duan, Yang Zhang, Ting Gong, Jin-Qiu Zhou Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China In budding yeast Saccharomyces cerevisiae, heterochromatin structure is found at three well defined chromosome regions, i.e. HM Loci, rDNA region and telomere region. The former two regions exhibit relatively stable DNA sequence, which do not shorten or lengthen through cycles of cell division. Telomeres are rather more dynamic: elongated through telomerase pathway and/or attacked/trimmed by nucleases. To understand how telomeric heterochromatin is assembled, we adopted a de novo telomere addition system, and analyzed whether telomere lengthening and heterochromatin formation correlate with each other. Our results indicated that telomere lengthening takes more cycles of cell division to reach a stable length, while Sir protein spreading proceeds much faster. We also monitored the dynamics of histone modification and nucleosome positioning, and found that both levels of H4K16 acetylation and H3K79 methylation declined, and telomere proximal nucleosomes become more stable during the prosess of heterochromatin assembling. Presented by: Duan, Yi-Min 107 Poster 36 Role of DNA end-binding in Ku heterodimer function Charlene Emersona,*, Christopher Lopezb, Albert Ribes-Zamorab, Alison Bertuchc a Molecular and Human Genetics, Baylor College of Medicine Pediatrics, Baylor College of Medicine c Pediatrics, Molecular and Human Genetics, Baylor College of Medicine b The evolutionarily conserved Ku heterodimer is a DNA end-binding (DEB) complex involved in both telomere maintenance and non-homologous end-joining (NHEJ). In Saccharomyces cerevisiae, the Yku70/Yku80 subunits bind DNA ends via a channel structure. We previously generated DEB defective Ku mutants by mutating residues in the DNA-binding channel, revealing that DEB is re uired for Ku’s telomeric functions. Paradoxically, we also found that these mutants demonstrated increased colony survival in an assay that probes for imprecise NHEJ (Lopez et al. 2011, PLoS Genet). A subsequent study also generated DEB defective mutants of Ku by deleting residues to constrict the DNA binding channel (Pfingsten et al. 2013, Cell). These mutants similarly demonstrated a re uirement for Ku’s DEB activity at telomeres. However, these mutants were defective for imprecise NHEJ. To reconcile the differences between the two studies, we generated additional DEB defective Ku heterodimers and found that they too had increased imprecise NHEJ and defective telomere function. We also identified a novel region in Yku70 that is required solely for NHEJ and is predicted to associate with residues deleted in the DEB mutants reported by Pfingsten et al. We propose that these results reconcile the imprecise NHEJ differences observed and support our hypothesis that Ku heterodimers solely defective for DEB can modulate NHEJ off DNA ends. Currently, we are assaying whether the DEB mutants bind to double strand breaks (DSBs) in vivo. We expect that DEB mutants will be unable to bind DSBs, suggesting a previously unknown role for Ku in imprecise NHEJ. Presented by: Emerson, Charlene 108 Poster 37 Ssu72 phosphatase regulates telomere length in S. pombe Jose Miguel Escandell, Clara Reis, Maria Gallo, Miguel Godinho-Ferreira Instituto Gulbenkian de Ciência. Oeiras, Portugal Telomeres protect chromosome-ends from being recognized as deleterious double strand breaks. Paradoxically, the DNA damage recognition machinery is, itself, required for normal telomere homeostasis. In fission yeast, Rad3 and Tel1 are required for telomerase recruitment through the phosphorylation of Ccq1 on T93. In contrast, shelterin components are negative regulators of telomere elongation by counteracting Rad3/Tel1 dependent Ccq1 phosphorylation. The precise mechanism whereby telomere length regulation is achieved is not fully understood. We have identified a new member of telomere length regulation in S. pombe. We screened the fission yeast whole genome deletion library looking for new telomere regulators. We found that cells devoid of Ssu72 exhibit longer telomeres. Ssu72 is a highly conserved phosphatase previously identified in humans as an RNA polII regulator and a cohesin-binding factor involved in sister-chromatid cohesion. Telomere elongation in ssu72Δ cells is both trt1+ and rad3+ -dependent, consistent with a role as a negative regulator of telomerase. Telomere elongation in ssu72Δ is similar to the one observed in rif1Δ mutants. However, ssu72Δ rif1Δ double mutants have longer telomeres than any of the single mutants, suggesting that these genes are involved in independent pathways. Surprisingly, deletion of ssu72+ in taz1Δ, rap1Δ or poz1Δ leads to telomeres shortening upon progressive cell divisions. Thus, in combination with these shelterin components, Ssu72 appears to work as a positive regulator of telomerase. To uncover the role of Ssu72 in telomere homeostasis, we are currently investigating Ssu72 control of G-overhang formation and Ccq1 phosphorylation. Presented by: Escandell, Jose Miguel 109 Poster 38 Roles of DNA repair factors in plant telomere maintenance Lucie Najdekrovaa, Miloslava Fojtovaa, Dagmar Zachovaa, Karel J. Angelisb, Jiri Fajkusa,* a Mendel Centre for Plant Genomics and Proteomics, Central European Institute of Technology (CEITEC) and Faculty of Science, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic b Institute of Experimental Botany, Academy of Sciences of the Czech Republic, v.v.i., Na Karlovce 1, 160 00 Praha 6, Czech Republic Plant telomeres have been investigated during past 20 years mainly in the most common model plant, Arabidopsis thaliana. Although our previous studies focused on evolutionary structural diversity of telomeres in a broad phylogenetic radiation of monocotyledonous and dicotyledonous plants showed interesting changes in telomere players, including telomeric DNA sequences and respective telomerases for their replenishment, functional studies have almost exclusively been limited to Arabidopsis. Here we set to perform comparative investigation of telomere maintenance in the moss model, Physcomitrella patens. Unlike its 450 million years younger relatives – land plants, Physcomitrella is one of a few known multicellular organisms, with highly efficient system of homologous recombination. Although its basic telomere maintenance machinery is apparently similar to that of most land plants, the crosstalk between DNA repair factors and telomeres may result in telomere phenotypes different from those observed in A. thaliana, thus challenging the generalized views on the role of repair factors on telomere maintenance. We will present our current results on telomere dynamics obtained in P. patens strains deficient in DNA repair factors. Funding: Czech Science Foundation (13-06595S), CEITEC (CZ.1.05/1.1.00/02.0068) and the European Social Fund (CZ.1.07/2.3.00/30.0019). Presented by: Fajkus, Jiri 110 Poster 39 HSF1 regulates TERRA transcription upon heat shock Sivan Koskas, Claire Vourc'h, Virginie Faure Albert Bonniot Institute- University of Grenoble- INSERM U823 38042 Grenoble cedex 9 FRANCE In response to metabolic or environmental stress, the key transcription factor named Heat Shock Factor 1 (HSF1) is activated and coordinates a cellular protective response. In heat-shocked human cells, HSF1 induces the transcription of heterochromatin pericentric regions resulting in the accumulation of non coding RNA satIII. A global transcriptional analysis of repetitive sequences revealed that telomeric UUAGGG repeat-containing RNA (TERRA) expression is up-regulated upon heat shock. Recently, it has been reported that heat shock induces also the dissociation of TRF2 from telomeres. TRF2 is a well-known component of the shelterin complex protecting telomeres. These data prompted us to investigate the role of HSF1 on TERRA regulation and on telomere stability upon heat shock. Here, we clearly demonstrate for the first time that HSF1 upregulates TERRA expression upon heat shock. We show that HSF1 is recruited on subtelomeric regions under stress. Loss of HSF1 leads to a decrease in RNA polymerase II binding at subtelomeric regions and reduces the level of TERRA without impacting TERRA stability. We also bring evidence that TERRA upregulation is not accompanied by major changes in the epigenetic status of subtelomeric regions. HSF1 also has not impact on telomerase activity. In contrast, we show that HSF1 is involved in the telomeric localization of telomeric repeat factor 2 (TRF2) upon heat shock. We are currently examining the impact of HSF1 on telomere stability. Our work shades light on the role of HSF1 in the upregulation of TERRA transcription, which may participate in telomere stability in stressed cells. Presented by: Faure, Virginie 111 Poster 40 Telomerase is essential for zebrafish heart regeneration Dorota Bednareka, Juan Manuel González-Rosaa, Oscar Gutiérrez-Gutiérreza, Tania Aguadoa, Gabriela Guzmána, Alfonso Cortésb, Agustín Zapatab, Jesús Jiménez-Borregueroa, Nadia Mercadera, Ignacio Floresa a Spanish National Centre for Cardiovascular Research (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain b Electron Microscopy Center, Complutense University, 28040 Madrid, Spain Adult mammalian hearts regenerate very poorly after infarction. By contrast, zebrafish hearts efficiently regenerate after an insult. An essential step in the regeneration process that distincts zebrafish from adult mammalian hearts is the strong proliferation response of zebrafish cardiomyocytes to injury. One route to proliferation is thought to be rejuvenation, whereby aged cells regain characteristics of younger cells. Telomerase can be considered a rejuvenation enzyme since its reactivation in artificially aged tissues restores youthful characteristics and delays the aging process. Using a model of heart regeneration in zebrafish, we show that, after injury, telomerase is hyperactivated and telomeres transiently rejuvenate. Subsequently a subset of cardiomyocytes marked with long telomeres strongly proliferates. In the absence of telomerase activity, cardiomyocytes shorten their telomeres, accumulate DNA damage and are unable to efficiently expand, which eventually leads to a blockade of the regeneration process. These findings highlight the importance of rejuvenation in organ regeneration, establish long telomeres as a proliferation-competent marker for zebrafish cardiomyocytes and points to telomerase therapy as a potential approach to awaken hibernating cardiomyocytes found after myocardial infarction. Presented by: Flores, Ignacio 112 Poster 41 CBX1 buffers DNA damage responses and senescence induction in response to oncogene activation, γ-irradiation and telomere dysfunction T. K. Leuchta, S. Foerschb,*, S. Taoc, A. Lechela, K. L. Rudolphc a Cooperation group of the Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena and the University of Ulm, Ulm, Germany b 1. Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany 2. Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany c Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany DNA damage responses (DDRs) impact on aging. CBX1 is a member of the heterochromatin-protein-1family, which binds to H3K9me. Upon DNA damage, CBX1 is phosphorylated leading to the mobilization of CBX1 and the activation of DDRs. It is unknown whether deletion of CBX1 would alter DDRs and how this would affect cellular aging and transformation. Here, we studied consequences of CBX1 gene deletion on DDRs, cellular proliferation and senescence in human fibroblasts. The study shows that CBX1 buffers the induction of DDRs in cells with shortened telomeres and in response to oncogene activation. CBX1 deletion limits the proliferation of damaged cells by induction of cellular senescence. Importantly, CBX1 deletion has no effects on the proliferation of undamaged cells with long telomeres. Foci of p-CBX1 and decreases in CBX1 levels appear in senescent cells and in aging tissues with short telomeres indicating that CBX1 degradation represents an integral process of telomere dysfunction induced aging. Additionally, the expression of endogenous CBX1 is essential to prevent premature formation of γH2AX foci at critically short telomeres. Accordingly, the knockdown of CBX1 significantly shortens the proliferative lifespan of human fibroblasts. The negative effects of CBX1-deletion on proliferative lifespan of non-transformed cells, are contrasted by improved protection of human cells from oncogene-induced transformation. First data on conditional KO-mice confirm that CBX1 is essential to impair the activation of DDRs in vivo. Together, we provide first evidence for a CBX1dependent molecular buffer alleviating the activation of DDRs in the context of telomere shortening, irradiation and oncogenic stress. Presented by: Foersch, S. 113 Poster 42 Epigenetic regulation of telomeres and telomerases in different model plants Miloslava Fojtovaa,*, Pavla Polanskab, Eva Majerovab, Jana Jureckovab, Anna Ogrockab, Jiri Fajkusc a Central European Institute of Technology (CEITEC), Masaryk University Brno, Czech Republic Faculty of Science and CEITEC, Masaryk University Brno, Czech Republic c CEITEC, Masaryk University Brno, Czech Republic and Institute of Biophysics, ASCR, Brno, Czech Republic b Telomeres, as chromatin structures, are targets for epigenetic molecular mechanisms. In this case, the result of regulation processes in not a change of gene expression but rather a change of telomere length or structure potentially modulating telomere protective function. We analyzed maintenance of telomeres in hypomethylated plant models; Nicotiana tabacum (tobacco) culture cells cultivated in the presence of hypomethylating drugs, and Arabidopsis thaliana with loss of function of enzymes crucial for the maintenance of stable methylation pattern or wild-type plants germinated in the presence of hypomethylation drugs. Surprisingly, opposed results were obtained. While in tobacco telomere length in hypomethylated cells was mainteined despite significantly increased telomerase activity, Arabidopsis telomeres were markedly shortened while telomerase activity and transcription were unchanged in hypomethylated individuals. Correspondingly, AtTERT (telomerase reverse transcriptase) chromatin was of euchromatic nature in both telomerase-positive and telomerase-negative tissues except of H3K27me3 loading, which was higher in telomerase-negative mature leaves. Analysis of epigenetic state of tobacco TERT chromatin (DNA methylation, histone modifications) is in progress. Majerova E, Fojtova M, Mozgova I, Bittova M, Fajkus J: Hypomethylating drugs efficiently decrease cytosine methylation in telomeric DNA and activate telomerase without affecting telomere lengths in tobacco cells. Plant Mol Biol 77:371–380 (2011). Ogrocka A, Polanska P, Majerova E, Janeba Z, Fajkus J, Fojtova M: Compromised telomere maintenance in hypomethylated Arabidopsis thaliana plants. Nucleic Acids Res, in press (2014). Funded by Czech Grant Agency (P501/11/0596), CEITEC (CZ.1.05/1.1.00/02.0068) and by the European Social Fund (CZ.1.07/2.3.00/20.0189). Presented by: Fojtova, Miloslava 114 Poster 43 A high content screen for telomerase trafficking regulators uncovers the cytosolic chaperonin TRiC as essential for TCAB1 folding and telomerase function Adam Freund, Steven Artandi Stanford University School of Medicine The identification of telomerase regulators is difficult due to the low levels of telomerase in most cells and subsequent challenges developing high-throughput screening assays for telomerase activity. The identification of TCAB1, a telomerase component required for the trafficking of telomerase to telomeres, offers a unique opportunity to develop a cell-based readout for telomerase activity, due to the easily detectable localization of TCAB1 to Cajal bodies. In a whole genome, high-content siRNA screen for inhibitors of TCAB1 localization, we identified the cytosolic chaperonin CCT/TRiC as required for TCAB1 protein homeostasis. We determined that TCAB1 is an obligate TRiC substrate: without TRiC, TCAB1 cannot properly fold, cannot bind hTERC, and telomerase localization is perturbed. This TRiC:TCAB1 interaction is mediated by the WD40 repeat domain of TCAB1. Furthermore, we found that all known disease-causing TCAB1 mutants are defective in precisely this folding step, providing a molecular explanation for these cases of human telomere dysfunction. Presented by: Freund, Adam 115 Poster 44 The Diversity and Evolution of Telomeres in Algae Jana Fulneckovaa,*, Tereza Sevcikovab, Jiri Fajkusa, Alena Lukesovac, Marek Eliasb, Eva Sykorovaa a CEITEC MU Kamenice 5/A2 CZ-62500 Brno & Inst. of Biophysics, ASCR,v.v.i., Královopolská 135, CZ-61265 Brno, Czech Republic b Department of Biology and Ecology, Faculty of Science, University of Ostrava, Chittussiho10, CZ-71000 Ostrava, Czech Republic c Institute of Soil Biology, Biology Centre ASCR,v.v.i.,Na Sadkach 7, CZ-37005 Ceske Budejovice, Czech Republic Algae are of interest for telomere biology, because they represent a substantial portion of the eukaryotic phylogenetic diversity. We employed an experimental approach to probe telomeric minisatellites. Our results support the view that there are few variants of the telomeric motifs that are conserved in particular groups of algae. Telomeric motif TTTAGGG, conserved in most green algal lineages, is ancestral for Chloroplastida. However, an interesting diversity of telomeres was described in the order Chlamydomonadales, where at least two independent evolutionary transitions from the ancestral type to TTTTAGGG occurred; moreover a change to TTAGGG was found in a subclade of Caudivolvoxa. Algae of the streptophyte class Klebsormidiophyceae possess TTTTAGGG telomeric repeat or a novel TTTTAGG motif. The TTTAGGG telomeric sequence is also present in telomeres of the photosynthetic alveolate Chromera velia and of Xanthophyceae, in contrast to the presence of the TTAGGG motif in other ochrophytes studied. Glaucophytes and haptophytes exhibit TTAGGG telomeric type. To expand knowledge about the distribution of different telomere types in framework of the eukaryotic phylogeny, we performed in silico analyses of genomic data from major eukaryotic lineages. These analyses confirm the TTAGGG telomeric repeat as the most common and possibly ancestral in eukaryotes, but alternative motifs replaced it along the phylogeny of diverse eukaryotic lineages. This work was supported by research grant from Czech Science Foundation 13-06595S, and by the European Social Fund (CZ.1.07/2.3.00/20.0189). Presented by: Fulneckova, Jana 116 Poster 45 Chromosomal instability and cancer cell stemness in the alternative lengthening of telomeres Fani-Marlen Roumelioti, Maria Chiourea, Christina Raftopoulou, Sarantis Gagos Laboratory of Genetics, Biomedical Research Foundation of the Academy of Athens Greece Extensive chromosomal instability in neoplasia (CIN) leads towards the selection of highly imbalanced cancer genomes. Many cancers are driven by cancer stem cells (CSCs) that may differentiate into a variety of cell types while maintaining the ability to self-renew. Human immortalized cell lines utilizing the Alternative Lengthening of Telomeres (ALT), lack telomerase activity and exert high rates of ongoing telomere dysfunction and CIN. To identify putative CSCs in the ALT-pathway, we combined single cell analysis by M-FISH/SKY, with a-CGH and Immunocytochemistry, in two human ALT cell lines, before and after exposure to ionizing radiation, topoisomerase inhibition, or DNA hyper-replication. Inducible genotoxic stress was convoyed by increased rates of random structural chromosome aberrations, polyploidization, and elevated frequencies of cells expressing the mesenchymal CSC marker CD133. Molecular karyotyping revealed several DNA damage-induced novel clonal structural chromosomal rearrangements. However by aCGH, the insulted ALT genome displayed remarkable propensity to maintain the major genomic imbalances of the control cells. Structural CIN was unequally dispersed between co-dividing cells both in control and challenged cell cultures. Mitotic survivors of structural CIN were products of polyploidization or polyploidy reduction and their percentages corresponded to the fraction of CD133+ cells/population. Furthermore, enrichment of CD133+ cells in culture, showed significant decrease at the frequencies of telomere dysfunction foci (TIF) and in random structural CIN, whereas the rates of whole genome endoreduplication or polyploidy reduction were highly elevated. Our results demonstrate a continuous process of ALT cancer genome homeostasis that is related to genotoxic therapy resistance and cancer cell stemness. Presented by: Gagos, Sarantis 117 Poster 46 Yeast Rap1 binds dsDNA in multiple binding modes Erik Feldmann, Roberto Galletto Department of Biochemistry and Molecular Biophysics Washington University School of Medicine, Saint Louis, MO 63110 Yeast Repressor activator protein 1 (Rap1) is a central player in establishing the shelterin-like complex at telomeres. In addition, Rap1 plays fundamental roles both as a transcriptional activator at ribosomal protein genes and glycolytic genes and as a repressor at the silencing mating-type loci. The data currently available provide a relatively simple picture where Rap1 binds with high affinity as a one-toone complex to its recognition sequences. In this work we show that on model DNA substrates containing a Rap1 recognition sequence, the DNA-binding domain of Rap1 (DBD) is able to access higher stoichiometries than expected. The data strongly suggest that the DBD is able to bind in multiple DNA-binding modes. We further show that the ability of the DBD to bind with higher stoichiometry than a 1:1 is modulated by the spacing between the two half-sites of the recognition sequence. Moreover, the data show that the ability to access different binding modes on DNA is strongly regulated by the C-terminal wrapping loop, observed in the crystal structure of the DBD. Also, access to different binding modes is not specific to the DBD but is conserved in larger Rap1 constructs, albeit the presence of additional domains appears to provide regulative regulatory roles. These novel findings suggest that the DNA-binding mode of Rap1 is dynamic and this may offer an additional mechanism of regulation for Rap1 that was previously unanticipated. Possible functional implications of our observations are discussed. Presented by: Galletto, Roberto 118 Poster 47 Telomere uncapping impairs K-RasG12V-induced lung carcinogenesis Maria Garcia-Beccariaa,*, Paula Martineza, Marta Cañamerob, Francisca Muleroc, Maria Blascoa a Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), b Histopathology Unit, Biotechnology Program, Spanish National Cancer Research Centre (CNIO) c Molecular Imaging Unit, Biotechnology Program, Spanish National Cancer Research Centre (CNIO) Lung cancer is the leading cause of cancer related deaths worldwide. In spite of some success of targeted therapies, new therapeutic strategies are needed. Telomeres are considered potential anticancer targets, as telomere maintenance above a minimum length is necessary for cancer cell growth. Indeed, inhibition of telomerase is being currently tested in various human cancer clinical trials. Telomerase inhibition, however, has the potential shortcoming of telomere length heterogeneity within tumors, which may result in selection of cancer cells with long telomeres. In addition, telomerase abrogation in mouse cancer models decreases tumor growth only after several mouse generations when telomeres reach a critically short length. Here, we address the induction of acute telomere uncapping by abrogation of the shelterin component TRF1 as an alternative strategy to target telomeres in cancer, independently of telomere length. To this end, we used the well-established KRasG12V lung cancer mouse model, shown to recapitulate the initiation and progression steps of human lung carcinogenesis. TRF1 abrogation reduced the size and malignancy of K-RasG12V-induced lung tumors, as well as increased mouse survival, compared to wild-type controls. The anti-tumorigenic effect of Trf1 deletion was concomitant with induction of telomeric DNA damage and apoptosis in the tumors. TRF1-abrogated tumors also showed less proliferation, G2-arrest, and signs of endoreduplication. These results demonstrate that Trf1 deletion impairs the growth and progression of tumors associated to increased oncogenic stress by induction of DNA damage and severe mitotic defects. Thus, induction of acute telomere uncapping emerges as a potential new therapeutic target for lung cancer. Presented by: Garcia-Beccaria, Maria 119 Poster 48 Interactions between the Elg1 clamp-unloader and the DNA damage checkpoint kinase Dun1 Inbal Gazy, Martin Kupiec Department of Molecular Microbiology and Biotechnology, Tel-Aviv University Ramat Aviv, 69978, Israel Elg1 is a protein with homology to Rfc1 that forms an RFC-like complex whose precise molecular function is still unkonwn. The Elg1 RFC-like complex interacts with the replication clamp PCNA, particularly when it becomes SUMOylated. It has been proposed that the role of Elg1 is to unload PCNA from stalled replication forks and from Okazaki fragments to allow their maturation. Deletion of ELG1 leads to a variety of genomic instability phenotypes, including, among others, hyper-recombination, chromosome loss and increased telomeric silencing. In addition, Elg1 plays a role in telomere regulation, and its deletion leads to elongated telomeres. In order to better understand Elg1’s role we investigated the synthetic interaction of ELG1 and several other genes known to play a role in telomere length regulation. We found that deletion of the gene encoding the protein kinase Dun1 supresses Elg1’s telomeric length phenotype. This kinase is under regulation of the DNA damage response and controls the level of dNTPs in the cells and deletion of DUN1 results in a reduction of the dNTP cellular pool. Accordingly, we found that elevation of the dNTP pools abolishes dun1 supression of elg1 elongated telomeres. We are currently investigating the mechanisms by which Dun1 and dNTPs affect Elg1's activity. Presented by: Gazy, Inbal 120 Poster 49 Telomeric Protein TRF2 Is an Angiogenic Target of WT1 That Regulates the Expression of PDFGRβ Mounir El Mai, Kay Wagner, Jean-François Michiel, Valerie Renault, Nicole Wagner, Eric Gilson Institut for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia-Antipolis, CNRS UMR7284/INSERM U1081, Faculty of Medicine, Nice, France We provide evidence that TRF2 is expressed in the vasculature of most human cancer types, where it co-localizes with the Wilms’ tumor suppressor WT1. We further show that TRF2 is a direct target of WT1 and is required for proliferation, migration, and tube formation of endothelial cells. These angiogenic effects of TRF2 are uncoupled from its function in telomere capping. In fact, TRF2 regulates the expression of the angiogenic tyrosine kinase PDGFRβ. These findings demonstrate a role of TRF2 in neoangiogenesis and delineate a distinct function of TRF2 as a regulator of angiogenic factor expression. Presented by: Gilson, Eric 121 Poster 50 Keeping telomere under wraps to avoid DNA damage: a job for TRF2 Delphine Bennaroch-Popivkera, Sabrina Pisanoa, Alexandre Gaya, Aaron Mendez-Bermudeza, Chrysa Latricka, Bei Peia, Simona Mironb, Marie-Hélène Le Dub, Eric Gilsona, Marie-Josèphe Giraud-Panisa,* a Institute for the Research on Cancer and Aging, Nice, UMR 7284 CNRS, U1081 INSERM, Faculté de Médecine, Université de Nice-Sophia Antipolis, Nice, France b DSV/IBi Tec-S/SB2SM/LBSR, Commissariat à l'Energie Atomique Saclay, Gif sur Yvette, France The human shelterin protein TRF2 is capable of condensing DNA in vitro, a property that is thought to be important for telomeres folding and during telomeres replication (Amiard et al, NSMB, 2007, Ye et al, Cell, 2010, Poulet et al, NAR, 2012). Using Atomic Force Microscopy, we have discovered that this topological activity of TRF2 is based on the wrapping of 100bp of DNA around the protein. Protein footprinting performed on the TRF2-DNA complex show that this wrapping involves residues in the homodimerization (TRFH) domain organized as a “DNA path” around this domain. Mutations of several of these residues gave rise to a mutant that we dubbed Top-less since it exhibits a topology activity largely reduced as seen by AFM, topology assays and single strand invasion assays, while it is still capable of binding telomeric DNA. Amazingly, expression of this mutant does not rescue the activation of the DNA damage response triggered at telomeres by TRF2 downregulation. We propose that TRF2dependent DNA wrapping is necessary for telomere protection against unsuited activation of the DNA damage response. Presented by: Giraud-Panis, Marie-Josèphe 122 Poster 51 RTEL1 mutations cause Hoyeraal-Hreidarsson syndrome by disrupting an essential function of RTEL1 in telomerase-dependent telomere maintenance Galina Glouskera,*, Zhong Dengb, Arturo Londoño-Vallejoc, Paul M. Liebermanb, Yehuda Tzfatid a Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem, 91904, Israel b Gene Expression and Regulation Group, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA. c Telomeres & Cancer Laboratory, Institut Curie, 26 rue d'Ulm, 75248 Paris, France; UPMC Univ. Paris 06, F-75005 Paris, France. “E uipe Labellis e Ligue Contre le Cancer” d Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem, 91904, Israel. Hoyeraal-Hreidarsson Syndrome (HHS), a severe form of the inherited telomeropathy dyskeratosis congenita (DC), is characterized by bone-marrow failure, immunodeficiency and developmental defects. HHS-causing mutations were found in DKC, hTERT, TINF2, and recently also in RTEL1, encoding a DNA helicase. Rtel1 was identified in mouse to be genetically associated with telomere length and RTEL1 deficiency in HHS patients was associated with severely short telomeres in blood leukocytes, implicating RTEL1 in telomere length maintenance. How RTEL1 regulates telomere length remains unclear. RTEL1-deficient cells display shortening of the telomeric 3’ overhang and telomere fragility1. An increase in telomere (T)- circles was detected by a rolling-circle DNA polymerization assay2 but not by two-dimensional gel electrophoresis1. However, telomere deletions caused by breaks or excision of T-circles do not readily explain the shortening of telomeres in telomerase-positive RTEL1-deficient cells and the normal telomere length in telomerase negative primary fibroblasts, while both telomerase positive and negative cells display telomere dysfunction-induced foci and telomere fragility. We are using lymphoblastoid cells and fibroblasts carrying single and compound heterozygous RTEL1 mutations, and a tetracycline-inducible system for the regulated expression of each of the RTEL1 splice variants. This system enables studying the different phenotypes associated with each variant, and with the disease-causing mutations. Our results suggest that RTEL1 is essential for telomerase action at telomeres; mutations in RTEL1 compromise the ability of telomerase to elongate telomeres, resulting in severe telomere shortening, which is the primary cause for HHS. 1Deng et al. (2013) PNAS 110:e3408-16 2Ballew et al. (2013) PLoS Genetics 9:e1003695 Presented by: Glousker, Galina 123 Poster 52 Development of a high-content, automated platform for rapid analysis of alternative lengthening of telomeres (ALT)-associated promyelocytic leukemia nuclear bodies (APBs) in human cancer cells David Halvorsen, Thomas Hunt, Manali Aggrawal, Haroldo Silva SENS Research Foundation Most cancers prevent telomere loss, senescence and apoptosis by expressing telomerase. Yet, about 10-15% of cancers employ a telomerase-free, homologous recombination-based mechanism known as alternative lengthening of telomeres (ALT). Unlike the telomerase-based pathway, no ALT-specific targets or therapeutics are yet available due in part to a lack of robust ALT assays. Here we develop and validate a high-speed, high-content version of the established ALT-associated promyelocytic leukemia (PML) nuclear body (APB) assay, wherein co-localization of PML with telomeric chromatin (e.g., TRF2) occurs prominently in ALT cells. Currently, the APB assay is impractical for high-throughput applications as it requires complex three-dimensional (3-D) confocal image acquisition, reconstruction and analysis using a supercomputer for robust statistical results. Here we use the Molecular Devices ImageXpress Micro Widefield High Content Screening System and MetaXpress imaging and analysis software running on a regular desktop computer for automated imaging and analysis of APBs in human ALT cells in multi-well plates. We validated our platform by treating cells with agents previously reported to modulate APB levels. During image acquisition, a maximum pixel intensity projection algorithm collapses Z-stacks into a two-dimensional (2-D) image. This platform reproduces key published data (e.g., average number of APBs per nuclei) in a less data-intensive format (2-D vs. 3-D) and therefore it is about 5-6 times faster than previous methods while keeping similar statistical power. Our platform provides useful parameters for the standardization of the APB assay and a powerful tool to screen drugs and genetic targets for functional impact on ALT activity. Presented by: Halvorsen, David 124 Poster 53 PARP1 and ATRX are Required for Proper Telomere Length Maintenance Adam Harveya,*, Duncan Bairdb, Eric Hendricksona a b University of Minnesota Cardif University Telomeres are the repetitive DNA:protein elements that protect the ends of linear chromosomes. Here, I describe the distinct roles of two genes not attributed to canonical telomere maintenance per se: PARP1 and ATRX, using genetic knock-out approaches in human somatic cells. PARP1 is known for its role in DNA break sensing and for its synthetic lethality with BRCA1/2 gene mutations in inherited breast and ovarian cancers. Unexpectedly, however, I have uncovered an underappreciated role for PARP1 in maintaining telomere length and integrity in human cells. Additionally, in collaboration with the laboratory of Duncan Baird, we have preliminary data suggesting that cells lacking PARP1 rarely escape telomere crisis induced by gradual telomere loss. The second accessory factor in my studies, ATRX, is a histone chaperone for the histone H3 variant H3.3. Mutations in ATRX have been strongly implicated in the genesis of alternative lengthening of telomeres-positive (ALT+) tumors. To experimentally test whether the absence of ATRX is necessary and/or sufficient for the genesis of ALT in the context of different cellular settings, I have functionally inactivated ATRX in 2 different cell types: i) a telomerase-positive, transformed and immortalized cancer cell line {HCT116} and ii) a telomerase-negative, transformed, but not immortalized cell line (JFCF). The inactivation of ATRX in HCT116 cell line did not lead to the formation of ALT. In contrast, when JFCF cells were subjected to gene targeting, 11 independent clones immortalized, were correctly gene targeted, lacked ATRX expression, and all have an ALT-phenotype. Presented by: Harvey, Adam 125 Poster 54 Local telomerase over-expression promotes cancer in a tissue-dependent manner Catarina M. Henriques, Miguel Godinho Ferreira Instituto Gulbenkian de Ciência, Portugal Ageing is characterized by an overall decline in tissue homeostasis, which sets a fertile ground for disease. In humans, progressive telomere dysfunction is a good candidate to explain tissue degeneration with age. Telomeres shorten during our lifespan. Critically short telomeres can impact tissue homeostasis both by cell autonomous (as a barrier to cell proliferation) and non-cell autonomous (by accumulation of senescent cells) mechanisms. Consistently, telomere dysfunction leads to premature ageing and disease in humans. However, it is currently not known whether functional telomeres are the limiting factor safeguarding from ageing. We have established zebrafish as a key model to address this question. We have shown that zebrafish, like humans; require telomerase for health and lifespan. In this model, telomere dysfunction leads to premature ageing, triggering systemic tissue degeneration in a time and tissue-dependent manner. We hypothesize that rescuing telomere dysfunction in key tissues may prevent age-associated degeneration. Our data suggest that local telomerase expression is sufficient to partially rescue telomerase mutant phenotypes, including both gut and muscle degeneration. This effect is likely to be dose and/or tissue dependent. Remarkably, telomerase over-expressing cells do not directly outcompete tert-/- cells in tissue-regeneration assays, suggesting they are not transformed/malignant. However, localized telomerase over-expression in otherwise tert-/- mutants (displaying old age phenotypes) appears to lead to hyperplasia and cell transformation. Thus, the “aged” microenvironment may itself promote the transformation of cells with un-restricted telomerase and, consequently, lead to cancer. These results impart caution for telomerase-activating therapies for tissue regeneration in old individuals. Presented by: Henriques, Catarina M. 126 Poster 55 Which domains are critical for TRF2-REST interaction? Milan Hluchý, Jan Paleček, Ctirad Hofr Chromatin Molecular Complexes, CEITEC and Faculty of Science, Masaryk University, Brno, CZ-62500, Czech Republic TRF2, one of the six shelterin subunits, is a key player in protection of telomeres from DNA-damage response pathways. Recently discovered interaction between TRF2 and protein REST showed new nontelomeric function of TRF2. REST is a master repressor in neuronal progenitor cells. TRF2 protects REST from proteasome degradation and helps to stall the neuronal progenitors in undifferentiated phase. This work aims to characterise the interacting domains on both proteins and determine the specificity of TRF2-REST interaction. Both TRF2 and REST were divided to three truncated protein variants according to their functional domains. REST proteins expressed in E. coli cells were used as bait in pulldown assay together with radioactively labelled TRF2 prey. Fragments with candidate interacting domains were used for further in vitro studies with surface plasmon and fluorescence spectroscopy. Both proteins interact physically with each other in vitro. The interacting domain in REST protein was found in the linker region covering amino acid residues 438 to 859. A candidate REST-interacting domain on TRF2 is in the TRFH dimerisation domain. Further investigation of these unexpected interacting partners can help to understand the dual role of REST in different cancer types where it can act both as a tumour suppressor or oncogene. This work was supported by CEITEC (CZ.1.05/1.1.00/02.0068), the European Social Fund (CZ.1.07/2.3.00/30.0019), GACR (P205/12/0550), and KONTAKT II (LH13054). Presented by: Hluchý, Milan 127 Poster 56 A rabbit ear mechanism by which human Rap1 modulates TRF2 attraction to telomeric DNA Eliška Janoušková, Ivona Nečasová, Michal Zimmermann, Milan Hluchý, Ctirad Hofr Chromatin Molecular Complexes, CEITEC and Faculty of Science, Masaryk University, Brno, CZ-62500, Czech Republic Regardless of a successful genetic research that has identified and assign main biological functions of shelterin proteins that safeguard telomeres, a molecular mechanism of how each protein subunit contributes to the protecting function of the whole shelterin complex remains elusive. Effects of full-length Rap1 protein on TRF2 binding to double-stranded DNA were investigated by quantitative biochemical approaches. We observed that Rap1 reduces the overall DNA binding affinity of TRF2. Despite the reduced DNA affinity in Rap1 presence, the binding selectivity of TRF2 to telomeric DNA is increased significantly. The lower salt dependence of the dissociation constant for TRF2 binding to DNA in presence of Rap1 indicates the neutralization of the total net charge of Rap1-TRF2 complex. The less pronounced electrostatic attraction between TRF2 and DNA in Rap1 presence explained the improved selectivity of TRF2 to telomeric DNA. Thus, Rap1 prompts more accurate and selective recognition of telomeric DNA by TRF2. Based on our quantitative measurements and known structural data, we speculate that Rap1, after binding to TRF2, might shield the positively charged basic domain on the N-terminus of TRF2 from mainly non-specific electrostatic interaction with DNA. An interaction model which resembles a happy and sad rabbit ear will be presented. The results of these quantitative studies contribute to the understanding of the selective recognition of telomeric DNA by the whole shelterin. This work was supported by CEITEC (CZ.1.05/1.1.00/02.0068), the European Social Fund (CZ.1.07/2.3.00/30.0019), GACR (P205/12/0550), and KONTAKT II (LH13054). Presented by: Hofr, Ctirad 128 Poster 57 Telomere transcripts without subtelomere sequences improve targeting of telomerase in cancer cells Sandra Sampl, Christian Stern, Doris Mejri, Klaus Holzmann Division of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University Vienna, Austria Transcripts from telomeres called TERRA were identified to bind and block telomerase activity (TA). TERRA can further function to guide the telomerase complex to specific chromosome ends. Therefore, TERRA without subtelomere sequences may act as decoy molecule independent of direct TA inhibition. We expressed decoy TERRA transcripts in cell culture cells to study in vitro cell growth and viability directly and in combination with telomerase inhibitors. Adeno- and lentivirus constructs (AV and LV) were established for transient and stable recombinant expression of approximately 120 units of UUAGGG repeats in sense (S) or anti-sense (AS) orientation under control of hH1 or CMV promoter. Telomere length (TL) and expression was analyzed by qPCR, TA by TRAP assay. Telomerase inhibitors and AVs expressing dominant-negative telomerase and shRNA against hTERT were used in MTT cell viability and clonogenicity assays. Moderate 1.5-1.7 fold elevation of TERRA-S compared to endogenous transcripts caused reduction of TA to 23-38% in tumor cell lines. In contrast, TERRA-AS expression reduced 3-8 fold endogenous TERRA without reduction of TA. TL, Population doubling time, cell viability and clonogenicity were not affected by TERRA-S/AS expression. Significant improvement of telomerase inhibition was identified as IC-50 values and clone formation capacities decreased 2.0-2.6 and 1.3–1.7 fold, respectively, in LV cell clones expressing moderate TERRA-S compared to TERRA-AS and controls. Fibroblasts showed 15-fold increased TERRA expression compared to tumor cells and were not affected by treatments applied. Our results suggest TERRA oligonucleotides as candidates for in vivo application in combination with telomerase targeting approaches. Presented by: Holzmann, Klaus 129 Poster 58 Telomerase modulates cyclinD1 gene in concert with NOL1 Juyeong Honga,*, Jihoon Leeb, In Kwon Chungc a Department of Integrated Omics for Biomedical Science, Yonsei University, Seoul, 120-749, Korea. Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea. c Department of Integrated Omics for Biomedical Science and Department of Systems Biology, Yonsei University, Seoul, 120-749, Korea. b Telomerase is upregulated in many human cancers. Telomerase, which is composed of telomerase reverse transcriptase (TERT) and the associated RNA component (TERC), adds telomeric repeats, thus allowing cells to keep dividing. Although recent studies have uncovered that extra-telomeric TERT functions as a transcription factor in oncogenic signaling pathway beyond de novo synthesis of telomere, regulatory mechanisms are still less understood. Here, we identify NOL1 as a transcription factor that regulates cyclinD1 gene expression by association with TERT, independently of telomerase activity. We find that NOL1 interacts with TERT using TERC and directs to TCF binding element (TBE) site of the cyclin D1 gene promoter. NOL1 does not affect the subcellular localization of TERT, telomerase activity, and telomere dysfunction induced foci (TIF) formation. Furthermore, Chromatin immunoprecipitation and luciferase analyses show that depletion of NOL1 prevents TERT from binding and activating CyclinD1 genes, which delays G1/S transition, subsequently leading to cell-cycle arrest. Together, these findings suggest a novel mechanism that contributes to understanding how TERT plays a role as a transcription factor in cellular proliferation. Presented by: Hong, Juyeong 130 Poster 59 Human Rap1 Induces TRF2 Release from Telomeric DNA Eliška Janoušková, Ivona Nečasová, Michal Zimmermann, Milan Hluchý, Ctirad Hofr Chromatin Molecular Complexes, CEITEC and Faculty of Science, Masaryk University, Brno, CZ-62500, Czech Republic Telomeric proteins TRF2 and Rap1 are components of a multi-subunit human protein complex called shelterin. Shelterin alters the spatial arrangement of telomeres by formation of specific structures, thereby affects telomerase and other enzymes access to DNA. In this way, shelterin proteins participate in the processes of aging and tumor cell transformation. Here we investigate the effect of protein Rap1 on TRF2 binding to double-stranded telomeric DNA. The quantitative analysis using fluorescence anisotropy, surface plasmon resonance, isothermal titration calorimetry and gel electrophoresis revealed that the binding affinity of TRF2 to Rap1 is comparable with the binding affinity of TRF2 to DNA. The presence of Rap1 reduces the overall DNA binding affinity of TRF2, however, increases the binding selectivity of TRF2 to telomeric DNA. Our results demonstrate that Rap1 induces a partial release of TRF2 from a preformed TRF2-DNA complex. In Rap1 presence, TRF2 affinity to DNA depends less on salt concentration, which shows that the specific TRF2-DNA interaction prevail; TRF2 binds telomeric DNA more selectively. The observed Rap1 induced charge neutralization might contribute to TRF2 release from DNA. Thus, Rap1 improves significantly the accuracy of the selective recognition of telomeric DNA by TRF2 and hence the whole shelterin complex. This work was supported by CEITEC (CZ.1.05/1.1.00/02.0068), the European Social Fund (CZ.1.07/2.3.00/30.0019), GACR (P205/12/0550), and KONTAKT II (LH13054). Presented by: Janoušková, Eliška 131 Poster 60 Influence of environmental factors on telomerase-independent survivor formation in fission yeast Kristi Jensena,*, Baumann Peterb a b HHMI and Stowers Institute for Medical Research HHMI and Stowers Intitute for Medical Research; Dept. of Mol and Int. Physiology, KUMC Whereas most human somatic cells do not express the catalytic subunit of telomerase; reactivation of telomerase is a signature of many human cancers. This lends clinical significance to finding a telomerase inhibitor that can be used as a chemotherapeutic drug. However, a sub-set of cancers lack active telomerase and chromosome ends are maintained by a telomerase independent process called alternative lengthening of telomeres (ALT). As we are learning more about the mechanisms underlying telomerase-independent telomere maintenance, it will be important to understand how readily cells can switch between different pathways to replenish telomeric sequences. In fission yeast, loss of telomerase results in a population of cells that initially experience chromosome instability and crisis after which telomerase-independent survivors arise. Three classes of survivors have been described: (i) so called circular survivors in which all three chromosomes have undergone circularization and chromosome ends are absent; (ii) linear survivors with long heterogeneous telomeres maintained through recombination reminiscent of human ALT cells; and (iii) HAATI survivors that accumulate large amounts of repetitive heterochromatic sequence at chromosome ends. How environmental conditions affect the choice of pathway that maintains telomeres in the absence of functional telomerase and whether cells can switch readily between pathways is poorly understood. We will present results on factors and conditions that affect survival in the absence of the RNA subunit of telomerase. Presented by: Jensen, Kristi 132 Poster 61 Sequence variants of the telomerase reverse transcriptase (TERT) gene in Nicotiana tabacum Jana Jureckovaa,*, Eva Sykorovab, Miloslava Fojtovac a Faculty of Science, Masaryk University Brno, Czech Republic Institute of Biophysics, ASCR, Czech Republic c CEITEC, Masaryk University Brno, Czech Republic b Study of telomeres evolution includes the analysis of arrangement of telomere and subtelomere regions and characterisation of the telomerase in different model organisms. Telomerase is a ribonucleoprotein enzyme complex providing the synthesis of telomeric repeats at the ends of eukaryotic chromosomes. Telomerase RNA (TR) serves as the teplate for elongation of the telomere repeat motif. The gene coding for catalytic subunit of telomerase - telomerase reverse transcriptase (TERT) is developmentally regulated and its transcription reflects the activity of telomerase in the tissue. Nicotiana tabacum (tobacco), an allotetraploid model plant, is the only plant representative in which more sequence variants of the TERT gene were identified (Sýkorová et al., 2012). Two NtTERT variants (NtTERT-C/s and NtTERT-D) were inherited from the tobacco‘s diploid progenitor Nicotiana sylvestris and one NtTERT variant (NtTERT-C/t) from Nicotiana tomentosiformis. The aim of our study was to analyse copy number of NtTERT-C/s, NtTERT-C/t and NtTERT-D variants in N. tabacum genome using qPCR and to determinethe transcription of these variants in tissues with different telomerase activity like seedlings, leaves, buds and roots by qRT-PCR. Sykorova E, Fulneckova J, Mokros P, Fajkus J, Fojtova M, Peska V. Three TERT genes in Nicotiana tabacum. Chromosome Res. (2012). The research is supported by Masaryk University Brno (MUNI/C/0979/2013), by Czech Science Foundation (13-06943S) and by the project “CEITEC - Central European Institute of Technology” (CZ.1.05/1.1.00/02.0068) from the European Regional Development Fund. Presented by: Jureckova, Jana 133 Poster 62 Fission yeast without subtelomeres Sanki Tashiro, Yuki Nishihara, Junko Kanoh Institute for Protein Research, Osaka University Recent studies have uncovered the functions of telomere, such as the maintenance of telomere DNA length, the protection of chromosomal ends, and the regulation of chromosome movements in mitosis and meiosis. However, little is known about the physiological roles of subtelomeres. S. pombe is an ideal organism for the analyses of subtelomere because it has only three chromosomes. The subtelomeres on chromosomes I and II consist of approximately 50 kb-long highly homologous DNA sequences, and the subtelomere on chromosome III contains the partial DNA sequence (approximately 15 kb-long) homologous to the other subtelomeres. To investigate the physiological roles of subtelomere, the all subtelomeres were deleted from the S. pombe chromosomes. The SD (subtelomere-deleted) strain was viable with the similar growth rate to that of the wild-type. It also showed the normal telomere DNA length, indicating that the subtelomeres are dispensable for the cell growth and the telomere DNA maintenance. Interestingly, the subtelomere deletion led to the invasion of heterochromatin to the subtelomere-adjacent regions (approximately 50 kb-long), which resulted in the silencing of genes located at the heterochromatin-invaded regions. These data suggested that the subtelomeres serve as buffer zones against the heterochromatin invasion to maintain gene expressions. Presented by: Kanoh, Junko 134 Poster 63 Multiple interactions within a primer influences Saccharomyces castellii telomerase activity in vitro Cecilia Gustafsson, Ahu Karademir, Roopesh Krishnankutty, Marita Cohn Department of Biology, Genetics group, Lund University, Lund, Sweden The telomerase ribonucleoprotein enzyme maintains genome integrity by elongating the telomeres. Telomerase adds tandem repeats on the telomeric DNAs via using its intrinsic RNA template. Translocation of the DNA 3´end on the RNA template provides telomerase to add several repeats on the substrate. The budding yeast Saccharomyces castellii synthesizes several 8-mer repeats (CTGGGTGT) onto the primer in vitro. In this work, we determined the the primer extension mechanisms of S. castellii telomerase by using an in vitro primer direct assay. We showed that a six nucleotide long primer and a single-stranded 3’ overhang of four nucleotides are efficiently extended. Moreover, we showed that multiple interactions within a primer influences the telomerase activity of S. castellii in vitro. While S. castellii telomerase is not able to utilize the non-telomeric 3’ ends of short primers, the interaction with the 5’ end of long primers can stimulate the extension of non-telomeric 3’ ends. Our results reveal intriguing details about the S. castellii telomerase activity. Presented by: Karademir, Ahu 135 Poster 64 Dissecting the telomeric and non-telomeric roles of human CTC1 through gene disruption Christopher Kasbek, Jason Stewart, Mary Chaiken, Caitlyn Goodwin, Carolyn Price University of Cincinnati Human CST (CTC1-STN1-TEN1) functions in DNA replication at telomeres and other genomic sites in response to replication stress. Mutations in the CTC1 subunit cause the disease Coats plus and can lead to dyskeratosis congenita. STN1 depletion revealed that CST functions in telomere duplex replication, C-strand fill in, and genome-wide replication rescue after hydoxyurea treatment. TEN1 depletion suggested additional roles as telomere loss and anaphase bridge formation were more pronounced than with STN1 depletion. However depletion of CTC1 with shRNA resulted in variable phenotypes. To clarify CTC1 function, we generated a conditional gene disruption in human cancer cells. Although CTC1 mRNA is undetectable 2 days after gene disruption, phenotypes due to CTC1 deletion are not observed until day 4. Non-telomeric defects arise earlier than telomeric defects. Initial phenotypes include decreased ability to rescue replication after fork stalling and increased anaphase bridge formation in the absence of telomere fusions, however growth rate remains normal. By days 5-7, Goverhang length is increased and chromosome ends lacking telomere signal become apparent. The growth rate decreases slightly. Telomere loss then increases steadily, chromosome fusions appear and DNA damage signaling rises. By three weeks after CTC1 disruption the cells cease to divide. These results support the idea that CST functions in response to multiple types of replication stress. They also suggest that the severity of symptoms in Coats plus patients stem from both telomeric and genome wide deficiencies in DNA replication. Presented by: Kasbek, Christopher 136 Poster 65 Interfacial inhibition of human telomerase reconstitution Guillaume Kellermanna,*, Markus Kaiserb, Florent Dinglic, Olivier Lahunad, Florence Mahuteau-Betzere, Damarys Loewdc, Evelyne Ségal-Bendirdjiana, Marie-Paule Teulade-Fichoue, Sophie Bombarda a INSERM U1007 / Université Paris Descartes Universität Duisburg-Essen c Institut Curie / laboratoire de spectrométrie de masse protéomique, France. d INSERM U-1016, Institut Cochin e Institut Curie / CNRS UMR 176, Orsay, France b Telomerase is an almost universal cancer target lacking of specific and efficient inhibitors. We present herein a method to produce high amount of the catalytic subunit of human telomerase and use it to identify small-molecules interfering with telomerase assembly. This test extends the identification of telomerase inhibitors to a new class of compounds, not detectable by conventionnal assays. Using this method, we identified a new molecule, TAI1, that can completely inhibit telomerase activity when added prior assembly, at a concentration which has no effect on an already assembled telomerase. Direct assay shows that TAI1 prevents the DNA polymerase activity of telomerase, and does not interfere with repeat addition processivity. EMSA showed that TAI1 only slightly reduces the binding of hTERT to hTR, in conditions where telomerase activity is completely inhibited. This means that TAI1 promotes the formation of inactive telomerase complexes. Surprisingly, these inactive complexes are irreversibly inhibited, since their 100fold dilution failed to restore any activity, even after several hours of incubation. These observations suggest that TAI1 is irreversibly trapped inside the telomerase complex, after assembly, in a manner reminiscent of interfacial inhibitors. Structure-activity relationships support a model where TAI1 binds both to hTERT and hTR, thus producing a ternary complexe. In conclusion, our data suggest that potent telomerase inhibitors have escaped detection during all previous screens performed with telomerase already assembled from cancer cells, and these observations therefore recommand the future use of the present method that can both detect assembly and catalytic inhibitors. Presented by: Kellermann, Guillaume 137 Poster 66 Therapeutic agents influence the TRF2-TIN2 interactions and their cellular distribution: study in human cancer cell lines Patrycja Klos, Ctirad Hofr, Ivona Necasova, Jiri Fajkus Central European Institute of Technology, Masaryk University, Molecular Complexes of Chromatin Group Among anti-cancer therapies, the use of chemicals interfering with the expression/function of telosome components has aroused recently much interest. In this study we examined the interactions between TRF2 and TIN2 shelterin proteins in human cancer cells with varied telomere length, treated with therapeutic chemicals that are known to interact with the DNA, which in turn might cause TRF2 redistribution accross the DNA, and/or influence its stability and interacting properties. To compare the number of TRF2-TIN2 interactions between the different cell lines we employed proximity ligation assay (PLA). Immunofluorescence colocalization study was performed to check the distribution pattern of the studied proteins in the cells. The expression level of TRF2 after the drug treatment was measured by qRT-PCR and Western blotting. Significant changes in the number of TRF2-TIN2 interaction spots, their distribution accross the chromatin, as well as TRF2 expression level were observed after treating the cells with the chosen chemicals. Interestingly, the obtained data varied in different studied cell lines. Altogether our results demonstrate a strong impact of the chosen therapeutics on cancer cell telosome, which in turn may have further implications in anti-cancer therapy. This work was supported by CEITEC (CZ.1.05/1.1.00/02.0068), the European Social Fund (CZ.1.07/2.3.00/30.0019), GACR (P205/12/0550), and KONTAKT II (LH13054). Presented by: Klos, Patrycja 138 Poster 67 Telomerase RNP complex assembly in disease background Elzbieta Kowalska, Georgeta Zemora, Michael Wildauer, Christina Waldsich Max F. Perutz Laboratories, Dr. Bohr-Gasse 9, 1030 Vienna, Austria RNA structure is relevant in telomerase ribonucleoprotein complex (RNP) formation, as specific nucleotide polymorphisms in the human telomerase RNA (hTR) have been implicated in disease pathologies. The main protein components of the human telomerase RNP complex are hTERT, a reverse transcriptase, and Dyskerin (DKC1), a protein important for telomerase biogenesis. As for hTR, there are known hot spots of single nucleotide polymorphisms in both of these proteins that are linked to dyskeratosis congenita and/or pulmonary fibrosis. To elucidate the structural basis of disease-linked mutations, I introduced single amino acid changes in these regions of the proteins. For hTERT, the motif B and the linker motif were mutated. In the case of DKC1, I targeted the PUA domain that contacts the hTR. The hTERT and DKC1 constructs containing the disease-related point mutations were expressed by transient transfection together with hTR in HEK293 cells. Employing the SHAPE chemistry, the structure of hTR can be investigated in living cells. After chemical modification, the cells were harvested and total RNA was extracted for reverse transcription. At modified nucleotides the reverse transcriptase will stall or fall off. The different cDNA fragments were separated on a denaturing polyacrylamide gel (PAGE) and intensities of bands were compared between hTR alone and hTR in the presence of the wildtype or point mutated hTERT or DKC1 proteins. The changes in structure of hTR in the presence of wildtype protein or the proteins with disease-related point mutations will provide novel insights into the architecture of the telomerase RNP complex. Presented by: Kowalska, Elzbieta 139 Poster 68 Alternative lengthening of telomeres detected in telomerase-positive canine histiocytic sarcoma Theresa Kreilmeiera,*, Sandra Samplb, Marlene Hauckc, Ingrid Walterd, Klaus Holzmannb, Miriam Kleitera a Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria b Division of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University Vienna, Austria c Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, Raleigh, NC 27607, USA d Vet Core Facility, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria The telomere maintenance mechanism (TMM) used in the absence of telomerase activity (TA) is called alternative lengthening of telomeres (ALT). ALT is found more often in human tumors of mesenchymal than of epithelial origin. Our aim was to screen a cohort of canine sarcomas for prevalence of TMM used. Tumor tissue from 17 spontaneous occurring canine soft tissue sarcomas were analysed for telomere length, TMM, and immunohistochemical expression of ATRX and DAXX. Telomere length was determined by qPCR, telo-qFISH hybridisation and TRF analyses including CHEF gelelectrophoresis. TA and ALT were measured by TRAP assay and the presence of C-circles, respectively. One histiocytic sarcoma showed elevated C-circle levels and long telomeres with heterogeneous lengths similar in appearance as in human cells with ALT. In contrast, TA was detected in 15 of 16 sarcomas including the case with ALT activity. In one sarcoma neither ALT nor telomerase activity was found. The case using two TMMs showed on tissue sections loss in ATRX but not DAXX expression, proteins known to localise at PML bodies. Further, colocalisation of DAXX at telomeres was elevated compared to cases without ALT. In conclusion, ALT exists in canine sarcomas with presumably less prevalence compared to human. Our data indicate some tumor cells use both ALT and TA, but this remains to be confirmed. TA seems to be the dominant TMM and even non-determined TMM might play a role. Therefore, all mechanisms have to be considered for therapeutic approaches. Presented by: Kreilmeier, Theresa 140 Poster 69 Telomere Length Kinetics Assay (TELKA) sorts the Telomere Length Maintenance (tlm) mutants into functional groups Martin Kupiec, Linda Rubinstein, Yaniv Harari, Vera Babin Department of Molecular Micro and Biotechnology, Tel Aviv University, Ramat Aviv 69978, Israel. Genome-wide systematic screens in yeast have uncovered a large gene network (the telomere length maintenance network, or TLM), encompassing more than 400 genes, which acts coordinatively to maintain telomere length. Identifying the genes was an important first stage; the next challenge is to decipher their mechanism of action and to organize then into functional groups or pathways. Here we present a new telomere-length measuring program, TelQuant, and a novel assay, TELKA (Telomere Length Kinetic Analysis), and use them to organize tlm mutants into functional classes. Our results show that a mutant defective for the relatively unknown MET7 gene has the same telomeric kinetics as mutants defective for the Ribonucleotide Reductase subunit Rnr1, in charge of the limiting step in dNTP synthesis, or for the Ku heterodimer, a well-established telomere complex. We confirm the epistatic relationship between the mutants, and show that physical interactions exist between Rnr1 and Met7. We also show that Met7 and the Ku heterodimer affects dNTP formation, and play a role in non-homologous end-joining (NHEJ). Thus, our telomere kinetics assay uncovers new functional groups, as well as complex genetic interactions between tlm mutants. Presented by: Kupiec, Martin 141 Poster 70 Involvement of SRSF11 in cell cycle-specific recruitment of active telomerase to telomeres at nuclear speckles Jihoon Leea,*, Sun Ah Jeongb, Prabhat Khadkaa, Juyeong Hongb, In Kwon Chungc a Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea. b Department of Integrated Omics for Biomedical Science, Yonsei University, Seoul, 120-749, Korea. c Department of Integrated Omics for Biomedical Science and Department of Systems Biology, Yonsei University, Seoul, 120-749, Korea. Telomerase, a unique ribonucleoprotein (RNP) complex that contains telomerase reverse transcriptase (TERT) and the telomerase RNA component (TERC), is required for continued cell proliferation in cancer and stem cells. By adding short telomere repeats to the 3’ ends of chromosomes, telomerase compensates for the progressive loss of telomeric DNA produced by incomplete replication of chromosome ends. Here we identify SRSF11 as a novel TERC-binding protein that localizes to nuclear speckles, subnuclear structures that are enriched in pre-messenger RNA splicing factors. SRSF11 associates with active telomerase enzyme within Cajal bodies and directs it to nuclear speckles specifically during S phase of the cell cycle. On the other hand, telomeres are shown to be constitutively present at nuclear speckles irrespective of cell cycle phase, suggesting that nuclear speckles may be the nuclear sites for telomerase recruitment to telomeres. Depletion of SRSF11 by RNA interference prevents telomerase from associating with nuclear speckles and disrupts telomerase recruitment to telomeres, thereby abrogating telomere elongation by telomerase. SRSF11 also associates with telomeres through the interaction with TRF2, which facilitates translocation of telomerase to telomeres. These findings suggest that SRSF11 acts as a nuclear speckle-targeting factor that plays a major role in the telomerase association with telomeres through the interactions with TERC and TRF2, and provide a new target for modulating telomerase activity in cancer. Presented by: Lee, Jihoon 142 Poster 71 Anticancer mechanism of TMPyP4 in breast cancer cell lines MCF7 and MDA-MB-231 Natalia Lipinskaa,*, Hanna Holysza, Aleksandra Romaniuka, Mariusz Kaczmarekb, Blazej Rubisa a Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Przybyszewskiego 49 St., 60-355 Poznan, Poland b Department of Immunology, Chair of Clinical Immunology, Poznan University of Medical Sciences, Rokietnicka 5D St., 60-806 Poznan, Poland Telomeres, the guanine-rich sequences, play a crucial role in human genome stabilization. Thus, they became a promising target in cancer fighting strategy. One of the potential effective tool in blocking the access of telomerase to telomeres, leading to a limited cancer cell divisions, is application of the telomeric ends (G-quadruplex) stabilizers. It was revealed that a cationic porphyrin, TMPyP4 is an efficient G-quadruplex stabilizing agent and hence might inhibit telomerase activity in human cancer cells. We analyzed the influence of clinically relevant TMPyP4 concentrations on morphology, cell cycle, proliferation, migration and adhesion of MCF7 and MDA-MB-231 breast cancer cells. It was shown that treatment with TMPyP4 caused a time (24, 48, 72h) and concentration (0.5µM 15µM) dependant proliferation decrease of breast cancer cells. Short-term treatment resulted in a significant telomere shortening, while longer times of treatment (up to 20 passages) induced restoration of the original length of telomeres (MDA-MB-231) or even telomere lengthening (MCF7). Surprisingly it was not associated with any significant changes in cell cycle. We suggest that this effect might be related to the mechanism of multidrug resistance since treatment with 0.5µM TMPyP4 caused a significant increase of ABCB1 gene expression after 24h. Interestingly, this effect was abolished after longer incubation time. Additionally, it was revealed that TMPyP4 treatment might affect adhesion and migration properties of breast cancer cells through influence on FAK, p-FAK, β1-integrin and paxillin expression. The work was supported by 2011/03/B/NZ7/00512 project. Presented by: Lipinska, Natalia 143 Poster 72 The Mechanism of Epigenetic Behavior at the Mre11/Rad50 Interface Danielle Tatum, In-Joon Baek, Heewon Park, Arthur Lustig, et al. Tulane University Medical Center We have identified an unusual characteristic that, to date, has only been observed in the mre11A470T allele that, based on crystallography, lies in one of the alpha helical residues present at the HLH domain of Mre11. We find that the pattern of recessive and dominant alleles in these mutants and the outcome is dependent upon the pre-existing chromatin present at the telomere. We have found identical results in CEN plasmid studies and ectopic integration studies of the effect. The pattern of heritability is stable to the genotype that is the recipient of the mutant gene and vise versa. We have ruled out possible copy number artifacts and differential turnover of Mre11 and Mre11A470T. We are finally determining whether this is truly an epigenetic state by determining the The DNA sequence of the mutant strain. We have begun on our next test in determining the mechanism of action of the putative epigenetic effect We have introduced one of the rap1 alleles, rap1-5, into both wild type and mutant chromatin states to determine if disrupted Rap1-based chromatin contributes to or removes the epigenetic heritable state. In this regard, our studies have shown an inability to heal broken chromosomes containing seed sequences while still stably maintaining a shorter telomere size. The inability of allele to act in the healing is unlikely to be due to the abundance of telomerase itself given parallel successful healing experiments with strains with similar sized telomeres e,g, the tel1 allele. As a result we are analyzing only replication at the telomere Presented by: Lustig, Arthur 144 Poster 73 Identification of the subtelomeric sequence of the long non-coding RNA TERRA unveils its role at telomeres Isabel López de Silanes, Osvaldo Graña, Maria Luigia De Bonis, Orlando Domínguez, David G. Pisano, Maria Antonia Blasco Spanish National Cancer Research Centre TERRAs are telomeric repeat-containing RNAs that are transcribed from the subtelomere towards the telomere. However, TERRA’s full se uence remains unknown to date. By using a whole-genome RNAsequencing approach, we have identified novel mouse transcripts arising from the subtelomere of chromosome 18 resembling TERRA in many key features. Namely, they are heterogeneous in size and are bound by TERRA RNA-binding proteins. These transcripts co-localize with TERRA and are found at the telomeres in interphase and metaphases, as well as are regulated in a similar manner to TERRA in response to stress and with induction of pluripotency. siRNA depletion of either these transcripts or TERRA causes an increase in telomere dysfunction-induced foci (TIF). We have also identified the promoter region responsible for their transcription. The identification of the subtelomeric sequence of TERRA will facilitate the design of molecular tools to understand the functional significance of TERRA transcripts in telomere biology and telomere-related diseases. Presented by: López de Silanes, Isabel 145 Poster 74 Epigenetic landscape of terminal and internal telomeric DNA using different plant models Eva Majerova, Terezie Mandakova, Miloslava Fojtova, Jiri Fajkus Department of Functional Genomics and Proteomics, Faculty of Science and Central European Institute of Technology, Masaryk University, CZ-62500 Brno, Czech Republic The epigenetic state of telomeres has been animatedly discussed but has not been clearly described yet. The most studied model plant, Arabidopsis thaliana, shows distinct epigenetic marks in dependence to the used methodology what correlates with the fact that considerable portion of telomeric repeats is found at intrachromosomal positions. One of the postulated hypotheses says that interstitial telomeric repeats (ITR) are heterochromatin while genuine telomeres show rather euchromatic character. Intriguingly, transcription of telomeres is found mostly at ITR with only small portion of TERRA and ARRET transcripts originating from terminal telomeres. Here, we present data on epigenetic landscape of telomeric repeats introducing two plant models – (i) Ballantinia antipoda with large blocks of ITR; and (ii) Nicotiana tabacum, plant with long telomeres and no detectable ITR. Our results show that the distribution of 5mC along telomeres is not uniform but proximal parts of telomeres are less densely methylated. Long tobacco telomeres have ambiguous character; we found marks for constitutive heterochromatin (H3K9me2) as well as for active or developmentally silenced genes (H3K4me3, H3K27me3). Moreover, telomeres are transcribed from both strands, which indicates that tobacco telomeres must include an active transcription start site. As for ITR, we found sparsely as well as densely methylated region, and both of them showed the same heterochromatic pattern (only H3K9me2 was found). Intriguingly, with no respect to different DNA methylation levels, both regions are transcribed to the same extent and from both strands. In addition, transcription of telomeric repeats is tissue-specific in both plants. This work was supported by CEITEC (CZ.1.05/1.1.00/02.0068) and by the European Social Fund (CZ.1.07/2.3.00/20.0043). Presented by: Majerova, Eva 146 Poster 75 Unraveling the changes in telomere composition during senescence, immortalization, and tumorigenic conversion of normal human fibroblasts Jana Majerská, Joachim Lingner Swiss Institute for Experimental Cancer Research (ISREC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, 1015, Switzerland The dynamic composition of telomere-associated proteins allows the cell to address the special challenges of telomere protection and maintenance. Perturbations of telomere function play an essential role in the induction of cellular senescence, genomic instability and cancer. However, the critical protein factors contributing to these processes have remained elusive. To address this question, we have employed a genetically defined cell culture model of tumorigenesis, developed by Hahn et al. [1, 2], which allows us to study telomeric states during different stages of the transformation process. Normal human lung fibroblasts were converted into malignant cells in a step-by-step fashion using serial introduction of genes encoding (a) the catalytic subunit of telomerase, (b) the SV40 large T and small T antigens, and (c) the H-RasV12 oncogene. We will discuss our progress in identifying differences in the telomeric protein composition between these cell lines, using the Quantitative telomeric chromatin isolation protocol (Q-TIP) [3] recently developed in our lab. 1. Hahn WC, Counter CM, Lundberg AS, Beijersbergen RL, Brooks MW and Weinberg RA (1999). Nature 400:464-8. 2. Hahn WC, Dessain SK, Brooks MW, King JE, Elenbaas B, Sabatini DM, DeCaprio JA and Weinberg RA (2002). Mol Cell Biol 22:2111-23. 3. Grolimund L, Aeby E, Hamelin R, Armand F, Chiappe D, Moniatte M and Lingner J (2013). Nat Commun 4:2848. Presented by: Majerská, Jana 147 Poster 76 Differential processing of double strand breaks and telomeres at the nuclear envelope in S. cerevisiae Isabella Marcomini, Chihiro Horigome, Susan M. Gasser Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland Telomeres, in yeast as in some higher eukaryotes, are anchored at the nuclear envelope, and this anchorage is necessary to prevent ectopic recombination and genomic instability. The anchorage pathway in S phase, dependent on yKu80-Est1 interaction, requires the SUN domain protein Mps3. When they become critically short in absence of telomerase, telomeres have been shown to colocalize with nuclear pores. Our lab has shown that in S.cerevisiae persistent double strand breaks (DSBs) are also relocated to the nuclear periphery. Two anchorage sites have been identified: nuclear pores throughout the cell cycle, and Mps3 in S phase in a resection-dependent way. It is remarkable that DSBs and telomeres share common anchorage sites considering that their fate in the cell must be very different: the activation of DNA repair pathways at telomeres could cause telomere-telomere fusions and senescence, while persistent DSBs lead to loss of somatic material and cell death. We propose that the flanking sequence near the DSB influences its binding site at the nuclear envelope, which in turn may determine the outcome of repair. We are testing this hypothesis with a construct made of a DSB site flanked by TG telomeric-like repeats of different lengths, integrated on a chromosome arm. The presence of long TG repeats (TG250) at a break is already known to have an anticheckpoint effect dependent on Rif1 and Rif2. The construct allows us to analyze DSB and telomeric processes at once and hopefully identify whether subnuclear positioning contributes to the identity of telomeres versus DSBs. Presented by: Marcomini, Isabella 148 Poster 77 Bridging ageing and metabolism through RAP1? Paula Martinez, David Pisano, Maria A. Blasco Spanish National Cancer Research Center (CNIO), Madrid, Spain RAP1 is one of the components of the shelterin complex that protects the telomeres. RAP1 binds to both telomeres and non-telomeric sites throughout the whole genome exerting a transcriptional regulatory role. Mice lacking RAP1 gain weight dramatically faster than wild type mice, show symptoms of diabetes type II and hepatic steatosis. Gene expression profile analyses in liver of adult mice show that in the absence of RAP1, several metabolic pathways are remarkably affected. Telomere length is highly dynamic and does vary among different cell types and along ageing. It is therefore conciveable that telomere length would affect the ratio between telomere-bound RAP1 and genomic DNA-bound RAP1 that is available to perform its transcriptional regulatory function. To test this hypothesis we have made used of the telomerase deficient mouse model. Succesive generations of Terc-/- mouse show a progressive telomere shortening that is accompanied by a proportional declive in regenerative capacity of the tissues that leads to an acceletared ageing. We have generated the Rap1-/- Terc-/compound mouse. We are currently analyzing whether Rap1-deletion has an impact on body weight and survival compared to (G1-G3) Rap1+/+ Terc-/-. Telomere length dynamics in these mouse cohorts is also under study to address whether RAP1 could have a role in telomere length homeostasis. Chipseq analysis in Rap1+/+ Terc+/+, G1 Rap1+/+ Terc-/- and G3 Rap1+/+ Terc-/- cells will shed light on whether Rap1 occupancy at telomeric-DNA versus genomic-DNA does change depending on telomere length. In this meeting we will report our results regarding the abovementioned studies. Presented by: Martinez, Paula 149 Poster 78 Telomere shortening induces skeletal and cardiac muscle failure in Duchenne Muscular Dystrophy Foteini Mourkiotia,*, Alex Changb, Alan Meekerc, Helen Blaub a Instructor Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Clinical Sciences Research Center, 269 Campus Drive, Stanford, CA 94305-5175, USA b Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Clinical Sciences Research Center, 269 Campus Drive, Stanford, CA 94305-5175, USA c Department of Pathology, Department of Oncology, Johns Hopkins Medical Institution, Baltimore, MD 21231, USA. Duchenne muscular dystrophy (DMD) is the most common inherited myopathy of childhood that leads to early death, generally before the age of 30. DMD is characterized by severe progressive muscle wasting due to mutations in dystrophin, which links the inner cytoskeleton with the extracellular matrix and therefore plays a key role in plasma membrane integrity in both skeletal and cardiac muscles. Paradoxically, mdx mice, which share the same dystrophin deficiency as DMD patients, exhibit only mild muscle weakness and minimal cardiac dysfunction in contrast to patients. We reasoned that telomere length differences between mice and humans could account for this discrepancy and developed a new mdx mouse model with shortened telomeres (mdx/mTR) (Mourkioti et. al., Nature Cell Biology, 2013 and Sacco, Mourkioti et. al., Cell, 2010). This model is the first dystrophic mouse that exhibits all the pathological hallmarks of human DMD: severe loss of muscle force, kyphosis, muscle membrane disruption, progressive wasting of limb and diaphragm muscles, and dilated cardiomyopathy, culminating in early death. Most importantly, we demonstrated 45% cardiomyocytespecific telomere shortening in both mice and DMD patients. Here, we will present new data on telomere shortening during DMD progression that will provide novel mechanistic insight into the role of telomeres in the etiology of disease. Presented by: Mourkioti, Foteini 150 Poster 79 HOMOLOGY-DEPENDENT REPAIR IS INVOLVED IN 45S rDNA LOSS IN FAS MUTANTS Veronika Muchovaa,*, Simon Amiardb, Iva Mozgováa, Martina Dvořáčkovác, Maria E. Gallegob, Charles Whiteb, Jiří Fajkusa a Mendel Centre for Plant Genomics and Proteomics, CEITEC, and Faculty of Science, Masaryk University, Kamenice 5, CZ-62500 Brno, Czech Republic b Génétique, Reproduction et Développement, UMR CNRS 6293 - Clermont Université - INSERM U1103, Campus des Cézeaux, Université Blaise Pascal, Clermont Ferrand, France c Institute of Biophysics ASCR, Královopolská 135, CZ-61265 Brno, Czech Republic In our previous studies we reported the progressive loss of 45S rDNA sequences and severe telomere shortening in A. thaliana CAF-1-deficient (fas1 or fas2 mutant) plants (Mozgova et al., 2010). Our results further demonstrate that the truncation of both repetitions is mitotic. Since fas mutants were reported to have increased levels of HR, we decided to investigate the possible involvement of this pathway in advertised phenomenon. For this purpose the double mutants in FAS and AtRAD51B (essential for mitotic HDR, paralogue of the Rad51 recombinase) genes were created. Here we demonstrate the significant role of AtRAD51B in 45S rDNA maintenance mechanism in fas mutants while the influence of this knock out in telomeres loss is negligible – suggesting different mechanisms of the loss of the two kinds of repeats at the ends of chromosomes. The involvement of imprecise DNA damage repair in rDNA dynamics in fas mutants is further supported by accumulation of double-strand breaks (measured as γ-H2AX foci) in 45S rDNA which are not associated with rDNA replication and are ATR-dependent. Our data also suggest that the repair of DSB present in the transcribed fraction of rDNA (in the nucleolus) is RAD51B-dependent and contributes to rDNA loss, in CAF-1 (FAS) plant mutants, presumably via single strand annealing pathway. This work was supported by CEITEC (CZ.1.05/1.1.00/02.0068), GACR (P501/11/0289) and by the European Social Fund (CZ.1.07/2.3.00/20.0043). Presented by: Muchova, Veronika 151 Poster 80 The effects of lifestyle factors on leukocyte telomere length dynamics: Results from the ESTHER Cohort Aysel Muezzinler, Aida Karina Dieffenbach, Katja Butterbach, Hermann Brenner Division of Clinical Epidemiology and Ageing Research, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120 Heidelberg, Germany In recent years, the influence of lifestyle factors on leukocyte telomere length (LTL) has drawn considerable attention. However, there is limited and contradictive evidence in the literature. Therefore, we aim to assess the association between BMI, smoking and LTL in both cross-sectional and longitudinal analyses. LTL of 3600 samples of an elderly population were measured by quantitative PCR method. 8-year follow-up LTL of 1000 samples were also measured for the longitudinal analyses. Terminal Restriction Fragment analysis was additionally performed in a sub-sample to obtain absolute TL in base pairs. Multivariate linear regression models were used to estimate the regression coefficients. Age was inversely associated with LTL (r=-0.098, p<0.0001), but not with yearly LTL change (r=0.005, p=0.882). Women had longer LTL than men (p<0.0001). Mean LTL was longer in never smokers (p=0.002), but the rate of yearly LTL change showed no difference. No difference in LTL and yearly LTL change was observed for intensity of smoking, different categories of BMI or duration of obesity. The analysed lifestyle factors point to a very limited role on LTL. Only some significant associations between smoking and BMI-related variables and LTL and yearly LTL change were observed. The power of the longitudinal analysis could be limited due to sample size. LTL dynamics differ in different age groups. Therefore, using a composite index for lifestyle to evaluate the value of LTL as a biological marker of ageing could be a more appropriate approach than focusing on individual factors. Presented by: Muezzinler, Aysel 152 Poster 81 Expression of ATRX represses the Alternative Lengthening of Telomeres phenotype Christine Napier, Jane Noble, Roger Reddel Cancer Research Unit, Children’s Medical Research Institute, Westmead, NSW, Australia Normal mortal fibroblasts contain repressors of the ALT mechanism, as demonstrated by analyses of somatic cell hybrids of ALT cells with normal mortal fibroblasts (1). Previous studies revealed that many ALT-positive tumours and cell lines have mutations in one or other member of the ATRX/DAXX chromatin remodelling complex (2,3). We have shown that knockdown of ATRX was not sufficient to activate ALT activity, but that it significantly decreased the length of time in crisis prior to activation of ALT and immortalisation. We also expressed exogenous ATRX in ALT cell lines that lack ATRX expression. Western blotting showed that the ATRX protein level peaked at day 2 and was undetectable by day 8 post-transfection. The C-circle levels decreased by up to 50% after 2 days of ATRX expression, accompanied by a significant decrease in the number of ALT-associated PML bodies in ATRX-positive nuclei. Cells transfected with ATRX were selected with G418 and resistant clones isolated. More than 80 clones were obtained from two ALT cell lines but only one clone expressed ATRX, suggesting a strong selection pressure against ATRX expression in ALT cells. These data are consistent with ATRX being an ALT repressor. 1. Perrem, K, et al., Oncogene 18: 3383-3390, 1999. 2. Heaphy, CM, et al., Science 333: 425, 2011. 3. Lovejoy, CA, et al., PLoS Genet. 8: e1002772, 2012. Presented by: Napier, Christine 153 Poster 82 In vitro assessment of the protective role of Cdc13 against degradation of telomeric single-stranded 3’ overhangs Saishyam Narayanan, Cecilia Gustafsson, Marita Cohn Department of Biology, Genetics group, Lund University Telomeres are nucleoprotein structures in eukaryotic chromosomes which protect the chromosomal ends from fusion and degradation by exonucleases. The telosome of budding yeast Saccharomyces castellii includes the Cdc13 protein, which binds to specific sequences on the TG-rich single-stranded 3’ overhangs of the telomere. Cdc13 plays a dual role in regulating the telomere lengths by recruiting telomerase by interaction with EST1 and by forming a protective cap (as Cdc13-Stn1-Ten1(CST) complex) at telomeric ends, making it inaccessible to telomerase. Remarkable fluctuations in the levels of Cdc13 along the cell cycle indicate that Cdc13/CST complex may not be essentially forming a protective cap in all the phases of cell cycle. Thus, Cdc13 binding and its functional roles are thought to be tightly regulated. We analyzed the end protection delimitations of Cdc13 using an in vitro Telomere End Protection Assay (TEPA). We found that Cdc13 on its own was able to provide protection to the single-stranded 3’ overhangs against degradation by various exonucleases, when analyzed on a short overhang of 20 nt. The distance of the Cdc13 minimum binding site on the single-strand from the ds-ss junction influenced the protection. Interestingly, Cdc13 bound at the minimum binding site conferred protection to the nucleotides beyond the minimum binding site in the 3’ overhang. Our studies proved that TEPA could be used as an effective method to determine the protective ability of Cdc13 at telomeres with different 3’ overhang lengths and 5’ end permutations. Presented by: Narayanan, Saishyam 154 Poster 83 TRF2 Overexpression Induces Telomeric Ultrafine Anaphase Bridges Bernadette Nera, Hui-Shun Huang, Thao Lai, Lifeng Xu University of California, Davis, Microbiology and Molecular Genetics, Davis, CA, 95616 Overexpression of full-length TRF2 is known to cause rapid telomere shortening. Here we report that TRF2 overexpression inhibits telomere replication and induces the formation of telomeric ultrafine anaphase bridges whose resolution leads to drastic telomere shortening. Ultra-fine anaphase bridges (UFBs) derive from sister chromatids that are held together at anaphase due to DNA catenation or incomplete replication. The BLM helicase protein is reported to align along UFBs. We found that elevated levels of TRF2 caused replication fork stalling at telomeres and induced telomeric UFBs. Depletion of BLM exacerbates TRF2-induced telomeric UFB formation. In contrast to the canonical UFBs, only a proportion of such telomeric UFBs are aligned by the BLM protein, suggesting that they represent a novel class of UFBs. The frequency of TRF2-induced telomeric UFBs correlates with the average telomere length of cells. Comparable levels of TRF2 expression can induce significant amount of telomeric UFBs in cells with pre-extended telomeres, but not in cells with very short average telomere lengths. We also found that the formation of telomeric UFBs correlate with the TRF2-induced rapid telomere shortening phenotype. Presented by: Nera, Bernadette 155 Poster 84 How specific is TRF2 binding to telomeric DNA? Ivona Nečasová, Michal Zimmermann, Ctirad Hofr Chromatin Molecular Complexes, CEITEC and Faculty of Science, Masaryk University, Brno, CZ-62500, Czech Republic Telomeres form caps at the very ends of linear chromosomes. Any disturbance in integrity of telomeres causes chromosomal instabilities leading to unfavorable chromosomal rearrangements or even to carcinogenesis. In mammals, telomere-associated proteins form a shelterin complex which protects chromosome ends from DNA repair machinery. Shelterin serves as a negative regulator of telomerase, a ribonucleoprotein enzyme that adds telomeric DNA to chromosomal ends to ensure complete genome replication. TRF2 plays a central role within shelterin. TRF2 binds to double-stranded telomeric DNA via a Cterminal Myb domain. It has been proposed that a N-terminal basic domain of TRF2 might be involved in DNA binding as well. Our results obtained by fluorescence anisotropy and isothermal titration calorimetry quantify the contribution of the basic domain to the overall TRF2 binding to telomeric DNA. Additionally, our results suggest the importance of the basic domain for long-range electrostatic interactions between TRF2 and DNA. Furthermore, NMR measurements indicate structural changes associated with the binding of TRF2 basic domain to DNA. Our studies provided the first detailed quantitative and thermodynamic description of the contribution of the basic domain to the DNA affinity of human TRF2. The obtained findings contribute to address interactions of shelterin proteins that are essential for the regulation of telomere length and thus maintenance of the whole genome stability. This work was supported by CEITEC (CZ.1.05/1.1.00/02.0068), the European Social Fund (CZ.1.07/2.3.00/30.0019), GACR (P205/12/0550), and KONTAKT II (LH13054). Presented by: Nečasová, Ivona 156 Poster 85 Selective Bioluminogenic HDAC Activity Assays for Profiling HDAC Inhibitors Andrew L. Niles1, Nathan J. Evans1, Kevin R. Kupcho1, Thomas A. Kirkland2, and Dan F. Lazar1 1 Promega Corporation, 2800 Woods Hollow Road, Madison, WI 53711 Promega Biosciences LLC, 277 Granada Drive, San Luis Obispo, CA 93401 2 Histone deacetylases (HDACs) play critical roles in the regulation of gene transcription and cell signaling events by deacetylating histones and other important non-histone substrates. Aberrant increases in HDAC enzyme activities are therefore implicated in a number of human infirmities, including cancers, metabolic disease and neurodegeneration. Fortunately, HDAC enzymes represent attractive pharmacological targets because they are readily tractable with small molecule inhibitors. In fact, several HDAC inhibitors (HDACi) have recently proceeded through (or are near) the FDA approval process for the treatment of hematologic malignancies. However, the promise of clinical HDACi therapy has been hampered by significant dose-limiting toxicities. These off-target effects have led to a renewed focus on basic HDAC biology and the development of isoenzyme-specific HDAC inhibitors which could avoid off-target effects. To help facilitate the discovery of compounds with better defined selectivity profiles, we have developed lysine deacetylase assays that selectively measure specific isoenzyme activities in cells, extracts, or purified recombinant preparations. These assays are based on substrates that are selective due to a combination of extended peptide sequence and novel chemical modifications. Deacetylase activity is measured by delivering a single, pro-luminogenic, homogeneous assay reagent to assay wells, resulting in luminescence proportional to HDAC activity. In addition to being isoenzyme selective, these novel substrates are cell permeable allowing for lytic and non-lytic cell-based HDAC assays. Lastly, these assays are also fully compatible with fluorescent viability and/or cytotoxicity assays. This provides additional flexibility for multiplexed formats which examine not only selective HDAC inhibition, but the functional consequences they exert on cell health. Presented by: Niles, Andrew L. 157 Poster 86 IDENTIFICATION OF NEW TERT GENE MUTATIONS IN FAMILIAL STUDIES OF PATIENTS PRESENTING WITH APLASTIC ANEMIA/HYPOCELLULAR MYELODYSPLASTIC SYNDROME Valeria Nofrinia,*, Valentina Pierinia, Francesco Berardinellib, Caterina Matteuccia, Valeria Di Battistaa, Antonella Sgurab, Tamara Iannottia, Alessandro Brozzic, Roberta La Starzaa, Cristina Mecuccia a Haematology and Bone Marrow Transplantation Unit, University of Perugia, Perugia, Italy Department of Science, University ‘Roma Tre’, Rome, Italy c Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Perugia, Italy b We describe new familial TERT gene mutations in 3 patients with diagnosis of severe aplastic anemia (AA) or hypocellular Myelodysplastic Syndrome (h-MDS). Pedigrees were obtained in all cases. A family history of telomere syndrome was documented in 2 cases. Telomere length was measured by Q-FISH comparing each proband with at least 3 age- and sex-matched healthy controls. First patient (female, 53) had severe AA, idiopathic pulmonary fibrosis, liver cirrhosis and died 41 months after diagnosis. She bore a missense mutation (c. 2093 G>A p. R698Q at exon 5) defined as damaging by in silico analysis (SIFT and PolyPhen-2 databases). Her brother (42 years) bore the same mutation but clinical effects were significantly different since he presented slight macrocytic anemia and mild pulmonary fibrosis. Q-FISH revealed, in the brother, significant telomere shortening (mean T/C%: 10.9 vs 17.4, p<0.05). Second patient (male, 18 years) presented severe AA and bore a new missense mutation (c. 2020 G>A p. G674S) at exon 5 defined as benign (SIFT and PolyPhen-2). Q-FISH revealed telomere shortening (mean T/C%: 18.6 vs 24.6, p<0.05). The patient’s mother, 1 sister and 1 brother had the same exon 5 mutation without phenotype. In these cases telomere length studies are ongoing. Third patient (female, 53 years) with family background had a stable (+136 months) h-MDS and an acquired 47,XX,+8 karyotype. Phenotype included idiopathic pulmonary fibrosis, skin hyperpigmentation, osteoarthritis. As compound heterozygous, she bore two germline mutations (nonsense mutation c.1209C>A p.C403*, exon 2; missense mutation c.2455C>T p.R819C, exon 8). Telomere length: mean T/C% 9.3 vs. 18.1 (p<0.05). Her healthy 23 year old son bore the exon 2 mutation and presented telomere shortening (mean T/C% 12.3 vs. 24.1, p<0.05). All new mutations had a striking different penetrance. Presented by: Nofrini, Valeria 158 Poster 87 Androgenetic fish as models in studies concerning healing of chromosome breaks by de novo telomere addition Konrad Ocalewicz Faculty of Environmental Sciences, University of Warmia and Mazury in Olsztyn, Poland Data concerning telomere healing of the chromosome breaks usually derive from the studies using cell lines exposed to the clastogenic agents including ionizing radiation that damage chromosomal DNA by inducing double strand breaks (DSBs). Ionizing radiation is also applied to inactivate nuclear DNA in fish eggs during induced artificial androgenesis in order to produce individuals possessing exclusively paternal chromosomes. Although X and gamma rays are thought to eliminate entire maternal nuclear DNA from the subsequent zygotes, we have found many radiation-induced fragments of the maternal chromosomes in the androgenetic rainbow trout and brook trout embryonic and adult cells. Distribution patterns of the telomeric hybridization signals on such chromosome fragments enabled to discriminate their several groups: 1) linear chromosome fragments with telomeric DNA sequences at both ends, 2) ring chromosomes with fused broken chromosome arms, 3) fragments with intact telomeres from one arm and fused unprotected sister chromatids of another broken arm and 4) acentric telomeric fragments incorporated into paternal chromosomes. Our observations proved, the some of the broken ends of the fish chromosomes could have been repaired with the telomeric DNA repeats synthesized de novo by telomerase or another mechanism capable of de novo telomere addition. Easy access to many fish species and availability of the techniques enabling study of the fish telomeres and telomerase make these organisms to be promising models in researches concerning healing of the broken chromosomal ends in the embryonic and adult cells. Presented by: Ocalewicz, Konrad 159 Poster 88 A new role for histone deacetylase 5 in the maintenance of long telomeres Catherine Polese1,3, Clara Lopes Novo1,2, Nicolas Matheus*, Anabelle Decottignies†, Arturo Londono-Vallejo‡,§, Vincent Castronovo*, and Denis Mottet3 *University of Liege Sart-Tilman, GIGA-Cancer, Metastasis Research Laboratory, Pathology Institute B23, Liege, Belgium †Genetic and Epigenetic Alterations of the Genome, de Duve Institute, Faculty of Pharmacy and Biomedical Sciences, Catholic University of Louvain, Brussels, Belgium ‡Telomeres and Cancer Laboratory, Labellis Ligue Contre le Cancer Institut Curie, Paris, France §Université Pierre et Marie Curie (UPMC), University of Paris 06, Paris, France 1These authors contributed equally to this work 2Current Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK 3Laboratory of Protein Signaling and Interactions, GIGA-Research, B34, Liege, Belgium Telomeres are major regulators of genome stability and cell proliferation. A detailed understanding of the mechanisms involved in their maintenance is of foremost importance. Of those, telomere chromatin remodeling is probably the least studied; thus, we intended to explore the role of a specific histone deacetylase on telomere maintenance. We uncovered a new role for histone deacetylase 5 (HDAC5) in telomere biology. We report that HDAC5 is recruited to the long telomeres of osteosarcoma- and fibrosarcoma-derived cell lines, where it ensures proper maintenance of these repetitive regions. Indeed, depletion of HDAC5 by RNAi resulted in the shortening of longer telomeres and homogenization of telomere length in cells that use either telomerase or an alternative mechanism of telomere maintenance. Furthermore, we present evidence for the activation of telomere recombination on depletion of HDAC5 in fibrosarcoma telomerase-positive cancer cells. Of potential importance, we also found that depletion of HDAC5 sensitizes cancer cells with long telomeres to chemotherapeutic drugs. Cells with shorter telomeres were used to control the specificity of HDAC5 role in the maintenance of long telomeres. HDAC5 is essential for the length maintenance of long telomeres and its depletion is required for sensitization of cancer cells with long telomeres to chemotherapy. Presented by: Polese, Catherine 160 Poster 89 Unusual telomeres in plants Vratislav Peska, Jiri Fajkus CEITEC MU Telomeres are physical ends of eukaryotic chromosomes that (i) compensate the loss of DNA due to incomplete replication, (ii) differ chromosomal ends from dsDNA breaks, (iii) prevent inappropriate recombination and end-fusions. These features are conditioned by the presence of telomere proteins interacting in a complex called shelterin. Telomerase - reverse transcriptase - plays major role in telomere length maintenance. Thanks to telomerase activity, telomeres consist of minisatelites with repetitive units type 5'-(TxAyGz)n-3 '. But more and more exceptions has been found with altered or unknown terminal DNA sequence, where telomerase activity is replaced by mechanism of alternative lengthening of telomeres (ALT) (e.g., Diptera, some laboratory yeast, some tumors and plants). Dicotyledonous plants of the Solanaceae family (Vestia, Sessea and Cestrum) and the monocotyledonous of the family Alliaceae (e.g. A. cepa and A. sativum) are an excellent opportunity to study how the eukaryotic cell cope with loss of telomerase system without loss of integrity and evolutionary perspective. Alternative telomeric sequence in plants remains unknown despite its importance. We have sequenced two Allium species to compare their repetitive DNA and to obtain telomeric candidates. This work was supported by CEITEC (CZ.1.05/1.1.00/02.0068) and by the European Social Fund (CZ.1.07/2.3.00/30.0019). Presented by: Peska, Vratislav 161 Poster 90 A study of telomere-related biomarkers in an arsenic-exposed Bangladeshi population Brandon Pierce, Shantanu Roy, Farzana Jasmine, Muhammad Kibriya, Habibul Ahsan University of Chicago Chronic exposure to inorganic arsenic through contaminated drinking water affects >100 million people worldwide and can lead to increased risk for various cancers, including lung, bladder, liver, skin, and kidney. The mechanisms by which arsenic contributes to carcinogenesis are not entirely understood, but telomere dysfunction has been hypothesized as one potential carcinogenic mechanism. Supporting this notion, recent experimental and observational research suggests that arsenic exposure may influence telomere length and telomere maintenance. Our group is currently conducting a large population-based study of arsenic-exposed individuals in Bangladeshi in order to characterize the relationships among arsenic exposure (measured in water and urine), various telomere-related biomarkers, and arsenic-related health outcomes. We are using a case-cohort design to assess the association of telomere length (measured prospectively in baseline blood samples) with incident cases of arsenic-induced skin lesions (n=1000), non-melanoma skin cancer (n=250), and overall mortality (n=500), as compared to control cohort members. Preliminary data emerging from this work suggests that arsenic exposure is associated with longer telomeres, consistent with a recent publication on this topic. Analysis of gene expression data for telomere-related genes suggests that telomere lengthening may occur through the “alternative lengthening of telomeres” (ALT) pathway, although lengthening via telomerase is also a possibility. As a part of this study, we also plan to measure several reported biomarkers of telomere dysfunction (CRMAP and chitinase) and assess associations with arsenic exposure and arsenic-related health outcomes. This ongoing study is a critical step towards understanding the mechanisms of arsenic toxicity in humans. Presented by: Pierce, Brandon 162 Poster 91 Long-term particulate air pollution in association with the mitochondrial-telomere axis of ageing Nicky Pietersa,*, Bram G. Janssena, Karen Smeetsa, Harrie Dewitteb, Bianca Coxa, Michelle Plusquina, Ann Cuypersa, Tim S. Nawrota a b Hasselt University Primary Health Care Center Genk Background: Telomere length and mitochondrial DNA content are markers of ageing and ageingassociated diseases including cardiovascular morbidity. Airborne particulate matter (PM) is associated with adverse effects on the cardiovascular system, but its effects on markers of ageing is currently inconclusive. Sirtuin1 down-regulation has been associated with the development of age-related diseases and might be an underlying mechanism in the etiology of PM-induced health effects. Aim: The present study examines the association between long-term PM exposure and leukocyte telomere length and mitochondrial DNA content in elderly. In addition, we examined the intermediateterm effects of Sirtuin1 on this association. Methods: Among 182 non-smoking elderly, leukocyte telomere length (LTL) was measured using a quantitative real-time polymerase chain assay (qPCR). Mitochondrial DNA content and gene expression of Sirtuin1 were measured using qPCR. Annual PM2.5 exposure was calculated for each participant’s home address using a Kriging interpolation model. Results: Annual PM2.5 concentrations ranged from 15 to 23 ug/m3. Independent of gender, age, BMI, socio-economic status and statin use, an annual 5 µg/m3 increase in PM2.5 was associated with a relative decrease of 23% (95% CI: -43% to -3%) in LTL and a relative decrease of 31% (95% CI: -50% to 12%) in mitochondrial DNA content. Mediation analysis showed Sirtuin1 as an intermediate mechanism for PM-related effects on markers of ageing. Conclusion: We found significant effects of PM2.5 exposure on the telomere-mitochondrial axis of ageing, mediated through a decreased expression of Sirtuin1. The studied targets may have important roles in the chronic health effects of PM2.5. Presented by: Pieters, Nicky 163 Poster 92 Mitochondria dysfunction in telomerase zebrafish mutants Inês Pimenta de Castro, Madalena Carneiro, Miguel Godinho Ferreira Instituto Gulbenkian de Ciência, Oeiras, Portugal Different biological processes cooperate progressively to diminish organ function with age. There are two main molecular theories of ageing: one proposes that short telomeres trigger cell senescence leading to tissue functional decline; the second postulates that cellular ageing results from the progressive loss of mitochondrial metabolism. Zebrafish have restricted telomerase expression and human-size telomeres. We recently showed that first generation telomerase mutant zebrafish die prematurely with shorter telomeres and displaying several ageing phenotypes. We now report that telomerase loss leads to intrinsic mitochondrial abnormalities in proliferative tissues such as the head kidney and gut. These include increased aberrant mitochondria that are not capable of respiring, producing ATP or generating ROS. Additionally, we observe a significant activation of the Akt pathway that results in the inactivation of the FOXO family of transcription factors and subsequent autophagy impairment. These data support a model in which telomere shortening leads to mitochondrial dysfunction through the activation of the Akt/FOXO pathway, thus orchestrating old age phenotypes. We anticipate that modulation of this pathway may attenuate the consequences of telomere shortening that naturally occurs with age. Presented by: Pimenta de Castro, Inês 164 Poster 93 Preclinical assessment of the telomerase inhibitor Imetelstat combined with radiotherapy in an orthotopic model of human glioblastoma Sylvain Ferrandona, Céline Mallevalb, Priscillia Battiston-Montagnea, Badia El Hamdania, Radu Bolbosc, Patrick Manasd, Sergei M Gryaznove, Claire Rodriguez-Lafrassea, Jérôme Honnoratb, Delphine Ponceta,*, et al. a EMR3738, Cellular and Molecular Radiobiology Laboratory, South Lyon Charles Mérieux Medicine Faculty, F-69921 Oullins, France b Team « neuro-oncology and neuro-inflammation », Lyon Neuroscience Research Center, INSERM U1028/CNRS UMR 5292, Laennec Medicine Faculty, 69372 LYON cedex 08, France c CERMEP-imagerie du vivant, Groupement Hospitalier Est, 59 Bd Pinel, 69677 Bron, France d UMS 3444 gerland CNRS P.B.E.S, Ecole Normale Supérieure Lyon, BP 7000, 69342 Lyon Cedex 07, France e Geron Corporation, 149 Commonwealth, Menlo Park, CA 94025, USA Glioblastoma (GBM) is the most frequent and aggressive adult brain tumor. Development of therapies in GBM faces two obstacles: a high tumoral molecular heterogeneity and the blood brain barrier (BBB) passage. The presence of a telomerase activity (TA) in most GBM and its level are predictive of a poor prognosis in GBM. Inhibiting the TA is thus a promising option to treat GBM. TA inhibition also increases the response to radiotherapy (RT) in other tumors. We thus tested an oligonucleotide targeting the telomerase (Imetelstat) alone and combined with RT to treat GBM. In vitro, after a shortterm pre-treatment by Imetelstat of the U87MG cell line, we observed an increase in telomeric damage and in cell death, both potentialized by RT. In vivo, using a murine orthotopic model of human GBM, µMRI imaging and a protocol easily transferable into clinical practice, we demonstrated that Imetelstat significantly : (i) inhibits the TA in the center of the tumor, (ii) reduces tumor volume; (iii) increases tumor response to RT, in terms of tumor volume regression and survival increase. Imetelstat has proven is efficiency in clinical trials in numerous cancers. These data support its clinical evaluation combined with RT in human GBM. Presented by: Poncet, Delphine 165 Poster 94 Human RTEL1 is required for the maintenance of long telomeres Rosa Maria Porreca, Christian Naucke, Mike Schertzer, Mylène Perderiset, Irena Draskovic, Arturo Londoño-Vallejo Telomeres & Cancer laboratory, UMR3244- UPMC, Institut Curie, 26 rue d’Ulm, 75005 Paris Rtel1, regulator of telomere elongation helicase 1, was initially discovered to be an essential factor for telomere length maintenance in mice. Further experiments showed that mouse cells lacking Rtel1 display telomeric loss and chromosome aberrations, caused by abnormal replication and inappropriate resolution of T-loops. It has been also suggested that Rtel1 is implicated in genome replication, recombination and DNA repair. Interestingly, recent germline mutations in RTEL1 have been found in patients with Hoyeraal-Hreidarsson syndrome (HHS), a severe form of dyskeratosis congenital. However, the precise mechanism of action of this protein in human cells remains largely unknown. To investigate the role of RTEL1 in human telomere metabolism we perform loss of function experiments with specific siRNAs and we make use of quantitative-FISH to measure telomere length after depletion of RTEL1 in a panel of cell lines. We show that down-regulation of RTEL1 induces telomere shortening exclusively in cells with very long telomeres (HT1080-ST and 1301) whereas the telomere length distributions in cells with short telomeres (HeLa and HT1080) remain unchanged. However, we find that chromosome aberrations were present regardless of the telomere length. Specifically, we observe chromatid/chromosome breaks, fusions and triradial or quadriradial structures, with activation of the DNA damage response through ATM-CHK2 and γH2AX signaling. Our recent experiments further demonstrate that upon depletion of RTEL1 there is a different stoichiometry of sheltering proteins at telomeres. Based on these results we are currently testing the hypothesis as to whether RTEL1 might be required for the overhang stability. Presented by: Porreca, Rosa Maria 166 Poster 95 Telomere Repeat Binding (TRB) proteins serve as functional components of telomeres and interact with telomerase (TERT) Petra Prochazkova Schrumpfovaa,*, Ivona Vychodilovaa, Martina Dvorackovab, Jana Majerskac, Ladislav Dokladalb, Sarka Schorovaa, Jiri Fajkusa a Mendel Centre for Plant Genomics and Proteomics, CEITEC and Department of Functional Genomics and Proteomics, NCBR, Faculty of Science, Masaryk University, Kamenice 5, CZ-625 00 Brno, Czech Republic b Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Královopolská 135, CZ61265 Brno, Czech Republic c Current address: Swiss Institute for Experimental Cancer Research (ISREC), Ecole Polytechnique Fédérale de Lausanne, Station 19, 1015 Lausanne, Switzerland Although telomeric proteins across the species share many similarities, in vivo data indicating the existence of a structure similar to mammalian shelterin complex in plants are still very poor. Partial characterization of a number of candidate proteins has not identified true components of plant shelterin or elucidated their functional mechanisms. Telomere repeat binding (TRB) proteins from Arabidopsis thaliana bind plant telomeric repeats through a Myb domain of the telobox type in vitro, and have been shown to interact with POT1b (Protection of telomeres 1). We discover co-localization of TRB1 protein with telomeres in situ using fluorescence microscopy, as well as in vivo interaction using chromatin immunoprecipitation. Classification of the TRB1 protein as a component of plant telomeres is further confirmed by the observation of shortening of telomeres in knockout mutants of the trb1 gene. Moreover, TRB proteins physically interact with plant telomerase catalytic (TERT) subunits. These findings suggest that TRB proteins are part of the telomeric interactome of A. thaliana. The research was supported by the Czech Science Foundation (13-06943S), by project CEITEC (CZ.1.05/1.1.00/02.0068) of the European Regional Development Fund, and project (CZ.1.07/2.3.00/20.0043) co-financed from European Social Fund. Procházková Schrumpfová P., Vychodilová I., Dvořáčková M., Majerská J., Dokládal L., Schořová Š. and Fajkus J. (2014) Telomere Repeat Binding proteins are functional components of Arabidopsis telomeres and interact with telomerase. The Plant Journal (2014) 77, 770–781 Presented by: Prochazkova Schrumpfova, Petra 167 Poster 96 In vivo selection for new telomerase RNA template regions in S. pombe Margaret Pruitt1,2, Richard Dannebaum1, Peter Baumann1,2 1 HHMI and Stowers Institute for Medical Research 2 Dept. of Mol. and Int. Physiology, KUMC The most common telomeric repeat sequence is GGTTAG, present in all vertebrates, and some invertebrates, fungi, and plant species. This predominance suggests that response to selective pressures drove many systems to this sequence to protect chromosome ends and solve the end replication problem. Yet, despite the apparent success of this repeat, different sequence repeats are found in diverse genera of algae, fungi, and ciliates. Perhaps the most substantial repeat diversity is found among fungi ranging from perfect 25 nucleotide repeats in Kluyveromyces lactis to highly heterogeneous repeats in Saccharomyces cerevisiae and Schizosaccharomyces pombe. Whereas perfect repeat sequences result from faithful reverse transcription of the RNA template, the synthesis of heterogeneous repeats requires stuttering, slippage or poorly defined boundary elements in the telomerase RNA subunit. Telomeric DNA sequence diversity is limited by the need for telomerase to reverse transcribe the sequence in a processive manner, the binding specificity of proteins that bind single- and double- stranded parts of the telomere, and possibly the requirement to form Gquadruplexes. The diversity of telomeric sequences found among fungi is surprising in light of these multiple constraints that favor a unique and constant repeat sequence. We aim to characterize the level of diversity of telomeric sequences that can exist in S. pombe. We replaced the TER1 template sequence with all 16,384 possible nucleotide combinations and allowed cells containing this library to grow competitively in liquid culture. Using Illumina sequencing technology, we followed dynamic changes in the abundance of different templates. Additionally, we are monitoring the length and sequence of telomeric DNA, the ability of Pot1 and Taz1 to bind and the extent to which alternative repeats can maintain the full spectrum of telomeric functions. This study will inform our understanding of the process and functional significance of the co-evolution of the Grich telomere and sequence-specific, telomere-associated proteins. Presented by: Pruitt, Margaret 168 Poster 97 Effects of dyskeratosis congenita patient mutations in hTERT on telomerase dimerization and activity Alix Christen, Sophie Redon, Sascha Feuerhahn, Joachim Lingner EPFL-ISREC Biochemical experiments and structural analysis by electron microscopy indicated that human telomerase forms dimers and that it contains two active sites (Sauerwald et al. 2013). Heterozygous mutations in hTERT cause dyskeratosis congenita. Whether hTERT mutations cause disease because of haploinsufficiency or whether hTERT mutations may have dominant negative effects has remained elusive. We now tested whether disease mutations in hTERT impinge on the ability of telomerase to multimerize and determined whether disease mutant hTERT interferes with wild type hTERT to catalyze telomere extension when co-assembled into heterodimers. We demonstrate that the sedimentation coefficient of endogenous telomerase in HEK293T cells is the same as that of “supertelomerase” that was assembled upon co-overexpression of hTERT and hTR in these cells. By performing co-immunoprecipitation assays with active sucrose gradient fractions we confirm that active telomerase is dimeric. We demonstrate that the tested dyskeratosis congenita hTERT mutations do not impair with the ability of telomerase to multimerize. We purify wild type/disease mutant telomerase heterodimers and characterize telomerase activity and processivity. Our data indicate that wild type/mutant telomerase heterodimers have a strongly reduced catalytic activity in comparison to wild type hTERT homodimers. Thus, hTERT mutations found in dyskeratosis congenita are dominant negative in heteromeric complexes interfering with wild type hTERT activity. Sauerwald, A., Sandin, S., Cristofari, G., Scheres, S.H., Lingner, J., and Rhodes, D. (2013). Structure of active dimeric human telomerase. Nature structural & molecular biology 20, 454-460. Presented by: Redon, Sophie 169 Poster 98 Quantile regression for flow-FISH telomere length measurement as a diagnostic tool for telomere diseases Fernanda Gutierrez Rodrigues, Priscila Santos Scheucher, Edson Zangiacomi Martinez, Bárbara Amélia Aparecida Santana, Rodrigo Tocantins Calado Ribeirão Preto Medical School, University of São Paulo Telomere diseases are a variety of disorders that may clinically manifest as bone marrow failure (BMF), dyskeratosis congenita, hepatic cirrhosis, and idiopathic pulmonary fibrosis (IPF). Telomere length measurement has important implications for diagnosis and therapy in telomere diseases. Methods capable of accurately measuring telomere length contribute to the improvement of diagnostic services. The goals of this study were evaluate the flow-FISH performance for telomere length measurement and assess the differential diagnosis contribution of this technique in patients with BMF syndromes and IPF. For this purpose, telomere length was assessed in peripheral blood leukocytes from a cohort of 180 healthy Brazilian subjects ranging in age from zero to 92 years. The data obtained fitted a thirdorder polynomial curve (R2 = 0.6099) and showed that telomeres decline with aging (rate of loss = 47 bp/year). The 1st, 10th, 50th, 90th, and 99th percentiles of healthy controls were adjusted to the curve applying quantile regression. Telomere length was also evaluated in 37 BMF patients and 17 IPF patients. Thirty-seven percent of individuals with BMF, 7% of sporadic IPF and 66% of familial IPF patients had short telomeres (<10th percentile of controls). The Bland-Altman agreement analysis between these methods was high. The mean difference was - 0,172 and the limits of agreement varied from -2,207 - 1,862. Therefore, for the first time, we were able to measure telomere length of Brazilians healthy controls and patients with BMF and IPF by flow-FISH. Our results reveal that applying quantile regression analysis for flow-FISH telomere length measurement is an accurate technique for the diagnosis of telomere diseases. Presented by: Rodrigues, Fernanda Gutierrez 170 Poster 99 MTV, a telomeric ssDNA-binding complex, protects Drosophila telomeres and recruits retrotransposon to chromosome ends yi zhanga, liang zhanga, guanjun gaob, yikang ronga,* a b lbmb, nci, nih tsinghua university, china Telomeres are protected by multiple protein complexes. In telomerase-maintained organisms, the Shelterin complex occupies primarily the duplex region of the telomere while the CST complex binds the single stranded region. Drosophila uses a transposon-based mechanism for end protection. We showed that the HipHop-HOAP complex occupies the duplex region. The revelation by the Gatti group that the Ver protein shares significant sequence similarities with Stn1 suggests that a similar ssDNAbinding complex exists in Drosophila. Using yeast 2-hybrid and recombinant proteins, we identified a trimeric complex consisting of Ver and Moi, two known capping proteins, as well as TEA, a novel capping protein that we identified. The Moi-TEA-Ver (MTV) complex purified in vitro binds and protects ssDNA in a sequence-independent manner. In vivo, MTV components are specifically enriched at chromosome ends where they prevent telomere fusion. Their telomeric localization is interdependent, and point mutations disrupting MTV interaction in yeast 2-hybrid assays behave as nulls in Drosophila, consistent with these proteins functioning as a complex in vivo. Interestingly, MTV as a complex may regulate end-elongation by participating in the recruitment of telomeric transposon RNPs to chromosome ends. MTV thus shares functional similarities with both the CST and the Tpp1-Pot1 complex in telomerase-maintained organisms, highlighting a possible conserved feature between telomerase-based and transposon-based telomere protecting mechanisms. Presented by: rong, yikang 171 Poster 100 Telomere length dynamics and hematopoietic differentiation in human DKC1-mutant iPSCs Flavia Sacilotto Donairesa,*, Thomas Winklerb, Cynthia Dunbarb, Rodrigo Tocantins Caladoa a Department of Genetics, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil. CTC – Center for Cell-based Therapy, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil. b Hematology Branch, National Heart Lung and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA. X-linked DC is caused by mutations in dyskerin (encoded by DKC1 gene), a protein that provides stability to the telomerase complex. Induced pluripotent stem cells (iPSCs) have been established as patient-specific disease models, as patients’ somatic cells can be reprogrammed to a pluripotent state, maintaining genotypic characteristics. Moreover, the pluripotent capacity of iPSCs enables differentiation into virtually any cell type. In an attempt to elucidate telomere dynamics in DC, we derived iPSCs from dermal fibroblasts of a DC patient carrying a DKC1 mutation (A353V substitution) by forced expression of OCT4, SOX2, KLF4, and MYC using an excisable polycistronic lentiviral vector (STEMCCA). We analyzed telomere length of three independent iPSC clones up to 40 passages, and observed telomere shortening during the first 20 passages, followed by length stabilization at a shorter state than in parental fibroblasts. We differentiated the three DKC1-mutant iPSCs clones towards the hematopoietic lineage, and observed a marked defect in generation of CD34+/CD45+ hematopoietic lineage cells. One clone had a shift towards hemogenic endothelial precursors (CD34+/CD31+). Our findings suggest that the control of telomere length in DKC1-mutant iPSCs is complex, with shortening as expected in early passages post reprogramming, followed by stabilization over time, suggesting an alternative pathway to maintain telomere length, and in contrast to findings reported in some previous studies. Telomere length was not completely recovered after reprogramming, suggesting that full dyskerin function is necessary for actual telomere elongation during reprogramming. Finally, our results also suggest that normal dyskerin function is necessary for adequate HSC differentiation. Presented by: Sacilotto Donaires, Flavia 172 Poster 101 Induced pluripotent stem cells as a model for telomeric abnormalities in ICF type I syndrome Shira Sagiea,*, Erika Ellrana, Hagar Katzira, Rony Shakeda, Alaa Ghanayimb, Maty Tzukermana, Sara Seliga a Laboratory of Molecular Medicine, Rambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion, Haifa 31096, Israel b Computer Science Department, Technion, Haifa 32000, Israel Human telomeric regions are packaged as constitutive heterochromatin, characterized by extensive subtelomeric DNA methylation and specific histone modifications. ICF (Immunodeficiency, Centromeric instability, Facial anomalies) type I patients carry mutations in DNA methyltransferase 3B (DNMT3B) that methylates de novo repetitive sequences during early embryonic development. ICF type I patientfibroblasts display hypomethylated subtelomeres, abnormally short telomeres and premature senescence. In order to study the molecular mechanism by which the failure to de novo methylate subtelomeres results in accelerated telomere shortening, we generated induced pluripotent stem cells (iPSCs) from three ICF type I patients. Telomeres were elongated in ICF-iPSCs during reprogramming, and the senescence phenotype was abolished despite sustained subtelomeric-hypomethylation and high TERRA levels. Fibroblast-like cells (FLs) isolated from differentiated ICF-iPSCs maintained abnormally high TERRA levels, and telomeres in these cells shortened at an accelerated rate, leading to early senescence, thus recapitulating the telomeric phenotype of the parental fibroblasts. These findings demonstrate that the abnormal telomere phenotype associated with subtelomeric hypomethylation is overridden in cells expressing telomerase, therefore excluding telomerase inhibition by TERRA as a central mechanism responsible for telomere shortening in ICF syndrome. The data in this study lend support to the use of ICF-iPSCs for modeling of phenotypic and molecular defects in ICF syndrome and for unraveling the mechanism whereby subtelomeric hypomethylation is linked to accelerated telomeric loss in this syndrome. Presented by: Sagie, Shira 173 Poster 102 Studies of G quadruplex-hRPA interactions in the telomere maintenance context Layal Safaa, Jean-François Rioub, Carole Saintoméb,* a Structure et instabilit des g nomes, INSERM U565, CNRS UMR 7196, Mus um National d’Histoire Naturelle, 43 rue cuvier, 75005, Paris, France b UMR 7196, Mus um National d’Histoire Naturelle, 43 rue cuvier, 75005, Paris, France Telomeres are the terminal protein-DNA complexes of linear eukaryotic chromosomes. They play an important role in critical processes underlying genome stability, cancer, and aging. Human telomeric DNA consists of a duplex region composed of TTAGGG repeats, which terminates in a 100–200 nucleotide G-rich single-stranded overhang (G-overhang) able to adopt secondary structures such as tloop and G-quadruplex. The G-overhang could be elongated in cancer cells through a reverse transcriptase named telomerase or by alternative lengthening of telomeres. Human Replication Protein A (hRPA) is able to bind and unfold G-quadruplexes in vitro. A binding model has been proposed suggesting the requirement of a single-stranded region created at one extremity of the G-quadruplex and due to the equilibrium between different conformations. Furthermore hRPA can modulate telomerase activity. In order to validate the previously described G-quadruplex-hRPA binding model, we have analyzed the interactions of hRPA with different sequences able to form G-quadruplex, using specific small molecule ligands (G4 ligands) that bind and stabilize the G-quadruplex conformation. We have also realized photocrosslinking experiments using the 4-thiothymine as probe to determine the nature of subunits implicated in G-quadruplex hRPA interactions. In this study, we showed that G4 ligands were able to efficiently inhibit hRPA binding to telomeric Gquadruplex. However, the addition of polyT tails with different length at either the 3’ or the 5’ extremity of the G-quadruplex sequence promotes hRPA binding. The presence of G4 ligands on these extended G-quadruplex coupled with photocrosslinking experiments permitted to reveal the binding and opening polarity of RPA. Presented by: Saintomé, Carole 174 Poster 103 The inhibition of protein-protein interactions within telomeres: a route to cancer chemotherapy? Twana Salih, Weng Chan, Lodewijk Dekker, Charles Laughton Division of Medicinal Chemistry and Structural Biology, School of Pharmacy and Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD Stable telomeres are critical to the survival of cancer cells; therefore a number of different cancer chemotherapeutic pipelines have been developed in order to disrupt or destabilize telomeres or telomerase. One of the newest approaches is the disruption of key protein-protein interactions in the telomere, such as that between shelterin components TRF1 and TIN2. The goal of this project is to synthesize and evaluate novel peptide-like molecules, analogues of a key interacting region of TIN2 that can compete effectively for the binding sites on TRF1 and so lead to the destabilization of telomere structure. We will report results from a combination of computational and experimental investigations. We will discuss the application of the MM-GBSA molecular modeling method to predict short TIN2-like peptides with optimal TRF1-binding affinities. We will describe the synthesis of selected peptides bearing a fluorescent tag designed to avoid compromising binding affinity. We will discuss the expression and purification of full-length hTRF1 protein and the development of an in vitro (fluorescence polarization) assay, with the aim of identifying a lead compound. Presented by: Salih, Twana 175 Poster 104 Uncapped telomeres as determinant for cell sensitivity to G-quadruplex ligands Erica Salvati, Angela Rizzo, Sara Iachettini, Pasquale Zizza, Chiara Cingolani, Carmen D'Angelo, Manuela Porru, Annamaria Biroccio Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy Over the past decade, many chemical classes of G-quadruplex (G4) ligands have been described for their ability to target and damage telomeres, leading to detrimental effects on tumor cells. The apparent selectivity of G4 ligands towards transformed cells, led us to ascertain the presence of differences in telomere structures and functions in normal vs transformed cells in a cell model system of genetically controlled transformation. Among the different comparative evaluations, the analysis of the telomere status, in interphasic and metaphasic nuclei, revealed a progressive increase in the number of telomere dysfunction (identified as TIFs) from normal to transformed cells. The majority of TIFs in interphasic nuclei exhibited the presence of gammaH2AX but not of 53BP1 foci, indicating that these telomere damages were not-processed by the DNA damage repair pathways. In agreement with this, gammaH2AX foci were detectable also in metaphase telomeres (meta-TIFs) in transformed cells and the presence of a basal telomere dysfunction correlated with the drug-induced telomere damage and cell death. The functional relevance of this observation was directly assessed by showing that fibroblasts with artificially uncapped telomeres, obtained by depleting components of the telomeric complex, became sensitive to G4 stabilization. On the other hand, telomeres of normal cells were not completely resistant to G4 stabilization, resulting damaged upon exposure to higher dosage of G4 ligands as well, and leading to cell death. Anyway the different degree of sensitivity offers a therapeutic window for a new class of molecules with anti-tumoral features. Presented by: Salvati, Erica 176 Poster 105 Telomere maintenance mechanisms without telomerase are of clinical relevance in colorectal carcinoma Sandra Sampla,*, Stefan Stättnerb, Fabienne Bastianc, Friedrich Wrbad, Brigitte Wolfe, Jeremy Hensonf, Loretta Laug, Roger Reddelh, Klaus Holzmanna a Division of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University Vienna, Austria b Department of Surgery, Paracelsus Medical University, Salzburg, Austria c Department of Surgery, Kaiser Franz Josef-Spital/SMZ-Süd, Vienna, Austria d Department of Pathology, University Hospital, Vienna, Austria e Department of General Surgery, Medical University Vienna, Austria f Prince of Wales Clinical School, Faculty of Medicine, UNSW Australia g The Children’s Hospital at Westmead, Sydney, NSW, Australia h Children's Medical Research Institute, Sydney, NSW, Australia Most colorectal carcinomas (CRCs) use telomerase as telomere maintenance mechanism (TMM). The estimated 10% without telomerase activity (TA) may use alternative lengthening of telomeres (ALT) or not defined TMM (NDTMM), with unknown clinical importance. This led us to study the potential association between TMM and outcome for CRC patients. Tumor and adjacent non-tumor tissues from 68 patients were analyzed for TA and ALT by qPCR-TRAP and C-circle assay, respectively. Negative qPCR-TRAP results were validated by IP-TRAP. TP53, ATRX, DAXX and TERT expression and ALT-associated PML bodies were investigated by immunohistochemistry and TP53 mutations by exon-sequencing. TA was detected in 90% (61/68) of tumors and 34% (23/67) of non-tumor tissues, with means of 31 and 1.8 total product generated (TPG) units, respectively. ALT and NDTMM were each identified in 6.0% (4/68) of tumors. All four ALT tumors showed either mutated TP53 or loss of ATRX expression. One ALT tumor additionally showed TA and heterogeneous TERT expression on tumor sections. TP53 was expressed in all cells of this tumor. In contrast, tumors with either TA or ALT showed TERT expression in all or none of the cells, respectively. Outcome was best when tumors used ALT compared to TA (p=0.0393) or NDTMM (p<0.0001), with median survival time of >116, 68.3 and 18.4 months, respectively. Screening a second cohort of 262 patients identified 8 further ALT cases with favorable outcome. TA and ALT tumorcells might coexist in CRC patient’s tumors. Our data suggest clinical relevance of TMM without telomerase as prognostic marker for CRC. Presented by: Sampl, Sandra 177 Poster 106 Telomere Structure and Maintenance in Trypanosoma brucei Ranjodh Sandhu, Bibo Li 1Cleveland State University, Center for Gene Regulation in Health and Disease, Dept. of Biological, Geological, and Environmental Sciences, Cleveland, OH 44115 Telomere recombination is particularly important for Trypanosoma brucei, a protozoan parasite and the causative agent of human African trypanosomiasis. T. brucei evades the host’s immune response by regularly changing its variant surface glycoproteins (VSG), and homologous recombination is one of several important mechanisms for VSG switching. Therefore, abnormal telomere recombination may affect VSG switching efficiency. So far, the telomere terminal structure in T. brucei is poorly understood. We have recently adopted adaptor ligation, ligation-mediated primer extension, and ligation-mediated PCR assays. These assays allowed us to determine the terminal nucleotide on both strands and the length of the telomere G-overhang. We observed that the majority of T. brucei telomeres have very short G-overhangs that end in 5’-TTAGGG-3’. In addition, terminal nucleotide of the C-rich strand is tightly regulated and mostly ends in 5’-ACC. We also identified the RNA component of T. brucei telomerase, TbTR, and provide phylogenetic and in vivo evidences for TbTR’s native folding and activity. TbTR is processed through trans-splicing and is a capped transcript that interacts and copurifies with TbTERT in vivo. Deletion of TbTR caused progressive shortening of telomeres at a rate of 3–5 bp/PD, which can be rescued by ectopic expression of a wild type allele of TbTR in an apparent dose-dependent manner. Remarkably, introduction of mutations in the TbTR template domain resulted in corresponding mutant telomere sequences. In addition, we found that the 5’-TTAGGG-3’ ending telomere G-overhangs are greatly diminished in cells lacking either telomerase or TbKU80, indicating that T. brucei telomerase contributes greatly to the normal telomere G-overhang structure. Presented by: Sandhu, Ranjodh 178 Poster 107 A Shelterin-RTEL1 interaction required for maintaining telomere integrity Grzegorz Sarek, Stephanie Panier, Jean-Baptiste Vannier, Simon Boulton DNA Damage Response laboratory, London Research Institute, Cancer Research UK, Clare Hall, South Mimms, EN6 3LD, UK Telomere homeostasis and DNA repair are fundamental processes, which are vital for genome integrity. RTEL1 (Regulator of telomere length 1) is an essential DNA helicase that resolves telomere loops (T-loops) and counteracts telomere fragility, thereby ensuring integrity of the telomere. Recently, we demonstrated that RTEL1 associates with the replisome through binding to proliferating cell nuclear antigen (PCNA) via a PCNA interacting protein (PIP) motif. Mutant cells deficient for the RTEL1-PCNA interaction exhibited telomere replication defects, evident from the accumulation of fragile sites at telomeres, which we attributed to a failure to unwind telomeric G4-DNA structures. In contrast, the RTEL1–PCNA complex was dispensable for dismantling T-loops. These data raise the possibility that RTEL1 engages other interacting partners to resolve T-loops. We show here that RTEL1 binds directly to Shelterin and this interaction occurs predominantly during S-phase. We identify key amino acids required for the RTEL1-Shelterin interaction, including specific residue within Shelterin and conserved residues within RTEL1, some of which are mutated in a subset of Dyskeratosis congenital (DC) patients. Analysis of mutant cells defective for the RTEL1-Shelterin interaction revealed inappropriate T-loop resolution accompanied with telomere loss and telomeric circle formation. In contrast to the RTEL1-PIP box mutant, loss of the RTEL1-Shelterin interaction had no measurable impact on genome-wide or telomere replication. Finally, we identify a single amino-acid substitution mutation within Shelterin that abolishes RTEL1 recruitment to telomeres and phenocopies RTEL1 deficiency. These findings define a critical RTEL1-Shelterin interaction, which is required to promote T-loop disassembly during S-phase and is compromised in a subset of RTEL1 DC patients. Presented by: Sarek, Grzegorz 179 Poster 108 Associations between five-factor model of personality and leukocyte telomere length in elderly men and women – the Helsinki Birth Cohort Study (HBCS) Katri Savolainena,*, Johan Erikssonb, Eero Kajantieb, Anu-Katriina Pesonena, Katri Räikkönena a Institute of Behavioural Sciences, University of Helsinki, FINLAND Diabetes Prevention Unit, Department of Chronic Disease Prevention, National Institution for Health and Welfare, Helsinki, FINLAND b Introduction Personality traits have been associated with longevity, and morbidity and mortality of cardiometabolic diseases and mental disorders. The underlying mechanisms are not fully understood. Accelerated cellular aging may play a role in this process. We studied if personality traits in later adulthood, as defined in the five-factor model, are associated with a biomarker of cellular vitality, leukocyte telomere length (LTL). Methods At a mean age of 63.4 (SD = 2.8) years, 1671 (742 men, 929 women) participants from the Helsinki Birth Cohort Study, filled in the Neuroticism, Extraversion and Openness Personality Inventory (NEOPI). LTL was measured at a mean age of 61.5 (SD = 2.9) years by using a real-time quantitative PCR method. Results None of the NEO-PI personality traits were significantly associated with the LTL in the analyses of both sexes combined. In the sex-specific analyses, men who scored higher on agreeableness (p = .016) or lower on neuroticism (p= .040) and women who scored lower on agreeableness (p = .016) had shorter LTL. Conclusions FFM dimensions of personality were not associated with LTL in a sample of elderly individuals. Our findings suggest that LTL, a biomarker of cellular aging, may not offer insight into the associations between personality, longevity and health. Presented by: Savolainen, Katri 180 Poster 109 A mouse model for ICF syndrome does not recapitulate the telomeric phenotype of human ICF syndrome patients Gal Laroma, Shiran Yehezkela, Robert J. Duszynskib, Lucy A. Godleyb, Claire Francastelc, Guillaume Velascoc, Sara Seliga,* a Rambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion b Department of Medicine, Section of Hematology Oncology, The University of Chicago c Centre Epigénétique et Destin Cellulaire, Université Paris Mutations in the human DNA Methyltransferase 3B (DNMT3B) gene result in autosomal-recessive ICF (Immunodeficiency, Centromeric instability, Facial anomalies) type I syndrome. The subtelomeres in these patients are abnormally hypomethylated and subtelomeric-hypomethylation is associated with high levels of TERRA, accelerated telomere shortening and premature senescence. With the aim of establishing a model system for studying the in vivo consequences of hypomethylated subtelomeres, we characterized the telomeric phenotype in mouse embryonic fibroblasts (MEFs) carrying human ICF type I mutations. Mouse subtelomeric regions in ICF-MEFs were only slightly hypomethylated in comparison to wild-type (WT) subtelomeres. Also dissimilar to human ICF cells, telomere-FISH analysis did not reveal missing signals, TRF analysis demonstrated no significant difference in the mean telomere length and TERRA levels were extremely low. We found normal levels of telomere sister chromatid exchange and no colocalization of PML bodies with telomeres. 3D analysis of centromeric and telomeric-nuclear positioning indicated that the position of telomeres, but not centromeres, differed significantly between WT and ICF MEFs, however human WT and ICF fibroblasts demonstrated similar nuclear positioning of these chromosomal regions. Our studies indicate that subtelomeric methylation, which plays an important role in telomere length and function in human cells, is a less significant player in the generation of the proper heterochromatinization of mouse telomeres. To this end, ICF-MEFs do not exhibit the main telomeric phenotypes observed in human ICF cells and therefore are not a good surrogate model for studying this phenotypic aspect of human ICF syndrome. Presented by: Selig, Sara 181 Poster 110 Dissecting Alternative Lengthening of Telomeres in the Nematode Caenorhabditis elegans Beomseok Seoa,*, Chuna Kima, Mark Hillsb, Sung Sanghyuna, Stephane Flibottec, Donald Moermanc, Lee Junhod a School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea, 151-742 b Terry Fox Laboratory, BC Cancer Agency, Vancouver V5Z 1L3, Canada c Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada d School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea, 151-742; Department of Biophysics and Chemical Biology, Seoul National University, Seoul, Korea 151-742 Eukaryotes have linear chromosomes, which causes the 'end-replication problem'. With consecutive rounds of DNA replication, the ends of the chromosomes cannot be completely replicated due to the inability to prime the lagging strand. One way to solve this problem is to use a reverse transcriptase telomerase, which adds telomeric repeats to the end of the chromosomes de novo. However, it has also been found that telomeres can be maintained without telomerase in yeast and some cancer cells. This is called Alternative Lengthening of Telomere (ALT) mechanism, which is proposed to be based on a process involving recombination although exact mechanisms are largely unknown. In the nematode Caenorhabditis elegans, the telomerase deficient trt-1(ok410) mutant strain becomes sterile after several generations as a result of shortening of telomeres (mortal germline phenotype). We mutagenized trt-1 mutant worms and isolated suppressor mutants whose mortal germline phenotype was suppressed. The transgenerational lifespan of these mutants are fully recovered. The Terminal Restriction Fragment (TRF) assays and fluorescent in situ hybridization (FISH) revealed that telomeres of the suppressor mutants were lengthened and had distinct properties from wild type telomeres. Whole genome sequencing revealed that the telomeres of suppressor mutants were amplified with variant repeats originated from either internal telomere-like repeats or subtelomeric DNA. N2 and CB4856 wild isolates of C. elegans seem to have different hot spots for telomere recombination. We are currently seeking candidate genes responsible for ALT mechanism in C. elegans. These results suggest that C. elegans ALT mechanism has a new mode of template selection, which can be differentially regulated in distinct genetic backgrounds. Presented by: Seo, Beomseok 182 Poster 111 Subtelomeric elements of the shortest telomere are required for TERRA expression and buffer senescence onset Kamar al zaman Serhala,*, Marco Grafb, Pascale Joliveta, Maria Teresa Teixeiraa, Brian Lukeb a Institut de Biologie Physico-chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, UMR8226 CNRS/UPMC b Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Deutsches Krebsforschungszentrum (DKFZ)-ZMBH Alliance, Heidelberg, Germany. Despite their heterochromatic state, telomeres express telomeric repeat containing RNAs (TERRA). In telomerase negative pre-senescent cells, TERRA RNA-DNA hybrids were shown to promote homologydirected repair (HDR) at telomeres. In a parallel study, it was determined that the shortest telomeres in senescing cells, are more prone to undergo processing events related to HDR. Taken together, we set out to determine whether critically short telomeres were subject to increased levels of HDR due to the up-regulation of TERRA and hence RNA-DNA hybrids at short telomeres. Here, we have employed the FLP-FRT system to engineer a yeast strain in which one specific telomere is the shortest telomere in the cell. We compared two variants of an inducible Very Short Telomere (VST), one where the endogenous subtelomere was left intact (VST-6R) and one where the subtelomere was removed, preventing TERRA expression (VST-7L). We found that upon telomerase deletion and shortening of the engineered telomere, the VST strain lacking subtelomeric elements senesced at a significantly increased rate compared to the strain containing the non-modified VST subtelomere. This shows that the subtelomeric elements of the shortest telomere delay senescence. Strikingly, TERRA levels were significantly increased at the VST-6R, supporting the idea that transcription at telomeres is derepressed as telomeres shorten in the absence of telomerase. Moreover, we detect an increase of RNA-DNA hybrids at this telomere. We propose a model whereby TERRA transcription is increased at critically short telomeres. This generates RNA-DNA hybrids, which in turn promote HDR at the shortest telomeres, delaying the senescence onset. Presented by: Serhal, Kamar al zaman 183 Poster 112 Telomere alterations and chromosome segregation defects induced by prolonged oxidative stress treatment Elisa Coluzzia, Rossella Buonsantea, Anthony J. Asmarb, Kelley L. Millerb, Daniela Ciminib, Antonella Sguraa a b University of Roma Tre, Department of Science, V.le G. Marconi, 446, 00146, Rome, Italy Virginia Tech, Department of Biological Sciences, 1015 Life Science Circle, Blacksburg, VA, 24061, USA Telomeres represent an important target of oxidative stress, and oxidative stress-induced damage may produce telomere length-dependent chromosome mis-segregation. In this study, we investigated the effects of prolonged oxidative stress on telomere length and chromosome segregation in human primary fibroblasts (MRC-5) treated daily with 10μM H2O2 and analysed at regular intervals over a 45– day period. Analysis of telomere length showed initial telomere shortening followed by lengthening in H2O2-treated cells over the first 20 days of treatment. This cycle also seemed to repeat at later times, but we focused our subsequent analyses on the first 20 days of treatment. We next investigate potential mechanisms of telomere elongation, and first performed RTQ-TRAP assay, which did not reveal any telomerase activity. We then asked whether telomere elongation may be induced by the ALT mechanism. To this end, we assessed two ALT markers: telomeric-sister chromatid exchanges (TSCE) and colocalization of telomeres with PML proteins. Both these ALT markers were observed at higher frequencies in H2O2-treated cells compared to untreated cells at the 15 day time point, which corresponded to the time of observed telomere elongation. Finally, live- and fixed-cell experiments showed a H2O2-induced increase chromosome bridges at 5 days of treatment and a reduction at 15 days, corresponding to the observed telomere shortening and elongation, respectively. –Taken together, these data suggest that oxidative stress initially induces telomere shortening and anaphase chromosome bridges in human primary fibroblasts. Under these conditions, these cells activate the ALT mechanism, which restores telomere length and suppresses chromosome bridge formation. Presented by: Sgura, Antonella 184 Poster 113 Development of a high-throughput, fluorescence-based, PCR-free method of C-circle detection to screen drugs and genes targeting the alternative lengthening of telomeres (ALT) pathway in human cancers David Halvorsen, Haroldo Silva SENS Research Foundation While most cancers rely on telomerase for immortalization, a significant proportion of tumors show no detectable telomerase activity. These cancers often use a homologous recombination-based pathway known as alternative lengthening of telomeres (ALT). However, no ALT-specific targets or therapies are currently available partially due to a lack of robust assays that quantitatively measure ALT hallmarks. Here we develop and validate a fast, high-throughput version of the established C-circle assay used to identify ALT-positive cancer cells and tumors. ALT causes abnormal telomere metabolism that generates linear and circular extra-chromosomal telomeric repeats, including partially double-stranded C-rich (CCCTAA)n circles or C-circles (CC). ALT activity is strongly correlated with and proportional to CC levels, which are measured by rolling circle amplification (RCA) using phi29 DNA polymerase. Currently, CC assays often take a few days to complete and use radioactive or chemiluminescent probes on DNAspotted membranes or complex PCR techniques that make them incompatible with high-throughput or clinical diagnostics. Our CC assay adapts established fluorescence-based methods previously validated to detect telomerase activity to quantitatively measure CC in human ALT cells from multi-well plates. We validated our assay using drugs and siRNA treatments previously established to regulate ALT activity. Unlike current CC assays, our method generates complete results in one hour post RCA or 8 hours including genomic DNA isolation. Our assay also has a broader linear response than conventional methods without loss of sensitivity. This novel CC detection platform enables large-scale screening of drugs and genetic targets for a functional role on the ALT mechanism. Presented by: Silva, Haroldo 185 Poster 114 TERT Promoter Mutations Are a Major Indicator of Poor Outcome in Differentiated Thyroid Carcinomas Paula Soaresa,*, Miguel Melob, Adriana Gaspar da Rochaa, João Vinagrec, et al. a IPATIMUP and Medical Faculty, University of Porto, Portugal Departments of Endocrinology, Diabetes, and Metabolism and Unit of Endocrinology,Faculty of Medicine, University and Hospital Center of Coimbra c IPATIMUP and ICBAS, University of Porto, Portugal b Telomerase promoter mutations (TERT) were recently described in follicular cell-derived thyroid carcinomas (FCDTC) and seem to be more prevalent in aggressive cancers. We studied 647 tumors and tumor-like lesions. A total of 469 patients with FCDTC treated and followed in five University Hospitals were included. Mean follow-up (±SD) was 7.8±5.8 years. We evaluate the predictive value of TERT promoter mutations and other clinico-pathological and molecular features (BRAF, NRAS) for distant metastasization, disease persistence at the end of follow-up and diseasespecific mortality. TERT promoter mutations were found in 7.5% of papillary carcinomas (PTC), 17.1% of follicular carcinomas (FTC), 29.0% of poorly differentiated carcinomas and 33.3% of anaplastic thyroid carcinomas. Patients with TERT-mutated tumors were older (p<0.001) and had larger tumors (p=0.002). In DTC, TERT promoter mutations were significantly associated with distant metastases (p<0.001) and higher stage (p<0.001). Patients with DTC harboring TERT promoter mutations were submitted to more radioiodine treatments (p=0.009) with higher cumulative dose (p=0.004), and to more treatment modalities (p=0.001). At the end of follow-up, patients with TERT-mutated DTC were more prone to have persistent disease (p=0.001). TERT promoter mutations were significantly associated with disease-specific mortality [in the whole FCDTC (p<0.001)] in DTC (p<0.001), in PTC (p=0.001) and in FTC (p<0.001). After adjusting for age at diagnosis and gender, the HR was 10.35 (95%CI 2.01-53.24; p=0.005) in DTC and 23.81 (95%CI 1.36-415.76; p=0.03) in PTC. TERT promoter mutations are an indicator of clinically aggressive tumors, being correlated with worse outcome and disease-specific mortality in DTC. TERT promoter mutations have an independent prognostic value in DTC and, notably, in PTC. Presented by: Soares, Paula 186 Poster 115 The role of DAXX and ATRX in telomere maintenance Zhou Songyanga,*, Quanyuan Heb, Mengfan Tangc a Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030; and SYSU-BCM Joint Research Center, School of Life Sciences, SYSU, Guangzhou, P.R. China b Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030 c SYSU-BCM Joint Research Center, School of Life Sciences, SYSU, Guangzhou, P.R. China Mutations in the DAXX and ATRX have been found in human pancreatic neuroendocrine tumors and pediatric glioblastoma, and correlate with the Alternative Lengthening of Telomeres (ALT) phenotype. DAXX has been shown to interact with ATRX and mediate histone H3.3 deposition. However, the role of DAXX and ATRX in telomere maintenance and development of ALT-type cancers remains unclear. We found that endogenous DAXX can localize to Cajal bodies, associate with telomerase, and regulate telomerase assembly and targeting. Furthermore, disease mutations located in different regions of DAXX differentially impacted its ability to interact with its binding partners, and its targeting to Cajal bodies and telomeres. These findings support a DAXX-centric pathway for telomere regulation. To further understand the function of DAXX and ATRX, we studied DAXX and ATRX protein complexes by IP mass spec and performed genome-wide Chip-seq analyses. Our data suggested that DAXX and ATRX have both overlaping and distinct functions in controling chromatin integrity, providing new insights into cellular mechanisms that regulate telomere stability and ALT. Presented by: Songyang, Zhou 187 Poster 116 Choice of processing dysfunctional telomeres influences stem cell aging and survival Tobias Sperka, Satjavani Ravipati, Omid Omrani, Lenhard Rudolph Leibniz Institute of Age Research, Fritz-Lipmann-Institute Telomere dysfunction endangers genome integrity, engages the DNA repair machinery and evokes checkpoint responses including the p53 network. The pool of proliferating stem and progenitor cells is protected from genomic instability and malignant transformation by checkpoint induction and depletion of damaged stem cells. Accumulating depletion subsequently impairs organ maintenance contributing to tissue aging. Our previous data indicates that critically short telomeres induce cell cycle arrest whereas telomere-fusion evokes apoptotic cell death. Here, we want to explore the possibility of directing the processing of dysfunctional telomeres in aging late generation mTerc-/- mice by co-deleting the DNA repair factors 53BP1, H2AX or MDC1, respectively. The homozygous deletions of genes encoding for inhibitors of DNA end-resection (MDC1, H2AX) leads to increased activation of p53 in telomere dysfunctional stem cells, telomere shortening, and chromosomal fusions. The affected mice have shorter lifespan and aggravated organ atrophy. In contrast, deletion of 53BP1 – an inhibitor of end-resection, which also mediates ligation of DNA ends – reveals evidence that end-resection per se is not limiting stem cell function. Instead the increase in chromosomal fusion in response to end-resection is mediating adverse effects of telomere dysfunction on stem cell maintenance. Presented by: Sperka, Tobias 188 Poster 117 Evidence for telomerase- and Rad52-independent sequence alterations at yeast telomeres Clémence Claussin, Michael Chang, Sonia Stinus European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen In the yeast Saccharomyces cerevisiae, cells lacking telomerase senesce after 60-80 generations but a small subset of cells can overcome senescence using recombination-mediated mechanisms to become ‘survivors’. Two main types of survivors have been described: type I and type II. Both types re uire Rad52, a protein necessary for almost all recombination events in yeast. Type I survivors involve the amplification of subtelomeric elements, while type II survivors resemble the majority of human ALT cancer cells in that they both exhibit amplification of the terminal telomere repeats. Recombination proteins are also known to be important in pre-senescent cells, before the formation of survivors. Telomerase mutants lacking Rad52 exhibit accelerated senescence, but the shortening rate of telomeres is unchanged. The precise function of recombination proteins in presenescent cells is unknown. Using a telomere sequencing approach, telomere recombination events have been detected in pre-senescent cells, although at a much lower frequency than in survivors. Surprisingly, we have found that these events still occur in rad52Δ mutants and therefore take place in a Rad52-independent manner. We are currently studying how these events occur as well as their significance with respect to telomere maintenance. Presented by: Stinus, Sonia 189 Poster 118 In vivo Interaction of Telomerase, Telomere homeostasis and the Human Papillomavirus Oncogenes Charis Achilleos, Stella Michael, Katerina Strati Department of Biological Sciences University of Cyprus Human papillomaviruses (HPVs) are responsible for the majority of cervical cancers as well as other types of epithelial cancers. Carcinogenesis is dependent on the long-term expression of the viral oncogenes E6 and E7. E6 has been shown to regulate the telomerase enzyme at the transcriptional and post-transcriptional level and even bind telomeric repeats in vitro. E7 has been shown to induce alternative lengthening of telomeres (ALT). The multiple ways of regulating in particular the telomerase enzyme suggest that it represents a crucial target for the virus. It has been speculated that this regulation may be important to the cancer-causing properties of the virus but also integral to its viral lifecycle. In order to understand the in vivo implications of the interaction of the HPV oncogene and telomere homeostasis we have employed mouse models. We are utilizing mice in which the keratin 14 promoter directs the expression of HPV16 E6 or E7 to stratified epithelial tissues of mice. These tissues are the target of natural infection. The expression of E6 and E7 leads to aberrant differentiation, increased and unscheduled DNA synthesis, a mobilization of epidermal stem cells, an expansion of stemness marker expression, formation of tumors etc. We have crossed these mice on a Terc-/background and will present acute phenotypes on the epidermis and their dependence on functional telomerase enzyme. Presented by: Strati, Katerina 190 Poster 119 in vivo isolation of telomeric proteins in the nematode Caenorhabditis elegans Sanghyun Sunga,*, Beomseok Seoa, Junho Leeb a Department of Biological Sciences, Seoul National University, Seoul, Korea, 151-742 Department of Biological Sciences & Department of Biophysics and Chemical Biology, Seoul National University, Seoul, Korea, 151-742 b Telomeres are special nucleoprotein complexes that cap the ends of the eukaryotic linear chromosomes to maintain genomic integrity. Many interesting aspects of the telomeres of Caenorhabditis elegans have been studied; for example, the extension of organismic lifespan by longer telomeres and alternative lengthening of telomeres at the organismic level have been reported. It is conceivable that the protein components of the telomeres may have critical roles in these processes as regulators of length, stability, and yet unknown interesting mechanisms of telomeres. Because most telomeric proteins found in higher mammals (or even yeasts) do not have homologous partners in C. elegans, there is a strong need for biochemical approaches to identify them in C. elegans. Previously, we used a nucleic acid affinity capture method to isolate specific proteins associated with telomeric DNA. Although this in vitro method was useful, it had limitations for describing all proteins that may act in the in vivo context. Therefore, in this study, we utilized the recently developed technique called proteomics of isolated chromatin segments(PICh). PICh uses unique nucleic acid to capture the endogenous segments of chromatin which are fixed before hybridization, so it can be a direct and quantitative tool. Currently, we are investigating the telomere-specific proteins of wild type C. elegans. Presented by: Sung, Sanghyun 191 Poster 120 The DNA-end-replication problem and the establishment of replicative senescence Julien Soudeta, Emilie Falleta, Pascale Joliveta, Michael Lisbyb, Eric Gilsonc, Maria Teresa Teixeiraa,* a Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, Institut de Biologie PhysicoChimique, UMR8226 CNRS/UPMC b Department of Biology, University of Copenhagen c Institute for Research on Cancer and Aging, Nice (IRCAN), University of Nice Sophia-Antipolis, CNRS UMR7284/INSERM U1081, Faculty of Medicine In the absence of telomerase, yeast telomeres shorten 2.5-5 nt/cell division leading to replicative senescence. However, the model for telomere shortening is currently incomplete and the exact contribution of the telomeric 3’-overhang to the shortening rate remains unclear. By following the dynamics of telomeres during replication at near-nucleotide resolution, we find that the leading strand synthesis generates blunt-end intermediates before being 5’-resected and filled-in. The shortening rate is set by positioning the last Okazaki fragments at the very ends of the chromosome. Thus, telomeres shorten in direct proportion to the 3’-overhang lengths of 5–10 nucleotides which are present in parental templates. Furthermore, Cdc13 coordinates leading and lagging strand syntheses. Yet, these telomere shortening “rules” do not apply when telomeres reach a critical length. By tracking individual telomeres, we show that telomeres are subjected to different pathways depending on their length. We demonstrate a progressive accumulation of subtelomeric ssDNA through 5’-3’-resection as telomeres shorten. We also uncovered that Rad5, a DNA helicase/Ubiquitin ligase of the error-free branch of the DNA damage tolerance (DDT) pathway associates with native telomeres. We propose that DDT acts in a length-independent manner whereas a HR-based repair using the sister chromatid as a template buffers precocious 5’-3’-resection at the shortest telomeres. Taken together, our data unravel a precise choreography of telomere replication elucidating the DNA-end-replication problem and provide a new framework to understand the control of the cell proliferation potential. Funded by ERC-STG-2010 D-END. Presented by: Teixeira, Maria Teresa 192 Poster 121 DNA DAMAGE SIGNALLING IN THE RECRUITMENT OF TELOMERASE TO TELOMERES Adrian Tonga,*, Josh Sterna, Scott Cohena, Xu-Dong Zhub, Tracy Bryana a b Children's Medical Research Institute, Sydney, Australia. McMaster University, Hamilton, Ontario, Canada. Telomerase recruitment to telomeres is a cell-cycle regulated process important in determining telomere length. Understanding this process is important for telomere biology and potential therapeutic drug design in regulating tumorigenesis. The yeast homologues of the DNA damage protein ATM is involved in telomerase recruitment, but ATM’s involvement in telomerase recruitment to telomeres has not been demonstrated in human cells. TRF1 has been shown to be phosphorylated by ATM, leading to telomere elongation. We therefore directly tested the hypothesis that ATM mediates telomerase recruitment to the telomere via phosphorylation of TRF1. We have employed an hTR/telomere fluorescent in situ hybridisation assay that allows the direct visualisation of endogenous telomerase to confirm that telomerase is recruited to the telomere at mid S phase in 293T cells. ATM silencing resulted in decreased telomerase recruitment, as well as a slight decrease in the ability of telomerase to assemble and form a functional protein complex, while TRF1 knockdown resulted in the deregulation of telomerase recruitment across the cell cycle. Co-knockdown of TRF1 partially rescued the phenotype of diminished telomerase recruitment resulting from ATM knockdown alone, demonstrating that these two proteins act in the same pathway. Treatment of cells with the ATM kinase inhibitor Ku55933 established that the kinase activity of ATM is vital in determining telomerase localisation to the telomere via TRF1. We have provided direct evidence that ATM phosphorylation events regulate telomerase recruitment, assembly and telomere accessibility through TRF1. Presented by: Tong, Adrian 193 Poster 122 Effects of bilirubin on telomere integrity and dynamics Anela Tosevskaa,*, Christine Moelzerb, Karl-Heinz Wagnera a b Research Platform Active Ageing, University of Vienna, Austria University of Vienna, Austria Bilirubin has long been considered a redundant byproduct of heme catabolism and potentially toxic if present in high concentrations in human serum. In contrast, mildly elevated bilirubin levels, characteristic of Gilbert Syndrome, are considered beneficial and are inversely correlated with cancer and CVD risk as well as the onset of metabolic syndrome. Mechanistically, this double nature of bilirubin is attributed to its concentration-dependent pro- or anti- oxidant capacity. Low grade hyperbilirubinemia (≈17,1µM in serum) is proposed to protect cellular macromolecules such as DNA from damage caused by excess generation of reactive oxygen species (ROS). We hypothesize that bilirubin also has protective effects against ROS-mediated telomere attrition. Our present study examines its possible direct or indirect effects on telomere integrity. To address this issue, we use a cell culture model of healthy primary human PBMCs, treated with unconjugated bilirubin (UCB) in concentrations physiologically present in the circulation. We use qPCRbased methods to assess telomere length and FPG-sensitive (8-oxo-dG) sites in telomeres. Furthermore, we evaluate the role of bilirubin in the recruitment of DNA repair mechanisms and telomere length maintenance under defined stress conditions. We combine these outcomes with flowcytometric cell cycle analysis in order to get an insight of cell survival and functionality following bilirubin treatment. Our findings suggest that telomeres could act as mediators in the bilirubin protective machinery against age-related and metabolic diseases. Presented by: Tosevska, Anela 194 Poster 123 Single-molecule imaging of RTEL1 in embryonic stem cells Evert-Jan Uringaa,*, Andrew Robinsonb, Karl Duderstadtb, Antoine Van Oijenb, Peter Lansdorpc a European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, The Netherlands b Zernike Institute for Advanced Materials, Centre for Synthetic Biology, University of Groningen, The Netherlands c European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, The Netherlands; Terry Fox Laboratory, BC Cancer Research Centre and Department of Medicine, University of British Columbia, Vancouver, BC Telomere maintenance and DNA repair are crucial processes that protect the genome against instability. The essential helicase Regulator of Telomere Length 1 (RTEL1) is required for unwinding DNA structures that arise during replication, recombination and repair. Substrates for RTEL1 include secondary structures formed from trinucleotide repeats and G-rich DNA. In addition, RTEL1 promotes T-loop disassembly and acts during homologous recombination by dismantling the displacement-loop. Fluorescence imaging can provide crucial information on the behavior of RTEL1 in living cells. Since overexpression of RTEL1 is toxic to cells, we generated mouse embryonic stem cells (ESCs) in which endogenous RTEL1 was replaced with a fluorescently-tagged RTEL1 fusion protein (RTEL1-venus). Our previous attempts to study RTEL1-venus in ESCs have been hampered by the very low expression levels of endogenous RTEL1. Here we report studies of RTEL1 using single-molecule localization microscopy. This technique has been used to study replication and repair in bacterial cells and allows studies of molecular properties of proteins that are not accessible through more traditional microscopy methods. Upon treatment with the topoisomerase II inhibitor etoposide, we observed a dose-dependent increase in the number of RTEL1-venus foci. Using single-molecule localization microscopy we determined that of these foci, 52% contain a single molecule of RTEL1-venus. To gain insight into the dynamic behavior of RTEL1 molecules, we also analysed the recovery of photobleached foci. We found that 50% of RTEL1 foci recover fluorescence after photobleaching, with a recovery rate similar to that of the diffuse signal. This finding suggests that RTEL1 molecules at sites of damage have either fast or slow exchange kinetics. Presented by: Uringa, Evert-Jan 195 Poster 124 Insights into telomere protection by Ku heterodimer Sona Valuchovaa,*, Eliska Janouskovab, Ctirad Hofrb, Karel Rihaa a b Gregor Mendel Institute of Molecular Plant Biology GmbH, Dr. Bohr-Gasse 3, 1030 Vienna, Austria Central European Institute of Technology, Kamenice 753/5, 601 77 Brno, Czech Republic Ku is a conserved protein present in all domains of life and viruses. It is involved in DNA double-strand break repair and telomere protection. Both processes are essential for maintenance of genome integrity and deficiencies in these processes were associated with human diseases. At first, the role of Ku in telomere protection in eukaryotic organisms seems counterintuitive, because it mediates fusions of unprotected telomeres. However, its action as a resection and recombination inhibitor seems to be beneficial for natural chromosome ends. We are trying to understand the mechanism that prevents initiation of DNA repair by Ku on telomeres while retaining its end-protective function. In Arabidopsis thaliana, presence of Ku heterodimer is necessary for integrity of blunt ended telomeres by inhibiting nucleolytic resection. Such resection takes place after DNA replication to create 3’ G-overhangs in many organisms. We hypothesize that Ku physically hinders/regulates the telomeric resection by binding to chromosome ends. To examine the importance of DNA-Ku interaction in telomere protection, we engineered series of mutant Ku complexes with altered DNA binding properties. We mutated conserved residues present on DNA interaction surface according to the available crystal structure. We identified human Ku proteins with in vitro decreased affinity to DNA and increased incidence on DNA termini. The same mutations of homologous residues had similar effect on complexes from A. thaliana. Analysis of the mutant complexes in vivo suggests that the DNA interacting residues are important for telomeric function(s) of the Ku complex. Presented by: Valuchova, Sona 196 Poster 125 Depression and shortened telomeres: implication on shelterin and telomerase Yabin Weia,*, Lena Backlundb, Lina Martinssonb, Aleksander A. Mathéc, Gregers Wegenerd, Martin Schallinga, Catharina Lavebratta a Department of Molecular Medicine and Surgery, Neurogenetics Unit, Karolinska Institutet, Stockholm, Sweden b Department of Clinical Neuroscience, Centre for Psychiatric Research and Education, Karolinska Institutet, Clinic for Affective Disorders, Karolinska University Hospital, Stockholm, Sweden c Department of Clinical Neuroscience, Section for Psychiatry, Karolinska Institutet, Stockholm, Sweden d Centre for Psychiatric Research, Aarhus University Hospital, Risskov, Denmark Eukaryotic telomeres are protective DNA-protein complexes that form the chromosome ends. A number of studies reported shorter leukocyte telomere length (LTL) to be associated with major depression, but telomere protective proteins, known as shelterin, has never been investigated. Further, how long-term lithium treatment protected against telomere shortening remains elusive. In the hippocampus region, shelterin components, telomerase activity and telomere length (TL) were assessed: 1) between the Flinders Sensitive Line (FSL), a genetic rat model of depression-like and its controls, the Flinders Resistant Line (FRL); 2) the FSL rats before and after dietary lithium treatment for 3 weeks. In addition, we assessed if the single nucleotide polymorphism (SNP, rs2736100) associated with depression or depressive episodes in a Swedish population based cohort (PART) and in bipolar patients (BP) respectively. Finally, by using human whole saliva DNA we compared the TL between depression patients and healthy controls, and testing the correlation of TL between DNA from whole saliva and leukocytes. The FSL rats, compared to its counterpart, exhibited significantly downregulation of Terf2, Rap1,Tert expression and telomerase activity, which were associated with shorter hippocampal TL. Lithium treatment only rescued the Tert expression and telomerase activity in the FSL. rs2736100 was associated with depression in PART cohort and increased depressive epidosdes in BP. TL, tested in whole saliva DNA, was decreased in depression patients compared to the controls, which positively correlated with leukocytes TL. This is the first study characterizing shelterin in depression-like rat model and lithium’s mechanism in protecting against telomere shortening. Presented by: Wei, Yabin 197 Poster 126 Chromosome Instability: Major Events Likely Start in the Telomeres, Stupid Ted Weinerta,*, Tracey Beyera, Pete Vintona, Rachel Langstona, Margherita Paschinib, Vicki Lundbladb a b University of Arizona Salk Institute for Biological Studies We have been studying how large-scale chromosome rearrangements arise. We developed a budding yeast model system in which loss of a gene (CAN1) allows identification of loss or recombination on a marker chromosome that is not essential (in a ChrVII disome). Surprisingly, a major event is one in which an unstable chromosome is formed, probably a dicentric. The dicentric undergoes additional events to form progeny cells (forming photogenic sectored colonies, if we do say so ourselves) that contain different chromosome structures. We believe that replication fork error leads to faulty template switch to form dicentrics. A major site of fusion was proposed to be 403 kb from the telomere end (on a 1.1Mb chromosome); we proposed this 403 site is "fragile" and where events initiate. We now find that the 403 site is NOT where events likely initiate, but rather is where instability events often resolve. Events appear to initiate, instead, in the telomere; events are up in est2 mutants (5 fold increase compared to wildtype cels), in cdc13 hypomorphs (13 fold increase) , and in tel1 rad17 double mutants (>300 fold increase)! (And since, as it’s widely known, at least one of us--TW--loves telomere people, that events initiate in or near a telomere is a lovely turn of events!) We are re-interpreting a lot of our earlier data with this telomere-centric view, and propose that replication forks stall in telomeres, then undergo template switching to form a first dicentric, which rearranges as cells struggle to divide. Presented by: Weinert, Ted 198 Poster 127 Structure-Function studies on Cdc13 Sofiane Y. Mersaoui, Raymund J. Wellinger Université de Sherbrooke Budding yeast Cdc13p is a key player acting to assure telomere maintenance and genome stability. Previous studies proposed an architectural organisation with five distinct domains for Cdc13p, which include four OB-fold domains and one recruitment domain. However, many details on the in vivo roles of those domains and their potential inter-domain interactions remained elusive. We therefore constructed tagged versions of the protein where each new allele lacks one of the five proposed domains. Highlights of the results show that the first 80 amino acids of the OB1 domain contribute to stabilization of the Cdc13p. We hypothesise that this stabilization occurs after dimerization of Cdc13p. Second, the data show that the poorly characterized third domain of the protein (putative OB2) is essential for the viability. Also, Cdc13-ΔOB2 is unable to bind telomeric DNA in vivo and results in a capping deficiency. We also introduced all these alleles into yeast strains that grow in the absence of Cdc13p. Although viable, those adapted cells are sensitive to genotoxic drugs; yet an ectopic expression of Cdc13p in these cells restores normal resistance, providing evidence for a new function of Cdc13p in resistance to DNA damage. Those adapted cells thus allow an assessment of functions of cdc13-alleles that lack essential domains and also permit a delineation of the domains involved in the presumed new function of Cdc13p. We will report our findings with these experiments and integrate them into the domain organization of Cdc13p that is pertinent to its in vivo functions. Presented by: Wellinger, Raymund J. 199 Poster 128 The structural organization of the human telomerase Michael Wildauer, Christina Waldsich Max F. Perutz Laboratories, Department of Biochemistry and Cell biology, University of Vienna, Austria Telomerase plays an essential role in the maintenance of chromosome ends. This ribonucleoprotein complex consists of two major players: an RNA component called telomerase RNA (TR) and the catalytic subunit termed telomerase reverse transcriptase (TERT). Together they prevent the shortening of the telomeres during each round of cell division. This problem is also called endreplication problem and was described for the first time by Russian theorist Alexey Olovnikov in the late 1970s. Despite on-going research for more than 3 decades the exact mechanism of the telomerase complex is not known. Describing the architecture of human telomerase will be critical to understand its mechanism. Our research focuses on studying the interface between the human telomerase RNA (hTR) and the human TERT protein. So far the detailed interaction pattern of hTR and hTERT in the functional core complex remains to be discovered. Via in vivo UV crosslinking we have been able to identify several crosslinks within hTR, mainly in the pseudoknot domain and the CR4/CR5 region. These structural elements have been implicated to be part of the hTERT binding site. Presented by: Wildauer, Michael 200 Poster 129 Roles of ATRX and H3.3 to assemble a repressive chromatin state at telomeres Maheshi Udugamaa, Fiona Changa, Lyn Chana, James McGhiea, Philippe Collasb, Jeffrey Mannc, Lee Wonga,* a Monash University, Victoria, Australia University of Oslo, Oslo, Norway c Murdoch Childrens Research Institute, Victoria, Australia b Telomeres are specialised ribonucleoprotein complexes found at chromosome ends and they are absolutely essential for maintaining chromosome and genome stability. The telomere DNA is enriched for typical heterochromatin marks such as hypoacetylated histone, H4K20me3 (H4 trimethylated lysine 20), H3K9me3 (H3 trimethylated lysine 9) and Heterochromatin Protein 1 (HP1). Telomere DNA is also hypermethylated. The heterochromatic nature of telomere chromatin is important for its structure and function. Beside these epigenetic marks, our previous studies show that Alpha Thalassemia Mental Retardation X-linked (ATRX) and H3.3 are also key regulators of the telomeric chromatin. Deposition of H3.3 by ATRX and its interacting partner DAXX is essential for chromatin repression at the telomere. Function of ATRX-H3.3 in maintaining telomere chromatin repression is further demonstrated by recent studies that show a strong association of ATRX mutation with alternative lengthening of telomeres (ALT) in human cancers; 90% of ALT cancer cell lines suffering from inactivating mutations of the ATRX gene and loss of ATRX function in somatic cell hybrids segregated with the ALT pathway. Here, we will investigate the roles of ATRX and H3.3 functions in controlling DNA replication fidelity and repressive chromatin assembly at the telomeres. The study is carried out using mouse embryonic stem cells (ESCs) carrying conditional gene knockout of h3f3a and h3f3b (H3.3a and b genes). We show evidence that H3.3 is required for assembly heterochromatin enriched with H3K9me3 at telomeres. Presented by: Wong, Lee 201 Poster 130 A cryptic route to senescence reveals a constitutive protective function of telomerase Zhou Xua,*, Emilie Falleta, Camille Paolettib, Steffen Fehrmannb, Gilles Charvinb, Maria Teresa Teixeiraa a Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, Institut de Biologie PhysicoChimique, Centre National de la Recherche Scientifique, Université Pierre et Marie Curie, UMR8226, 13 rue Pierre et Marie Curie 75005 Paris, France b Institut de Génétique et de Biologie Moléculaire et Cellulaire, 1 rue Laurent Fries 67400 Illkirch Cedex, France In the absence of telomerase, eukaryotic cells still undergo divisions but eventually arrest. To study the dynamics of this transition in Saccharomyces cerevisiae, we used a microfluidics-based approach and tracked down individual cell lineages from telomerase inactivation to cell death. We found that most lineages are characterized by a sharp on/off transition from normal cell divisions to an arrested state. This is consistent with a single event controlling senescence onset when telomere(s) reach a critical length due to the end-replication problem. To our surprise, we also observed the existence of a cryptic noncanonical route leading to replicative senescence, which comprises frequent cell cycle delays early after telomerase inactivation, signalled through Mec1 and inconsistent with gradual telomere attrition. Rad51 is required in both types of lineages to maintain viability, in an age-independent manner. Conversely, we quantified the major contribution of telomerase to cell viability when homologous recombination is compromised, and showed a two-fold reduction in the mortality rate when telomerase is active. This suggests that in wild-type cells, telomeres are stochastically damaged with a measurable low frequency, regardless of the end-replication problem, and require telomerase and/or homologous recombination for repair. This work reveals a constitutive protective role for telomerase elongation activity, distinct from the rescue of progressively shortened telomeres. Funded by ERC-STG-2010 D-END Presented by: Xu, Zhou 202 Poster 131 TPP1 is a substrate for the deubiquitinating enzyme USP7 Ivo Zemp, Patrick Reichenbach, Joachim Lingner Swiss Institute for Experimental Cancer Research (ISREC), Ecole Polytechnique Fédérale de Lausanne (EPFL), Switzerland The shelterin component TPP1 has apparently opposing roles in telomerase regulation. On one hand, its depletion leads to telomere decapping and telomere extension, indicating that TPP1 is necessary for telomerase inhibition. On the other hand, TPP1, and more specifically the TEL patch in its OB domain, is required for efficient telomerase recruitment to telomeres and telomere elongation. How the opposing roles of TPP1 are regulated remains poorly understood. To gain further insights into the regulation of TPP1, we performed a yeast two-hybrid screen using the OB domain of TPP1 as a bait, and identified the deubiquitinating enzyme USP7 as an interacting protein of TPP1. We show that TPP1 is ubiquitinated at several residues in its OB fold and C-terminal region, and USP7 deubiquitinates TPP1 both in vivo and in vitro. We observe that TPP1 protein levels are regulated by the proteasome, but USP7 appears not to be involved in this process, raising the intriguing possibility that TPP1 ubiquitination and its deubiquitination by USP7 regulate TPP1 functions. We show that TPP1 ubiquitination is not required for its proper localization, since ubiquitinationdeficient mutants of TPP1 still interact with the shelterin components POT1 and TIN2, and co-localize with the telomeric marker TRF1. We will report on whether, in contrast, ubiquitination of TPP1 interferes with its binding to shelterin components or recruitment of telomerase. Presented by: Zemp, Ivo 203 Poster 132 SUMOylation of Tpz1 controls telomerase action in fission yeast Mansi Garg, Sahar Mansoubi, Resham Lal Gurung, Steve Plumb, Felicity Watts, Alessandro Bianchi Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK Previous work had indicated that SUMO controls telomerase action in fission yeast. In a systematic search for SUMOylated proteins in S. pombe, we have identified Tpz1 as a telomeric factor that is subject to modification by SUMO. We have identified a single lysine residue in Tpz1 that is targeted by the modification. Mutation of this lysine leads to telomere elongation in a telomerase-dependent manner (recombination is not involved). Genetic and phenotypic analysis indicates that Pli1 is the SUMO E3 ligase primarily active in the modification of Tpz1 and that this single modification on the Tpz1 protein is largely responsible for the effect of SUMO in the telomerase pathway in fission yeast. The telomere association of several telomeric proteins in only minimally affected in the SUMOylationdefective tpz1 mutant, with the notable exception of Stn1, which shows diminished telomere binding. Intriguingly, Stn1/Ten interact with a SUMO-Tpz1 fusion protein, and also with Tpz1 and SUMO independently, in two-hybrid assays. Our data suggest that SUMOylation down-regulates telomerase in fission yeast by promoting Stn1/Ten1 action at telomeres through Tpz1, likely through a direct interaction between SUMOylated Tpz1 and Stn1. These findings uncover an evolutionary conservation of the regulation of CST function by SUMOylation. Presented by: Bianchi, Alessandro 204 Participant List Eric Aeby Swiss Institute for Experimental Cancer Research (ISREC), Ecole Polytechnique Fédérale de Lausanne (EPFL) [email protected] Julio Aguado IFOM - the FIRC Institute of Molecular Oncology [email protected] Shawn Ahmed University of North Carolina [email protected] Patrizia Alberti Museum National d'Histoire Naturelle [email protected] Genevieve Almouzni Institut Curie [email protected] Sara Anjomani Virmouni Brunel University [email protected] Helene Antoine-Poirel Cliniques Universitaires Saint-Luc , de Duve Institute, UCL [email protected] Mary Armanios Johns Hopkins Univeristy School of Medicine [email protected] Nausica Arnoult The Salk Institute [email protected] Rajika Arora Institute of Biochemistry / ETH Zurich [email protected] Steven Artandi Stanford [email protected] Usha Aryal University of Leicester [email protected] Claus Azzalin ETHZ Zurich [email protected] Duncan Baird Cardiff University [email protected] Christian Bär CNIO [email protected] Aurelia Barascu UMR CNRS 8226 Institut de Biologie PhysicoChimique [email protected] Peter Baumann HHMI, Stowers Institute for Medical Research [email protected] Tara Beattie University of Calgary [email protected] Oliver Bechter UZ Leuven [email protected] Karen Beemon Johns Hopkins University [email protected] 205 Petra Bencurova Masaryk University [email protected] Patrick Brest IRCAN [email protected] Carlos Benitez Spanish National Cancer Research Center (CNIO) [email protected] Tracy Bryan Children's Medical Research Institute [email protected] Maname Benyelles ENS-Lyon, France [email protected] Stefano Cacchione Sapienza University of Rome, Dept of Biology & Biotechnology [email protected] Benjamin Best Life Extension Foundation [email protected] Alessandro Bianchi Genome Damage and Stability Centre, School of Life Sciences, University of Sussex [email protected] Elizabeth Blackburn University of California, San Francisco [email protected] Maria Blasco Spanish National Cancer Research Centre [email protected] Sophie Bombard INSERM UMR-S-1007, Universite Paris Descartes [email protected] Sumit Borah University of Colorado Boulder [email protected] Joanna Boros de Duve Institute / Catholic University of Louvain [email protected] Simon Boulton London Research Institute, Clare Hall Laboratories [email protected] Maria Isabel Cano Universidade Estadual Paulista Julio de Mesquita Filho, UNESP, Biosciences Institute, Genetics Dept. [email protected] Chris Caridi University of California, Riverside [email protected] Jaime Carrillo Garcia Instituto de Investigaciones Biomedicas "Alberto Sols" [email protected] Maria Antonietta Cerone UCL Cancer Institute [email protected] Weihang Chai Washington State University, USA [email protected] Alex Chang Stanford [email protected] Michael Chang University of Groningen [email protected] Razan Charif AP-HP [email protected] 206 Pascal Chartrand Universite de Montreal [email protected] Elisa Coluzzi University of Rome [email protected] Liuh-Yow Chen Institute of Molecular Biology / Academia Sinica [email protected] Dimitri Conomos Children's Medical Research Institute [email protected] Julian J.-L. Chen Arizona State University [email protected] Julia Cooper National Cancer Institute, NIH [email protected] Shantanu Chowdhury CSIR - Institute of Genomics and Integrative Biology [email protected] Sandro Cosconati Second University of Naples [email protected] Dmitri Churikov Marseille Cancer Research Center [email protected] Stephane Coulon CRCM [email protected] Alessandro Cicconi "Sapienza" University of Rome [email protected] Jannie De Jong European Research Institute for the Biology of Ageing / Laboratory of telomeres and genome integrity [email protected] Francesca Cipressa University of Rome 'La Sapienza' [email protected] Kate Clark Newcastle University [email protected] Veryan Codd University of Leicester [email protected] Marita Cohn Lund University / Department of Biology [email protected] Kathleen Collins UC Berkeley [email protected] Titia de Lange The Rockefeller Univeristy [email protected] Anita De Rossi University of Padova [email protected] Anabelle Decottignies de Duve Institute / Catholic University of Louvain [email protected] Katharina Deeg German Cancer Research Center (DKFZ) [email protected] Erin Degelman Univeristy of Calgary [email protected] 207 Jerome Dejardin IGH CNRS INSERM [email protected] Jiri Fajkus Masaryk University and CEITEC [email protected] Salvatore Di Maro University of Naples "Federico II" [email protected] Virginie Faure Albert Bonniot Institute - University of Grenoble [email protected] Aurelie Diman de Duve Institute / Catholic University of Louvain [email protected] Miguel Ferreira Instituto Gulbenkian de Ciencia [email protected] Irena Draskovic Institut Curie [email protected] Ignacio Flores CNIC [email protected] Rachid Drissi Cincinnati Children's Hospital Medical Center [email protected] Sebastian Foersch Leibniz Institute for Age Research [email protected] Yimin Duan Shanghai Institute of Biochemistry and Cell Biology [email protected] Miloslava Fojtova Masaryk University [email protected] Katerina Duskova University of Alcala [email protected] Adam Freund Stanford [email protected] Charlene Emerson Baylor College of Medicine [email protected] Jana Fulneckova CEITEC MU & Inst. Of Biophysics, ASCR [email protected] Harikleia Episkopou de Duve Institute / Catholic University of Louvain [email protected] Sarantis Gagos Biomedical Research Foundation of the Academy of Athens Greece (BRFAA) [email protected] Jose Escandell Planells Instituto Gulbenkian de Ciencia [email protected] Laura Esteban Lafuente University of Alcala [email protected] Roberto Galletto Washington University School of Medicine [email protected] Maria Garcia Beccaria Spanish National Cancer Research Center (CNIO) [email protected] 208 Inbal Gazy Tel Aviv University, Israel [email protected] Adam Harvey University of Minnesota [email protected] Vincent Geli CRCM Marseille [email protected] Makoto Hayashi The Salk Institute [email protected] Eric Gilson IRCAN [email protected] Sandra Henkelman ERIBA [email protected] Marie-Josephe Giraud-Panis IRCAN [email protected] Catarina Henriques Instituto Gulbenkian de Ciencia [email protected] Galina Glousker The Hebrew University of Jerusalem [email protected] Milan Hluchy Masaryk University [email protected] Daniel Gomez Universidad Nacional de Quilmes [email protected] Dirk Hockemeyer UC Berkeley [email protected] Marco Graf ZMBH [email protected] Ctirad Hofr CEITEC - Masaryk University [email protected] Roger Greenberg University of Pennsylvania [email protected] Klaus Holzmann Institute of Cancer Research, Comprehensive Cancer Center, Medical University Vienna, Austria [email protected] Fernanda Gutierrez University of Sao Paulo [email protected] David Halvorsen SENS Research Foundation [email protected] Lea Harrington Institute for Research in Immunology and Cancer/Univ Montreal [email protected] Juyeong Hong Department of Integrated Omics for Biomedical Science / Yonsei University [email protected] Jacqueline Jacobs The Netherlands Cancer Institute [email protected] Eliska Janouskova Masaryk University [email protected] 209 Clare Jelinska Nanyang Technological University [email protected] Patrycja Klos Masaryk Univeristy [email protected] Kristi Jensen Stowers Institute for Medical Research [email protected] Elzbieta Kowalska Max F. Perutz Laboratories (MFPL) [email protected] Jana Jureckova Masaryk University, Faculty of Science [email protected] Theresa Kreilmeier Department for Companion Animals and Horses, University of Veterinary Medicine Vienna, Veterinarplatz 1, 1210 Vienna, Austria [email protected] Junko Kanoh Institute for Protein Research, Osaka University [email protected] Ahu Karademir Lund University / Department of Biology [email protected] Jan Karlseder The Salk Institute for Biological Studies [email protected] Galina Kartasheva LLC "Telomerase Activation Sciences" [email protected] Christopher Kasbek University of Cincinnati [email protected] Guillaume Kellermann Universite Paris Descartes [email protected] Chuna Kim Seoul National University [email protected] Antonis Kirmizis University of Cyprus [email protected] Martin Kupiec Tel Aviv University, Israel [email protected] Mattia la Torre Sapienza University of Rome [email protected] Peter Lansdorp European Research Institute for the Biology of Ageing [email protected] Eros Lazzerini Denchi The Scripps Research Institute [email protected] Junho Lee Seoul National University [email protected] Jihoon Lee Department of Integrated Omics for Biomedical Science / Yonsei University [email protected] Ming Lei National Center for Protein Science, Shanghai, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences [email protected] 210 Paul Lieberman The Wistar Institute [email protected] Isabella Marcomini Friedrich Miescher Institute [email protected] Joachim Lingner EPFL [email protected] Dries Martens Universiteit Hasselt [email protected] Natalia Lipinska Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences [email protected] Paula Martinez CNIO [email protected] Arturo Londono-Vallejo Institut Curie [email protected] Isabel Lopez de Silanes Spanish Cancer Research Centre [email protected] Brian Luke University of Heidelberg, ZMBH [email protected] Vicki Lundblad The Salk Institute for Biological Studies [email protected] Arthur Lustig Tulane University [email protected] Paulina Marzec Institute of Human Genetics (IGH), CNRS [email protected] Aaron Mendez Bermudez IRCAN [email protected] Javier Miralles Fuste The Salk Institute [email protected] Beth Moorefield Nature Structural & Molecular Biology [email protected] Foteini Mourkioti Stanford University [email protected] David Lydall Newcastle University [email protected] Thanchanok Muangman Thailand Institute of Scientific and Technological Research [email protected] Eva Majerova Masaryk University, Brno, CZE [email protected] Veronika Muchova CEITEC MU [email protected] Jana Majerska EPFL SV ISREC [email protected] Aysel Muezzinler German Cancer Research Center (DKFZ) [email protected] 211 Toru Nakamura University of Illinois at Chicago [email protected] Ozgun Ozer University of Copenhagen [email protected] Christine Napier Children's Medical Research Institute [email protected] Raquel Paiva University of Sao Paulo [email protected] Saishyam Narayanan Lund University / Department of Biology [email protected] Lili Pan Stowers Institute for Medical Research [email protected] Ivona Necasova CEITEC MU [email protected] Stephanie Panier London Research Institute [email protected] Nya Nelson Baylor College of Medicine [email protected] Omesha Perera Children's Medical Research Institute [email protected] Bernadette Nera UC Davis [email protected] Vratislav Peska CEITEC - Masaryk University [email protected] Drew Niles 10 - Promega Madison [email protected] Verena Pfeiffer EPFL [email protected] Valeria Nofrini University of Perugia [email protected] Hilda Pickett Children's Medical Research Institute [email protected] Konrad Ocalewicz Univeristy of Warmia and Mazury in Olsztyn, Poland [email protected] Brandon Pierce University of Chicago [email protected] Galina Orlova LLC "Telomerase Activation Sciences" [email protected] Nicky Pieters Hasselt University [email protected] Jessica Owens Stowers Institute for Medical Research [email protected] Ines Pimenta de Castro Instituto Gulbenkian de Ciencia [email protected] 212 Catherine Polese Laboratory of Protein Signaling and Interactions, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liege [email protected] Delphine Poncet Claude Bernard University Lyon1 [email protected] Rosa Maria Porreca Institut Curie [email protected] Florian Poulain de Duve Institute / Catholic University of Louvain [email protected] Carolyn Price University of Cincinnati [email protected] Petra Prochazkova Schrumpfova Masaryk University and CEITEC [email protected] Margaret Pruitt Stowers Institute for Medical Research [email protected] Robert Radford The Salk Institute [email protected] Grazia Daniela Raffa Sapienza Universita di Roma [email protected] Roger Reddel Children's Medical Research Institute [email protected] Sophie Redon ISREC / EPFL [email protected] Patrick Revy Imagine Institute [email protected] Daniela Rhodes Nanyang Technological University [email protected] Yikang Rong National Institute of Health [email protected] Francesca Rossiello IFOM - the FIRC Institute of Molecular Oncology [email protected] Karl Lenhard Rudolph Leibinz Institute for Age Research/Fritz-Lipman Institute Jena [email protected] Flavia Sacilotto Donaires University of Sao Paulo [email protected] Mahito Sadaie Kyoto University [email protected] Isabella Saggio Sapienza University of Rome [email protected] Shira Sagie Technion [email protected] Carole Saintome Museum National d'Histoire Naturelle [email protected] Twana Salih University of Nottingham [email protected] 213 Erica Salvati Regina Elena National Cancer Institute [email protected] Antonella Sgura University of Rome [email protected] Sandra Sampl Institut for Cancer Research / Department of Medicine I / Medical University of Vienna [email protected] Yusuke Shima Kyoto University [email protected] Ranjodh Singh Sandhu Cleveland State University [email protected] Grzegorz Sarek London Research Institute, Cancer Research UK [email protected] Sharon Savage National Cancer Institute, NIH [email protected] Katri Savolainen University of Helsinki [email protected] Jens Schmidt University of Colorado Boulder [email protected] Sara Selig Rambam Health Care Campus and Rappaport Faculty of Medicine and Research Institute, Technion [email protected] Beomseok Seo Seoul National University [email protected] David Shore University of Geneva [email protected] Stefan Sigurdsson University of Iceland [email protected] Haroldo Silva SENS Research Foundation [email protected] Susan Smith The Skirball Institute NYU SOM [email protected] Paula Soares IPATIMUP [email protected] Zhou Songyang Baylor College of Medicine [email protected] Tobias Sperka Leibniz Institute for Age Research - Fritz Lipmann Institute [email protected] Kamar al Zaman Serhal Institut de Biologie Physico - Chimique / UMR 8226 [email protected] Sonia Stinus ERIBA - European Research Institute for the Biology of Ageing [email protected] Agnel Sfeir NYU/Skirball Institute [email protected] Katerina Strati University of Cyprus [email protected] 214 Estelle Suaud Active Motif [email protected] Jean-Baptiste Vannier Cancer Research UK [email protected] Sanghyun Sung Seoul National University [email protected] Hartmut Vodermaier The EMBO Journal [email protected] Maria Teresa Teixeira Laboratoire de Biologie Moleculaire et Cellulaire des Eucaryotes, Institut de Biologie Physico-Chimique, UMR8226 CNRS/UPMC [email protected] Leonid Vorslov LLC "Telomerase Activation Sciences" [email protected] Nico Thoma Friedrich Miescher Institute for Biomedical Research [email protected] Kazunori Tomita University College London [email protected] Adrian Tong Children's Medical Research Institute [email protected] Anela Tosevska University of Vienna [email protected] Yehuda (Dudy) Tzfati The Hebrew University of Jerusalem [email protected] Evert-Jan Uringa European Research Institute for the Biology of Ageing, University Medical Center Groningen [email protected] Yabin Wei Karolinska Institutet [email protected] Ted Weinert University of Arizona [email protected] Mundy Wellinger Universite de Sherbrooke [email protected] Brigitte Wevers The Netherlands Cancer Institute [email protected] Michael Wildauer University of Vienna, Department of Biochemistry and Cell Biology [email protected] Lee Wong Monash University [email protected] Woodring Wright UT Soutwestern Med Ctr [email protected] Sona Valuchova Gregor Mendel Institute of Molecular Plant Biology GmbH [email protected] 215 Zhou Xu Laboratoire de Biologie Moleculaire et Cellulaire des Eucaryotes, Institut de Biologie PhysicoChimique, Centre National de la Recherche Scientifique, Universite Pierre et Marie Curie UMR8226 [email protected] Lifeng Xu UC Davis [email protected] Ivo Zemp Institute for Experimental Cancer Research, EPF Lausanne [email protected] 216 Reference Maps Brussels North Train Station to Husa President Park 217 Husa President Park to Grand-Place 218 Grand-Place to La Manufacture 219 La Manufacture to Husa President Park 220 Brussels’ Metro 221 Brussels, General map 207