248
 SCIENTIFIC REPORT 2013 SPANISH NATIONAL CANCER RESE ARCH CENTRE, CNIO

INFORME CNIO 3013

Embed Size (px)

DESCRIPTION

actividades del cnio durante 2013

Citation preview

  • 5/19/2018 INFORME CNIO 3013

    1/248

    SCIENTIFICREPORT 2013

    SPANISH NATIONAL CANCER RESE ARCH CENTRE, CNIO

  • 5/19/2018 INFORME CNIO 3013

    2/248

  • 5/19/2018 INFORME CNIO 3013

    3/248

    SCIENTIFICREPORT 2013

  • 5/19/2018 INFORME CNIO 3013

    4/248

  • 5/19/2018 INFORME CNIO 3013

    5/248

    SCIENTIFICREPORT 2013

  • 5/19/2018 INFORME CNIO 3013

    6/248

    CONTENTS

    ORGANISATIONOF RESEARCH

    FOREWORD 6

    VICE-DIRECTIONOF BASIC RESEARCH

    14

    12

    Molecular Oncology Programme 18

    Tumour Suppression Group 20

    Experimental Oncology Group 24

    Telomeres and Telomerase Group 26

    Cell Division and Cancer Group 30

    Genomic Instability Group 34Chromosome Dynamics Group 38

    DNA Replication Group 42

    BBVA Foundation-CNIO Cancer Cell Biology Programme 46

    Genes, Development and Disease Group 48

    Epithelial Cell Biology Junior Group 52

    Growth Factors, Nutrients and Cancer Junior Group 54

    Seve Ballesteros Foundation-CNIO Brain Tumour Junior Group 56

    Structural Biology and Biocomputing Programme 58

    Structural Computational Biology Group 60

    Macromolecular Crystallography Group 64

    Computational Biophysics Junior Group 68

    Cell Signalling and Adhesion Junior Group 70

    Structural Bases of Genome Integrity Junior Group 72

    Spectroscopy and Nuclear Magnetic Resonance Core Unit 74

    Bioinformatics Core Unit 76

    National Bioinformatics Institute Core Unit 78

    Molecular Pathology Programme 84

    Melanoma Group 86

    Epithelial Carcinogenesis Group 90

    Stem Cells and Cancer Group 94

    Tumour Markers Junior Group 98

    Human Cancer Genetics Programme 100

    Human Genetics Group 102

    Molecular Cytogenetics Group 106

    Hereditary Endocrine Cancer Group 1 10

    Genetic and Molecular Epidemiology Group 11 4Familial Cancer Clinical Unit 1 18

    Human Genotyping-CEGEN Core Unit 120

    Clinical Research Programme 122

    Gastrointestinal Cancer Clinical Research Unit 124

    Breast Cancer Junior Clinical Research Unit 128

    CRIS Foundation-CNIO Prostate Cancer and Genitourinary

    Tumours Junior Clinical Research Unit 130

    Molecular Diagnostics Unit 132

    Translational Bioinformatics Unit 134

    Biobank 136

    VICE-DIRECTION OFTRANSLATIONAL RESEARCH

    80

    SCIENTIFIC REPORT 2013 4

  • 5/19/2018 INFORME CNIO 3013

    7/248

    COMMUNICATION 180

    Deans Ofce 192

    Women in Science Ofce 194

    CNIO OFFICES 190

    Competitive Funding 198

    Education and Training Programmes 210

    Scientic Events 216

    Administration 232

    Board of Trustees 232

    Scientic Advisory Board 234

    Management 236

    CNIO Personnel 2013 238

    FACTS & FIGURES 196

    CREATIVE TEAM 242

    Biotechnology Programme 142

    Genomics Core Unit 144

    Transgenic Mice Core Unit 146

    Monoclonal Antibodies Core Unit 148

    Histopathology Core Unit 150

    Molecular Imaging Core Unit 152

    Flow Cytometry Core Unit 154

    Confocal Microscopy Core Unit 156

    Proteomics Core Unit 158

    Animal Facility 160

    Experimental Therapeutics Programme 162Medicinal Chemistry Section 164

    Biology Section 168

    CNIO-Lilly Cell Signalling Therapies Section 172

    CNIO-Lilly Epigenetics Section 174

    Technology Transfer and Valorisation Ofce 176

    Private Sponsors 178

    DIRECTION OF INNOVATION 138

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 5

  • 5/19/2018 INFORME CNIO 3013

    8/248

    SCIENTIFIC REPORT 2013 6

  • 5/19/2018 INFORME CNIO 3013

    9/248

    2013 has been the

    most productive yearin more than 10 yearsof CNIOs history,both in terms of

    scientic discoveriesand innovationachievements. Thissuccess is to be credited

    to CNIO scientists.MARIA A. BLASCODirector

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 7

  • 5/19/2018 INFORME CNIO 3013

    10/248

  • 5/19/2018 INFORME CNIO 3013

    11/248

    First of all, I would like to take this opportunity to thank all of

    those who have once again collaborated in the elaboration of thisyears Annual Report. My special thanks goes out to the ScientificManagement team that was heavily involved in its production,and in particular to Ana Merino and Sonia Cerd, who once moredemonstrated their efficiency and professionalism. I also wish tothank our external collaborators, Amparo Garrido (photography)and Underbau (design team).

    I am very glad to start by saying that 2013 has been the mostproductive year in more than 10 years of CNIOs history, bothin terms of scientific discoveries and innovation achievements.This success is to be credited to CNIO scientists; this year theirscientific performance has excelled to an even greater extent.

    In 2013, the CNIO published a total of 229 papers, 55 of whichwere published in journals with impact factors in the range of10 to 15 and >15. This represents a 22% increase in high impactfactor publications compared to 2012. This excellent performanceconsolidates our impressive output of scientific publications intop journals since 2010.

    We are particularly proud of the groundbreaking discovery madeby CNIO researchers from the Tumour Suppression Group, led byManuel Serrano, who achieved in vivogeneration of pluripotentstem cells; this important step forward in the field of regenerative

    medicine was published inNatureand was considered as one ofthe notable advances of 2013 byNature Medicine. SerranosGroup deserves double congratulations, seeing as they alsoparticipated in the discovery of a role for cellular senescence inembryonic development, where, together with apoptosis, it playsan essential role in final organ design and functionality. This isa key discovery for the cellular senescence field; the study waspublished in Cell.

    Maria A. Blasco DirectorFOREWORD

    FOREWORD

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 9

  • 5/19/2018 INFORME CNIO 3013

    12/248

    Erwin Wagner s Group alone published several papers i n avariet y of top journals (Cell Metabolism,Immunity, Genes &Dev, among others), strengthening our understanding on therole of inflammation in several types of cancer. CNIO scientists

    also made significant contributions in high-throughput cancergenome analysis through landmark papers published by theCNIO Groups led by Francisco Real on bladder cancer and JavierBenitez on breast and ovarian cancer; both were published in

    Nature Genetics.

    2013 has also been outstanding in terms of CNIO translationalresearch activities. In addition to consolidating the in-houseClinical Research Units of the Clinical Cancer ResearchProgramme, directed by Manuel Hidalgo, the CNIO has signedagreements to create new Associated Clinical Units in twoHospitals of the Regional Government of Madrid, including

    the Paediatric HospitalHospital Nio Jessand theFundacinJimnez-Daz. We are particularly proud of launching an earlyphase Paediatric Clinical Trial Unit at theHospital Nio Jess.Paediatric Clinical Trials Units are scarce in Spain and the one ledby CNIO oncologists will be the first early phase trial in Madrid.Moreover, CNIO oncologists are coordinating two large, country-

    wide, Clinical Trials Networks in Breast and Prostate Cancer.

    In addition to the Clinical Research Programme, the CNIOperforms important clinical activity through the Familiar CancerUnit located at theHospital Universitario de Fuenlabrada, as wellas through the Molecular Diagnostics Unit and the MolecularCytogenetics Group at CNIO; we provided genetic counselling

    to more than 160 patients and performed almost 1000 geneticdeterminations.

    During 2013, the Experimental Therapeutics Programme (ETP)worked closely together with our researchers in order to validatenew therapeutic targets. The Programmes current projects haveattracted the interest of several companies who are interestedin developing them into clinical candidates. 2013 proved to bean exceptional year thanks to our closing two very importantlicensing deals; namely, the licensing of the PIM inhibitor seriesto the Irish drug development company,Inflection Biosciences inMarch 2013, and the licensing of CNIO ATR inhibitors to Merck

    Serono in December 2013. For the first time, two very excitingprojects headed by the Experimental Therapeutics Programme,in collaboration with CNIO scientists, have been partnered withindustry for further development and commercialisation. We arehopeful that our joining forces with the pharmaceutical industry

    will allow us to take full advantage of our ability to find the nextgeneration of breakthrough therapies to fight cancer. The finalproof-of-concept for this Program me will emerge once one ofour molecules enters a clinical trial. We believe that we are onthe right track!

    Research Collaborations with industrial partners have also beenreinforced in 2013. Significantly, CNIO extended a long-standing

    FOREWORD

    SCIENTIFIC REPORT 2013 10

    partnership with the pharmaceutical company Eli Lilly in orderto establish a new section in the field of cancer epigenetics.Furthermore, during 2013 two new research projects were grantedfunding under the Extended Innovation Network of Roche, which

    CNIO joined in 2012. This makes a total of three highly innovativeand translational projects that are being funded at CNIO underthe umbrella agreement with Roche. Our objective is to worktogether with industry to translate research results obtained atthe CNIO into innovative ideas and products to improve diagnosis,prevention and treatment in the field of cancer.

    It is my pleasure to announce that in 2013, Alfonso Valencia,who is the Director of the Structural Biology and BiocomputingProgramme at the CNIO, was also appointed Vice-Director ofBasic Research, and since then he has been helping the CNIODirection in key managerial issues. I want to take this opportunity

    to wholeheartedly thank the former Vice-Director of BasicResearch, Erwin Wagner, for his dedication to the CNIO andhis enthusiastic efforts to make the CNIO a truly internationalCentre. Erwin will continue with this important task as Directorof the BBVA Foundation-CNIO Cancer Cell Biology Programme.

    In February 2013, the new CNIO Scientific Advisory Boardundertook a site visit to CNIO in order to conduct a globalevaluation of its activities, as well as carry out the 5 Year Evaluationof the BBVA Foundation-CNIO Cancer Cell Biology and ClinicalResearch Programmes. During this evaluation, the Groups ledby Erwin Wagner, Christopher Heeschen and Manuel Hidalgo,

    were very positively evaluated. Two Junior Group Leaders of

    the Cancer Cell Biology Programme, Mirna Prez-Moreno andNabil Djouder, were also positively evaluated and their JuniorPosition at CNIO was extended for an additional three years.Congratulations Mirna and Nabil!

    During 2013, two Junior Groups left the CNIO to take tenuredpositions at different institutions. On one hand, Marta Snchez-Carbayo, who was with us for more than six years as Head of theTumour Markers Group at the Molecular Pathology Programme,accepted a full professor position at the prestigious CIC-BIOGUNE; a multidisciplinary research centre in the BasqueCountry, Spain. We wish Marta lots of success in the beautiful

    city of Bilbao. Francesco Gervasio, a brilliant researcher in thefield of computer biology, started at the CNIO as a Junior GroupLeader three years ago, and was offered a full professorship atThe University College of London. In spite of the fact that Martaand Francesco are no longer at CNIO, we maintain numerouscollaborations with them.

    In 2013, we celebrated the CNIOs 10thinauguration anniversaryduring the thirdNature-CNIO Cancer Symposium. On thisoccasion the subject was Tumour Heterogeneity and Plasticity.Thanks again to all theNatureeditors who were involved in theevent! A special thank you goes out to Barbara Marte, NicolaMcCarthy and Alexia-Ileana Zaromytidou, as well as to our

  • 5/19/2018 INFORME CNIO 3013

    13/248

    in-house organisers, Erwin Wagner and Mirna Prez-Moreno, andto our external organiser, Scott Lowe, for the exciting conferencethat they managed to put together.

    I would like to take this opportunity to thank those who sponsoredour students, postdoctoral programmes, and the stays of severalresearchers. I hereby thankBanco SantanderFoundation forfunding postdoctoral stays at CNIO; Obra Social la CaixaFoundation for fostering international PhD fellowships; SeveBallesteros Foundation that supports the Seve-BallesterosFoundation-CNIO Brain Tumour Group; and the Jesus SerraFoundation for supporting the Visiting Scientists Programme.During 2013, Robert Benezra from the Memorial Sloan-KetteringCancer Center in New York, was beneficiary of the Jess SerraFoundations Visiting Researcher Programme. I am also grateful

    to the Foundation Banc Sabadell for sponsoring conferences givenby out-ofthe box spea kers who provided novel perspectivesthat contribute to the CNIOs trans-disciplinary environment.During 2013, we had the privilege of listening to: Pedro Alonso,one of the worlds foremost global authorities on malaria research;Carl Djerassi, chemist and novelist but better known for hiscontribution to the development of the contraceptive pill; J.L.

    Arsuaga, a leader in human palaeontology and member of theresearch team of Atapuerca Pleistocene deposits, and finallyJose Mara Ordovs, globally known specialist in nutrition,nutrigenetics and nutrigenomics. In addition, I extend mythanks toFundacin BBVAthat generously supports the BBVA

    Foundation-CNIO Cancer Cell Biology Programme, as well as tothe CRISFoundation against Cancer, which supports the CRISFoundation-CNIO Prostate Cancer and Genitourinary TumoursClinical Research Unit at CNIO. Further words of thanks are dueto AVON, which funds the Breast Cancer Clinical Research Unit,and to the Botn Foundation for supporting the Telomeres andTelomerase Group, the Tumour Suppression Group and from2014 onwards also the Genomic Instability Group; all of themlocated under the Molecular Oncology Programme.

    I would also like to make a special mention of the success achievedby the Communications Department in 2013. After two years ofhard work since its creation, the department has succeeded in

    FOREWORD

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 11

    increasing CNIOs press visibility by 75% compared with 2012.One of the top stories this year was undoubtedly the study on invivogeneration of pluripotent stem cells published in the journalNature. All of the ma in Spanish media outlets reported the

    discovery, including print media, TV and radio. The discoverywas also picked up by foreign media outlets around the world,including the BBC, The Wall Street Journal, T he FinancialTimes,Le Mondeand The Telegraph, amongst many others.

    Another example that I would like to highlight is the CNIO andMerck license agreement, which was deemed a success a ndreflected as such by national and international media, includingthe prominent Wall Street Journal. These listed examples are

    just a small selection amongst several, which stand testament toour motivation and commitment to society in terms of sharingthe latest breakthroughs, as well as CNIOs capacity to translateknowledge into applications for the benefit of cancer patients.

    As well as increasing our presence in traditional media, CNIOhas also increased visibility on social media networks, such asTwitter and YouTube. During 2013, CNIOs press releases inthe global news service EurekAlert! received over 70,000 views.

    Finally, I want to whole-heartedly thank all CNIO employeesfor their professionalism during 2013. In a year of restrictions,it became more evident than ever before that we all considerourselves to be an intrinsic part of the CNIO, and that we allcontinue to work closely together to ensure that the CNIO is oneof the best scientific institutions in Europe. s

  • 5/19/2018 INFORME CNIO 3013

    14/248

    SCIENTIFIC REPORT 2013 12

    MOLECULAR ONCOLOGY

    PROGRAMME

    ALFONSO VALENCIA VICE-DIRECTOR OF BASIC RESEARCH

    MARIA A. BLASCO DIRECTOR

    Manuel Serrano Programme Director

    Manuel Serrano

    Tumour Suppression GroupMariano BarbacidExperimental Oncology Group

    Maria A. BlascoTelomeres and Telomerase Group

    Marcos MalumbresCell Division and Cancer Group

    scar Fernndez-Capetillo

    Genomic Instability GroupAna Losada

    Chromosome Dynamics Group

    Juan MndezDNA Replication Group

    BBVA FOUNDATION-CNIO

    CANCER CELL BIOLOGY

    PROGRAMME

    Erwin F. Wagner Programme Director

    Erwin F. WagnerGenes, Development and Disease Group

    Mirna Prez-MorenoEpithelial Cell Biology Junior Group

    Nabil DjouderGrowth Factors, Nutrients and

    Cancer Junior GroupMassimo SquatritoSeve Ballesteros Foundation-CNIOBrain Tumour Junior Group

    STRUCTURAL BIOLOGY

    AND BIOCOMPUTING

    PROGRAMME

    Alfonso Valencia Programme Director

    Alfonso ValenciaStructural Computational Biology Group

    Guillermo MontoyaMacromolecular Crystallography Group

    Francesco L. Gervasio (until June)

    Computational Biophysics Junior GroupDaniel LiethaCell Signalling and Adhesion Junior Group

    Santiago Ramn-MaiquesStructural Bases of GenomeIntegrity Junior Group

    Ramn Campos-OlivasSpectroscopy and Nuclear MagneticResonance Core Unit

    David G. PisanoBioinformatics Core Unit

    Alfonso ValenciaNational Bioinformatics Institute Core Unit

    MOLECULAR PATHOLOGY

    PROGRAMME

    MANUEL HIDALGO VICE-DIRECTOR OF TRANSLATIONAL RESEARCH

    Mara S. Soengas Programme Director

    Mara S. SoengasMelanoma Group

    Francisco X. RealEpithelial Carcinogenesis Group

    Christopher HeeschenStem Cells and Cancer Group

    Marta Snchez-Carbayo (until May)Tumour Markers Junior Group

    ORGANISATION OFRESEARCH

  • 5/19/2018 INFORME CNIO 3013

    15/248

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO

    13

    CLINICAL RESEARCH PROGRAMME

    EXPERIMENTAL THERAPEUTICS

    PROGRAMME

    BIOBANK

    Manuel Hidalgo Programme Director

    Joaqun Pastor Programme Director

    Manuel M. Morente Director

    Manuel HidalgoGastrointestinal Cancer

    Clinical Research Unit

    Miguel Quintela-FandinoBreast Cancer Junior Clinical Research Unit

    David OlmosCRISFoundation-CNIO Prostate

    Cancer and Genitourinary Tumours

    Junior Clinical Research Unit

    Sonia MartnezMedicinal Chemistry Section

    Carmen BlancoBiology Section

    Luis J. LombardaMolecular Diagnostics Unit

    Ftima Al-ShahrourTranslational Bioinformatics Unit

    Susana VelascoCNIO-Lilly Cell Signalling Therapies Section

    Maria Jos Barrero (since July)CNIO-Lilly Epigenetics Section

    BIOTECHNOLOGY PROGRAMME

    MARISOL QUINTERO (until October) DIRECTOR OF INNOVATION

    Fernando Pelez Programme Director

    Orlando DomnguezGenomics Core Unit

    Sagrario OrtegaTransgenic Mice Core Unit

    Giovanna RoncadorMonoclonal Antibodies Core Unit

    Marta Caamero (until June)Histopathology Core Unit

    Francisca MuleroMolecular Imaging Core Unit

    Lola MartnezFlow Cytometry Core Unit

    Diego MegasConfocal Microscopy Core Unit

    Javier MuozProteomics Core Unit

    Isabel Blanco (Charles River Laboratories)Animal Facility

    HUMAN CANCER GENETICS

    PROGRAMME

    Javier Bentez Programme Director

    Javier BentezHuman Genetics Group

    Juan C. CigudosaMolecular Cytogenetics Group

    Mercedes RobledoHereditary Endocrine Cancer Group

    Nria MalatsGenetic and Molecular Epidemiology Group

    Miguel UriosteFamilial Cancer Clinical Unit

    Anna Gonzlez-NeiraHuman Genotyping-CEGENCore Unit

    TECHNOLOGY TRANSFER

    AND VALORISATION OFFICE

    Marisol Quintero Head of Office(until October)

    Anabel Sanz Head of Office (since October)

  • 5/19/2018 INFORME CNIO 3013

    16/248

    SCIENTIFIC REPORT 2013 14

    Vice-Directionof BasicResearch

  • 5/19/2018 INFORME CNIO 3013

    17/248

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 15

    Molecular Oncology Programme 18Tumour Suppression Group 20

    Experimental Oncology Group 24

    Telomeres and Telomerase Group 26

    Cell Division and Cancer Group 30Genomic Instability Group 34

    Chromosome Dynamics Group 38

    DNA Replication Group 42

    BBVA Foundation-CNIO Cancer Cell Biology Programme 46Genes, Development and Disease Group 48

    Epithelial Cell Biology Junior Group 52

    Growth Factors, Nutrients and Cancer Junior Group 54

    Seve Ballesteros Foundation-CNIO Brain Tumour Junior Group 56

    Structural Biology and Biocomputing Programme 58Structural Computational Biology Group 60

    Macromolecular Crystallography Group 64Computational Biophysics Junior Group 68

    Cell Signalling and Adhesion Junior Group 70

    Structural Bases of Genome Integrity Junior Group 72

    Spectroscopy and Nuclear Magnetic Resonance Core Unit 74

    Bioinformatics Core Unit 76

    National Bioinformatics Institute Core Unit 78

  • 5/19/2018 INFORME CNIO 3013

    18/248

    ALFONSO VALENCIAVice-Director of Basic Research

    SCIENTIFIC REPORT 2013 16

  • 5/19/2018 INFORME CNIO 3013

    19/248

    My main mission, as

    Vice-Director for BasicResearch, is to worktogether with CNIOsBasic Research Groups

    to enhance scienticexcellence, fostercollaboration andoptimise the use of our

    resources.

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 17

  • 5/19/2018 INFORME CNIO 3013

    20/248

    MANUEL SERRANO Programme Director

    MOLECULARONCOLOGY PROGRAMME

    VICE-DIRECTION OF BASIC RESEARCH

    SCIENTIFIC REPORT 2013 18

  • 5/19/2018 INFORME CNIO 3013

    21/248

    The Molecular OncologyProgramme is CNIO sagship Programme. Partof our success lies in ourcollective goal to make ourresearch more innovativeand efcient. Many of the

    discoveries made in ourProgramme are already intranslational phases and are,thus, contributing towardsthe improvement of cancertreatment.

    It may sound obvious, but it is always good to remember thatthe so-called translational or applied research necessarilyrequires basic science to be translated or applied. This isprecisely the mission of the Molecular Oncology Programme:to generate knowledge that can be translated into better carefor cancer patients.

    This year, the Molecular Oncology Programme has continued tobe on the frontline of oncology research. We were particularly

    proud this year because the Genomic Instability Group, incollaboration with the Experimental Therapeutics Programme,have generated extremely promising chemotherapeuticcompounds that inhibit the DNA damage signalling kinaseATR, having signed an ambitious licensing agreement withthe German pharmaceutical giant Merck Serono. Thanks tothis agreement, these compounds will be tested in clinicalassays, most likely with the participation of the CNIO ClinicalResearch Programme. This is a prime example of the greatsynergies that the CNIO can create, bringing together basicscientists, medical chemists and clinical oncologists. This isan ongoing story of great success and a motivation for all ofus to continue along this road of progress.

    I want to mention two other examples of research projects ledby scientists of the Molecular Oncology Programme that furtherillustrate our ability to catalyse creative collaborations. Thefirst one involves the collaboration of the CNIO Telomeres andTelomerase Group with the Spanish Project of the InternationalCancer Genome Consortium (ICGC), which resulted in thediscovery of a novel type of oncogenic mutation that affects thestability of chromosomes in chronic lymphocytic leukaemia(CLL). The second example is a multi-partnered collaborationbetween the CNIO Tumour Suppression Group, almost allthe Core Units at the CNIO, and a research group from the

    CNIC. This remarkable collaborative effort demonstrated theinduction of pluripotency in vivo; the significance of this workwas recognised byNature Medicine as the most importantdiscovery of the year 2013 in the field of stem cells. s

    MOLECULAR ONCOLOGY PROGRAMME

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 19

  • 5/19/2018 INFORME CNIO 3013

    22/248

    Adelaida Palla VENLluc Mosteiro ESP

    Elena Lpez-Guadamillas ESPDaniela Piazzolla ITADaniel Muoz ESPCian J. Lynch IRL

    Pablo J. Fernndez-Marcos ESPTimothy Cash USAMara Abad ESPCristina Pantoja ESP

    Antonio Maraver ESPSusana Llanos ESPHan Li CHNManuel Serrano ESP

    Staff Scientists

    Luis Enrique Donate (until February),Han Li, Susana Llanos, Antonio Maraver,Cristina Pantoja

    Manuel SerranoGroup LeaderTUMOUR SUPPRESSION

    GROUP

    VICE-DIRECTION OF BASIC RESEARCH

    SCIENTIFIC REPORT 2013 20

  • 5/19/2018 INFORME CNIO 3013

    23/248

    MOLECULAR ONCOLOGY PROGRAMME | TUMOUR SUPPRESSION GROUP

    OVERVIEW

    To characterise cellular senescence as a tumoursuppression mechanism.

    To investigate cellular pluripotency and the involvementof tumour suppressors in the regulation of reprogrammingto induced pluripotent stem (iPS) cells.

    Tumour suppressors are genes that can prevent the developmentof cancer. All our cells have a functional set of these genes.However, despite their efficient protection against cancer, thesegenes can become defective over time. The affected cells thusbecome partially unprotected from cancer and, in combination

    with additional mutations in other genes, can give rise to thedevelopment of cancer.

    Understanding how tumour suppressor genes work may help usto design drugs that block cancer. Our Group also manipulatesthe mouse genome to create novel alterations that increaseor decrease tumour suppression potency.

    The goals of our Group are:

    To understand the mechanisms of tumour suppressionand identify new tumour suppressor regulators.

    To study the interplay between tumour suppression and

    ageing. To analyse the involvement of tumour suppressors in the

    regulation of metabolism and protection from metabolicdamage.

    Graduate Students

    Katharina Hess (until May), ElenaLpez-Guadamillas, Luca Morgado,

    Lluc Mosteiro, Adelaida Palla

    Muoz, Sandrina Nbrega (until July),Daniela Piazzolla

    Post-Doctoral Fellows

    Mara Abad, Timothy Cash, Pablo J.Fernndez-Marcos, Cian J. Lynch, Daniel

    During 2013, we have shown in micethat in vivoconditions are permissiveto cellular de-differentiation andreprogramming. Moreover, in vivoreprogramming achieves a state of

    pluripotency that is more primitivethan the one achieved in vitro. Thesendings were published in Natureandwere considered Discovery of Year bythe journal Nature Medicine.

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 21

    RESEARCH HIGHLIGHTS

    expression in pre-tumoural murine thyroids revealed

    that SIRT1 stabilises c-MYC protein and increases c-MYCtranscriptional programmes. Interestingly, in the case ofhuman thyroid cancer, SIRT1 is frequently overexpressedand its levels correlate positively with those of c-MYC. Ourresults implicate SIRT1 as a new candidate target for thetreatment of thyroid carcinomas.

    Cell reprogramming in vivo

    Reprogramming into pluripotency is an intense field ofinvestigation that is already providing many insights intocell plasticity. Cell reprogramming had so far been achieved

    SIRT1 promotes thyroid cancer

    While the existing evidence in mice indicated that SIRT1has potent tumour suppressor activity in a variety of cancermodels, we wanted to study the impact of SIRT1 on cancerassociated to PTEN loss; for this purpose, we crossed our2 lines ofSirt1transgenic mice withPten-deficient mice.Surprisingly, rather than protecting,Sirt1overexpressiondecreased the survival of thePten-deficient mice (FIGURE1). Contrary to previous accumulated evidence in othercancer types whereSirt1acts as a tumour suppressor,Sirt1is oncogenic in the thyroid and prostate (FIGURE 1). Thiseffect was particularly prominent in the case of thyroidcancers, which were all metastatic. The analyses of mRNA

  • 5/19/2018 INFORME CNIO 3013

    24/248

    nostic marker and candidate therapeutictarget for NSCLC. Int J Cancer132, 1986-1995.

    Gonzlez-Navarro H, Vinu A, Sanz MJ,Delgado M, Pozo MA, Serrano M, BurksDJ, Andrs V (2013). Increased dosage ofInk4/Arf protects against glucose intoler-

    ance and insulin resistance associated withaging.Aging Cell12, 102-111.

    Redrejo-Rodrguez M, Muoz-Espn D, Hol-guera I, Menca M, Salas M (2013). Nuclear

    and nucleoid localization are independentlyconserved functions in bacteriophage ter-

    minal proteins. Mol Microbiol90, 858-868. Porteiro B, Daz-Ruiz A, Martnez G, Senra

    A, Vidal A, Serrano M, Gualillo O, LpezM, Malagn MM, Diguez C, Nogueiras

    Colvin EK, Njicop EN, Sutherland RL, LiuT, Serrano M, Bouwens L, Real FX, BiankinAV, Rooman I (2013). Sirtuin-1 RegulatesAcinar-to-Ductal Metaplasia and SupportsCancer Cell Viability in Pancreatic Cancer.Cancer Res73, 2357-2367.

    Herranz D, Maraver A, Caamero M, G-mez-Lpez G, Inglada-Prez L, Robledo M,Castelblanco E, Matias-Guiu X, Serrano M(2013). SIRT1 promotes thyroid carcinogen-esis driven by PTEN deficiency.Oncogene32, 4052-4056.

    Oliemuller E, Pelez R, Garasa S, Pajares MJ,Agorreta J, Po R, Montuenga LM, Teijeira A,Llanos S, Rouzaut A (2013). Phosphorylatedtubulin adaptor protein CRMP-2 as prog-

    embryonic development. Cell155, 1104-1118. Fernandez-Marcos PJ, Serrano M (2013).

    Sirt4: the glutamine gatekeeper. CancerCell23, 427-428.

    Gomez-Sanchez JA, Gomis-Coloma C,

    Morenilla-Palao C, Peiro G, Serra E, SerranoM, Cabedo H (2013). Epigenetic induction ofthe Ink4a/Arf locus prevents Schwann celloverproliferation during nerve regenerationand after tumorigenic challenge. Brain136,2262-2278.

    Ortega-Molina A, Serrano M (2013). PTEN

    in cancer, metabolism, and aging. TrendsEndocrinol Metab24, 184-189.

    Wauters E, Sanchez-Arvalo Lobo VJ, Pinho

    AV, Mawson A, Herranz D, Wu J, Cowley MJ,

    PUBLICATIONS

    Abad M, Mosteiro Ll, Pantoja C, Caam-ero M, Rayon T, Ors I, Graa O, Megas D,Domnguez O, Martinez D, Manzanares M,

    Ortega S, Serrano M (2013). Reprogrammingin vivo produces teratomas and iPS cells withtotipotency features. Nature502, 340-345.

    Lpez-Otn C, Blasco MA, Partridge L, Ser-rano M, Kroemer G (2013). The hallmarks

    of aging. Cell153, 1194-1217. Muoz-Espn D, Caamero M, Maraver

    A, Gmez-Lpez G, Contreras J, Murillo-Cuesta S, Rodrguez-Baeza A, Varela-NietoI, Ruberte J, Collado M, Serrano M (2013).Programmed cellular senescence during

    VICE-DIRECTION OF BASIC RESEARCH

    SCIENTIFIC REPORT 2013 22

    under carefully controlled in vitroculture conditions, whereasthe in vivo tissue microenvironment would in principlesteer towards cellular differentiation and is opposed to

    reprogramming. Bearing the former in mind, we havenevertheless attempted to achieve reprogramming in vivo.

    This year, we have demonstrated that transitory induction ofthe factors Oct4,Sox2,Klf4and c-Mycin mice results in rapiddedifferentiation in multiple tissues, including the stomach,intestine, pancreas and kidney. Dedifferentiation occurredto a variable extent, including the loss of keratin expressionand the acquisition of NANOG expression, a pluripotency

    marker indicative of reprogramming (FIGURE 2). At laterperiods of time, m ice developed multiple teratomas. Thereprogrammable mice also present circulating induced

    pluripotent stem cells (iPS cells) in the blood and, at thetranscriptome level, these in vivogenerated iPS cells arecloser to embryonic stem cells (ES cells) than standard in vitrogenerated iPS cells. Interestingly, in vivoiPS cells efficientlycontribute to the trophectoderm lineage, suggesting that theyachieve a more plastic or primitive state than ES cells. Weconcluded that reprogrammingin vivois feasible and conferstotipotency features absent in standard iPS or ES cells.

    Figure 1 Sirt1overexpression in mice promotes thyroid tumorigenesis and correlateswith increased c-MYC activity. (A)Survival of cohorts of male mice of the indicatedgenotypes. (B, C)Incidence of thyroid and prostate carcinomas, respectively, in 5to 7-month-old mice of the indicated genotypes.

    Figure 2 Many cell types are dedifferentiated and reprogrammed in vivo.Double immunohistochemistry of cytokeratin 19 (CK19, magenta, indicativeof differentiation) and of NANOG (dark brown, indicative of reprogramming)in the stomach (top), the large intestine (middle), and the pancreas (bottom)of reprogrammable mice. All scale bars correspond to 100 m.

  • 5/19/2018 INFORME CNIO 3013

    25/248

    AWARDS AND RECOGNITION

    Jimnez Daz Distinction Award, XLVJimnez Daz Memorial Lecture, Spain.

    Dominguez O, Schonthaler HB, Wagner EF,Serrano M (2013). Reprogramming activityof NANOGP8, a NANOG family memberwidely expressed in cancer. Oncogene(inpress). PMID 23752184.

    PATENTS

    Fujita K, Harris C, Horikaza I, Mondal A, Park,Pine SR, Serrano M (2013). Manipulationsof stem cell functions by p53 isoforms. EP11771334.

    Serrano M, Abad M, Mosteiro L (2013).Novel Induced Pluripotent Stem Cells andmethods of preparation. EP13382187.

    Serrano M, Ramrez JM. (2013). Super p53mice display retinal astroglial changes.PLoS

    One8, e65446.

    Articles in press

    Garca-Rodrguez JL, Barbier-Torres L,Fernndez-lvarez S, Juan VG, Monte MJ,Halilbasic E, Herranz D, Alvarez L, Aspi-chueta P, Marn JJ, Trauner M, Mato JM,Serrano M, Beraza N, Martnez-Chantar ML

    (2013). SIRT1 controls liver regeneration byregulating BA metabolism through FXRand mTOR signaling.Hepatology(in press).PMID: 24338587.

    Palla AR, Piazzolla D, Abad M, Li H,

    R (2013). Ghrelin Requires p53 toStimulate Lipid Storage in Fat and Liver.Endocrinology154, 3671-3679.

    Georgakopoulou EA, Tsimaratou K, Evan-

    gelou K, Fernandez-Marcos PJ, ZoumpourlisV, Trougakos IP, Kletsas D, Bartek J, SerranoM, Gorgoulis VG (2013). Specific lipofuscinstaining as a novel biomarker to detectreplicative and stress-induced senescence.A method applicable in cryo-preserved andarchival tissues.Aging5, 37-50.

    Li H, Marple T, Hasty P (2013). Ku80-deletedcells are defective at base excision repair.Mutat Res 745-746, 16-25.

    Salazar JJ, Gallego-Pinazo R, de Hoz R,Pinazo-Durn MD, Rojas B, Ramrez AI,

    MOLECULAR ONCOLOGY PROGRAMME | TUMOUR SUPPRESSION GROUP

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 23

    Developmentally-programmed cellular senescence

    Cellular senescence is emerging as an important aspect of

    tissue remodelling. Until now, senescence had been associatedto accidental, non-programmed, tissue damage; as it occursin pathological states including cancer. This year, we havediscovered that senescence also occurs during mammalianembryonic development in a programmed manner and activelyparticipates in multiple tissue remodelling processes. Wefocused on the mesonephros and on the endolymphatic sac ofthe inner ear. Senescence at both structures depends strictlyon p21, and it is independent of DNA damage, p53 or other cell

    cycle inhibitors. We have also demonstrated that the TGF/SMAD and PI3K/FOXO pathways regulate developmentalsenescence (FIGURE 3). Importantly, developmental senescence

    is followed by subsequent macrophage infiltration andclearance of the senescent cells, thus completing the cycle oftissue remodelling. In the absence of p21, loss of senescence ispartially compensated by apoptosis, but still results in detectabledevelopmental abnormalities. Human embryos also presentsenescence markers at the mesonephros and endolymphaticsac. We propose that senescence emerged during the course ofevolution as an embryonic tissue remodelling process, and thatit was subsequently adapted for tissue repair upon damage. s

    Figure 3 Summary of the molecularmechanisms and function of develop-mentally programmed senescence.

  • 5/19/2018 INFORME CNIO 3013

    26/248

    Staff Scientists

    Matthias Drosten, Carmen Guerra,

    Monica A. Musteanu, David Santamara

    Mariano Barbacid

    Group LeaderEXPERIMENTAL

    ONCOLOGY GROUP

    VICE-DIRECTION OF BASIC RESEARCH

    SCIENTIFIC REPORT 2013 24

    Role of Ras signalling in skin development

    Proliferation in the epidermis is a tightly controlled process.

    During skin development, epidermis formation and hair follicle

    morphogenesis crucially depend on the regulated balance between

    proliferation and differentiation. We have deleted all threeRas

    loci (H-Ras,N-RasandK-Ras) from keratinocytesin vitroas well

    as specifically from the epidermis in mice using a K5Cre strain.

    Upon Ras elimination, keratinocytes ceased proliferation and

    entered into senescence without any signs of apoptosis induc-

    tion. Constitutive activation of the MAPK pathway was able topartially rescue the proliferative defects. In mice, Ras signalling

    was essential for proper development of the epidermis and hair

    follicles. Deletion of the threeRasloci during epidermis formation

    in mouse embryos caused a dramatic decrease in proliferation,

    substantially thinner epidermis and delayed appearance of dif-

    ferentiation markers. We did not detect apoptotic or senescent

    cells suggesting that loss of Ras protein expression only leads to

    severe hypoproliferation. These observations provide genetic

    evidence for an essential role of Ras proteins in the control of

    keratinocyte and epidermal proliferation.

    Identification of cancer initiating

    cells in K-Rasdriven lung adenocarcinoma

    Ubiquitous expression of a K-RasG12Voncogene in adult mice

    only induced overt tumours in lungs. To identify these trans-

    formation-permissive cells, we inducedK-RasG12Vexpression in

    a very limited number of adult lung cells. Four weeks later, 30%

    of these cells had proliferated to form small clusters. However,

    only surfactant protein C stained (SPC+) alveolar type II (ATII)

    cells were able to form hyperplastic lesions, some of which

    progressed to adenomas and adenocarcinomas. In contrast,induction of K-RasG12Vexpression in lung cells by intratra-

    cheal infection generated hyperplasias in all regions except

    the proximal airways. Bronchiolar and bronchioalveolar duct

    junction hyperplasias were primarily made of CC10+ Clara cells.

    Some of them progressed to benign adenomas. However, only

    alveolar hyperplasias made up of ATII cells, progressed to yield

    malignant adenocarcinomas. Adenoviral infection induced

    inflammatory infiltrates primarily made of T and B cells. This

    inflammatory response was essential for the development of

    K-RasG12V-driven bronchiolar hyperplasias and adenomas, but

    not for the generation of SPC+ ATII lesions. Finally, activation

    ofK-RasG12Vduring embryonic development under the control

    OVERVIEW

    K-Rasoncogenes have been implicated in about one fifth of

    all human cancers including those with the worst prognosis,

    such as lung adenocarcinoma and pancreatic ductal

    adenocarcinoma. We have developed genetically modified

    mouse models (GEMMs) that closely recapitulate the natural

    history of these human cancers. We have used these strainsto validate targets of potential therapeutic value, with the

    ultimate goal of translating these findings into the clinic.

    We have crossed these GEM strains with mice that carried

    conditional knock-out mutations in loci encoding potential

    therapeutic targets. These targets can be ablated once the

    tumour has been generated to determine whether they are

    essential, or at least relevant, for tumour development. This

    genetic-based strategy has significant advantages over classical

    pharmacological studies since it does not rely on the quality

    of the drug/inhibitor and the observed effects are always

    mechanism-based, not off-target effects. Moreover, if the

    target is eliminated systemically, our studies offer relevant

    information regarding potential toxic effects that may occurwhen the target is pharmacologically inhibited in normal

    tissues. More recently, we are replacing the conditional

    knock-out strains with conditional knock-in mice so that we

    will express kinase dead isoforms in tumour tissue instead

    of eliminating the target. We hope that this experimental

    approach will mimic more accurately those pharmacological

    responses that will be observed in the clinic.

    RESEARCH HIGHLIGHTS

  • 5/19/2018 INFORME CNIO 3013

    27/248

    Research Associates

    Jose Javier Berenguer, Juan VelascoTechnicians

    M. Carmen Gonzlez, Beatriz Jimnez,Marta San Romn, Raquel Villar

    Graduate Students

    Mara Teresa Blasco (since February),Magdolna Djurec, Isabel Hernndez,Sara Mainardi (until August), CarolinaNavas (until August), Patricia Nieto,Luca Simn, Catherine E. Symonds

    Post-Doctoral Fellows

    Chiara Ambrogio, Emilie Bousquet (untilSeptember), Sarah Francoz, Raquel

    Garca, Harrys K.C. Jacob

    MOLECULAR ONCOLOGY PROGRAMME | EXPERIMENTAL ONCOLOGY GROUP

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 25

    Figure Proliferating K-RasG12Voncogene express-ing cells are SPC+ alveolar type II cells. Repre-sentative sections of X-Gal stained lungs wereincubated with antibodies against (A) SPC andCC10 and (B) SPC and AQP5. Merged imagesrepresent the overlay of X-Gal staining and IFimages. Nuclei were counterstained with DAPI.

    Scale bars represent 20 m.

    PUBLICATIONS

    Berns A, Barbacid M (2013). Mouse modelsof cancer. Mol Oncol7, 143-145.

    Guerra C, Barbacid M (2013). Geneticallyengineered mouse models of pancreaticadenocarcinoma.Mol Oncol7, 232-247.

    Drosten M, Lechuga CG, Barbacid M . Rassignaling is essential for skin develop-ment. OncogeneJuly 8, 2013. doi:10.1038/onc.2013.254.

    Alvarez R, Musteanu M, Garcia-Garcia E,Lopez-Casas PP, Megias D, Guerra C, MuozM, Quijano Y, Cubillo A, Rodriguez-Pascual J,Plaza C, de Vicente E, Prados S, Tabernero S,Barbacid M, Lopez-Rios F, Hidalgo M (2013).

    Stromal disrupting effects of nab-paclitaxel inpancreatic cancer.Br J Cancer109, 926-933.

    Azrak SS, Ginel-Picardo A, Drosten M, Bar-

    bacid M, Santos E (2013). Reversible, interre-

    lated mRNA and miRNA expression patterns

    in the transcriptome of Rasless fibroblasts:functional and mechanistic implications. BMC

    Genomics14, 731. Gonzlez-Snchez E, Martn-Caballero

    J, Flores JM, Hernndez-Losa J; ngelesMontero, Corts J, Mares R, Barbacid M, Re-cio JA (2013). Lkb1 Loss Promotes TumorProgression of BRAFV600E-Induced LungAdenomas. PLoS One8, e66933.

    Drosten M, Lechuga CG, Barbacid M (2013).Genetic analysis of Ras genes in epidermaldevelopment and tumorigenesis.SmallGTPases4,4.

    Articles in press

    Hu W, Nevzorova YA, Haas U, Moro N, Si-

    cinski P, Geng Y, Barbacid M, Trautwein C,Liedtke C . Concurrent deletion of cyclin

    E1 and cyclin-dependent kinase 2 in he-patocytes inhibits DNA replication andliver regeneration in mice. Hepatology(inpress). PMID: 23787781.

    Mainardi S, Mijimolle N, Francoz S, Vicente-Dueas C, Snchez-Garca I, Barbacid M .Identification of cancer initiating cells in K-Ras driven lung adenocarcinoma. Proc Natl

    Acad Sci USA(in press). PMID:24367082. Cerqueira A, Martn A, Symonds CE, Oda-jima J, Dubus P, Barbacid M, Santamara D.Genetic characterization of the role of theCip/Kip family of proteins as Cdk inhibi-tors and assembly factors. Mol Cell Biol(inpress). PMID: 24515438.

    Lawler M, Le Chevalier T, Murphy MJ Jr,Banks I, Conte P, De Lorenzo F, Meunier F,

    Pinedo HM, Selby P, Armand JP, BarbacidM, Barzach M, Bergh J, Bode G, Cameron

    DA, de Braud F, de Gramont A, Diehl V,Diler S, Erdem S, Fitzpatrick JM, Geissler J,Hollywood D, Hjgaard L, Horgan D, JassemJ, Johnson PW, Kapitein P, Kelly J, KloezenS, La Vecchia C, Lwenberg B, Oliver K,Sullivan R, Tabernero J, Van de Velde CJ,Wilking N, Wilson R, Zielinski C, Zur HausenH, Johnston PG. A catalyst for change: theEuropean cancer patients Bill of rights.Oncologist(in press). PMID: 24493667.

    AWARDS AND RECOGNITION

    Foreign Member, National Academy ofSciences of the US, Section 41: MedicalGenetics, Hematology and Oncology.

    of aSca1promoter exclusively yielded CC10+ lesions, includingadenomas. These results illustrate that different types of lungcells, at various developmental stages, can generate tumourlesions in response toK-Ras. However, in adult mice, onlySPC+ ATII cells were able to yield malignant adenocarcinomas.

    K-RasV14I-induced Noonan syndrome

    predisposes to tumour development in mice

    Noonan syndrome (NS) is an autosomal dominant genetic disordercharacterised by short stature, craniofacial dysmorphism andcongenital heart defects. A significant fraction of NS patients alsodevelop myeloproliferative disorders (MPD). Mutations respon-sible for NS occur in at least eight different loci includingK-RAS.

    We have generated a mouse model for NS induced by the mostfrequentK-RASmutation in NS patients. Mutant mice displayedmultiple NS-associated developmental defects such as growthdelay, craniofacial dysmorphia, cardiac defects and haematologicalabnormalities including MPD. Homozygous animals had perinatallethality, increased predisposition to tumour development otherthan MPD and cooperated with tumour suppressors. Exposure ofpregnant mothers to a MEK inhibitor rescued perinatal lethality

    and prevented craniofacial dysmorphia and cardiac defects, butnot haematological disorders including MPD. However, thesedefects were not corrected when mice were treated after wean-ing. These mice offer an experimental tool to study the molecularalterations underlying the clinical manifestations of NS and totest new therapies aimed at preventing or ameliorating thosedeficits associated with this syndrome. s

  • 5/19/2018 INFORME CNIO 3013

    28/248

    OVERVIEW

    Telomere length defects are associated to cancer and ageingprocesses, and have a profound effect on stem cell behaviour.

    We aim to determine the role of genetic and epigenetic telomereregulators in cancer and ageing by generating new mouse modelsand studying the role of these factors in stem cell biology.

    Our research aims at:

    Understanding the biology of the telomeres and telomeraseby generating mouse models to probe the role of telomeresand telomerase in cancer and ageing.

    We study the mechanisms by which tumour cells are immortaland normal cells are mortal. The immortality of cancer cellsis one of their most universal characteristics. The enzymetelomerase is present in more than 95% of all types of humancancers but is not present in normal cells in the body. Telomeresare nucleoprotein complexes located at the ends of chromosomesthat are essential for chromosome protection and genomicstability. One of the many factors that lead to ageing is theprogressive shortening of telomeres associated with organismageing. When telomeres are altered (in their length or theirintegrity) adult stem cells have a maimed regenerative capacity.

    Nora Sobern MEX

    Mara Garca-Beccaria ESPAksinya Derevyanko RUSMartina Stagno dAlcontres ITAMaria Luigia e Bonis ITA

    Paula Martnez ESPRosa M. Marin ESPIsabel Lpez de Silanes ESPMaria A. Blasco ESP

    Staff Scientists

    Isabel Lpez de Silanes, Rosa M. Marin,Paula Martnez, Marinela Mndez (sinceApril), gueda M. Tejera, Elisa Varela

    Maria A. BlascoGroup LeaderTELOMERES AND

    TELOMERASE GROUP

    VICE-DIRECTION OF BASIC RESEARCH

    SCIENTIFIC REPORT 2013 26

  • 5/19/2018 INFORME CNIO 3013

    29/248

    Deciphering the interplay between telomeres and DNArepair pathways.

    Characterising the role of telomeric heterochromatin. Developing strategies for telomerase activation. Elucidating the role of telomerase and telomeres in

    adult stem cell biology and in nuclear reprogramming ofdifferentiated cells to induced Pluripotent Stem (iPS) cells.

    Rosa M. Serrano ESPMercedes Gallardo ESPJuan Manuel Povedano ESPIanire Garrobo ESP

    Christian Br DEUElisa Varela ESPgueda M. Tejera ARGMarinela Mndez COL

    Technicians

    Mercedes Gallardo, Rosa M. Serrano,Nora Sobern

    Juan Manuel Povedano,Ralph P. Schneider (until March)

    Graduate Students

    Aksinya Derevyanko, Miguel Foronda,Mara Garca-Beccaria, Ianire Garrobo,

    Post-Doctoral Fellows

    Christian Br, Bruno Bernardes de Jess(until June), Maria Luigia de Bonis,

    Benjamin Kumpfmller (until August),Martina Stagno dAlcontres

    MOLECULAR ONCOLOGY PROGRAMME | TELOMERES AND TELOMERASE GROUP

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 27

    Our exploration of a new mechanism,by which mutations in the telomericprotein POT1 contribute to thedevelopment of human chroniclymphocytic leukaemia, may facilitatenovel approaches for therapeuticintervention and clinical managementof this disease.

  • 5/19/2018 INFORME CNIO 3013

    30/248

    ture predicts biological age in mice.AgingCell12, 93-101.

    Martinez-Delgado B, Gallardo M, Tanic M,Yanowsky K, Inglada-Perez L, Barroso A,Rodriguez-Pinilla M, Caamero M, BlascoMA, Benitez J (2013). Short telomeres arefrequent in hereditary breast tumors and areassociated with high tumor grade. BreastCancer Res Treat141, 231-242.

    Vera E, Bernardes de Jesus B, Foronda M,Flores JM, Blasco MA (2013). Telomerase

    is essential for the generation of inducedpluripotent stem cells.Nat Commun 4, 1946.

    Bernardes de Jesus B, Blasco MA (2013).Telomerase at the intersection of cancer andaging. Trends Genet29, 513-520.

    Stout GJ, Blasco MA (2013). TelomereLength and Telomerase Activity Impactthe UV Sensitivity Syndrome XerodermaPigmentosum C.Cancer Res73, 1844-1854.

    Toms-Loba A, Bernardes de Jesus B, MatoJM, Blasco MA (2013). A metabolic signa-

    rano M, Kroemer G (2013). The hallmarksof aging. Cell153, 1194-1217.

    Mondal AM, Horikawa I, Pine SR, Fujita K,Morgan KM, Vera E, Mazur SJ, Appella E,Vojtesek B, Blasco MA, Lane DP, Harris CC(2013). p53 isoforms regulate aging- andtumor-associated replicative senescence inT lymphocytes.J Clin Invest123, 5247-5257.

    Schneider RP, Garrobo I, Foronda M, Pala-cios JA, Marin RM, Flores I, Ortega S, BlascoMA (2013). TRF1 is a stem cell marker and

    PUBLICATIONS

    Ramsay AJ, Quesada V, Foronda M, CondeL, Martnez-Trillos A, Villamor N, RodrguezD, Kwarciak A, Garabaya C, Gallardo M,Lpez-Guerra M, Lpez-Guillermo A, PuenteXS, Blasco MA, Campo E, Lpez-Otn C(2013). POT1 mutations cause telomeredysfunction in chronic lymphocytic leu-kemia. Nat Genet45, 526-530.

    Lpez-Otn C, Blasco MA, Partridge L, Ser-

    VICE-DIRECTION OF BASIC RESEARCH

    SCIENTIFIC REPORT 2013 28

    Human chronic lymphocytic leukaemia and telomere

    dysfunction due to mutations in telomeric protein POT1

    Chronic lymphocytic leukaemia (CLL), the most frequentleukaemia in adults in Western countries, affects more thanone thousand new patients in Spain each year. Analysis ofsequencing data from CLL patients uncovered that the gene

    POT1is one of the most frequently mutated genes in thisillness.POT1encodes the telomeric protein POT1; a proteinthat, by acting as a staple, fixes in place the protective hoodthat safeguards the telomeres.

    Somatic mutation ofPOT1affects key protein residues that

    are required for binding to telomeric DNA, thus preventingthis gene from fulfilling its function. We showed thatPOT1-mutated CLL cells have numerous telomeric and chromosomalabnormalities (FIGURE 1) that suggest thatPOT1mutationsfavour the acquisition of the malignant features of CLL cells.The study of the biochemical pathways that lead from thesechromosomal abnormalities to the uncontrolled growth of Blymphocytes could provide clues for a better understandingof CLL in particular and of human cancer in general.

    Telomeric protein TRF1 is a stem cell markerand is essential for nuclear reprogramming

    Deficiency of the telomeric protein TRF1 leads to earlyembryonic lethality suggesting an essential role for TRF1 inearly mouse development and to severe organ atrophy anddysfunction when deleted in adult tissues; this suggests thatTRF1 may also be important for adult stem cell compartmentsand maintenance of organ homeostasis. To study the role of TRF1in stem biology and tissue homeostasis, we have generated areporter mouse carrying eGFP-TRF1. In this context, generationof a reporter mouse for TRF1 is a valuable tool to understandthe regulation of TRF1 expression in normal and pluripotent/adult stem cells, and also to address if cells with high TRF1 levels

    are associated with higher stemness/pluripotency.

    We found that eGFP-TRF1 expression in mice is maximal

    in known adult stem cell compartments (FIGURE 2), andshowed that TRF1 ensures their functionality. This discoveryis useful for identifying and eventually isolating the stemcell population in tissues; something that is importantfor the development of regenerative medicine. We havedetermined that eGFP-TRF1 is highly expressed in inducedpluripotent stem (iPS) cells and that TRF1 expression isuncoupled from the telomere elongation associated withreprogramming. We observed that selection of eGFP-TRF1-high iPS cell populations correlates with higher pluripotencyas indicated by their ability to form teratomas and chimaeras.We also showed that TRF1 is necessary for both induction

    and maintenance of pluripotency and that TRF1 is a directtranscriptional target of Oct3/4.

    Telomeric protein RAP1 protectsfrom obesity through its extratelomeric roleregulating gene expression

    To investigate the non-telomeric roles of RAP1 in vivo, wehave now generated a RAP1 whole-body knock-outmouse.These mice presented an early onset of obesity, which is moresevere in females than in males and is aggravated under ahigh-fat diet.Rap1-deficient mice showed accumulation of

    fat in abdominal depots, developed hepatic steatosis, andhad high fasting plasma levels of insulin, glucose, cholesterol,and alanine transaminase. Gene expression analyses of theliver and visceral white fat from Rap1-deficient mice beforethe onset of obesity indicated deregulation of key metabolictranscriptional programmes including fatty acid metabolism,PPARsignalling and glucose metabolism. We identified

    PparandPgc1, as well as their target genes, as the keymetabolic pathways affected byRap1deletion in the liver. Wefurther showed that RAP1 binds toPparandPgc1loci andmodulates their transcription.These findings have shownan unprecedented role for a telomere-binding protein in the

    regulation of metabolism. s

    RESEARCH HIGHLIGHTS

  • 5/19/2018 INFORME CNIO 3013

    31/248

    ES, Sikora E, Gradinaru D, Doll M, SalmonM, Kristensen P, Griffith H, Libert C, Grune T,Breusing N, Simm A, Franceschi C, Talbot D,CaiafaP, Friguet B, Slagboom E, Hervonen A,

    Aspinall R (2013). Determination of biologicalage in human beings with a chronologicalage of 35-74 years. EP 13 001 450.

    AWARDS AND RECOGNITION

    Honorary Ambassador of the Spain Brand

    2013 in the Science and Innovation category,Leading Brands of Spain Forum.

    Honorary Award from the Ctedra Real Ma-dridfor an outstanding scientific career, Spain.

    Corresponding Fellow of the Spanish Royal

    Academy of Pharmacy. Member of the Selection Committee, 2013

    Pezcoller Foundation-AACR InternationalAward for Cancer Research.

    Member of the Jury, 2013 Inbev Baillet LatourHealth Prize, Leuven, Belgium.

    Wilson JS, Tejera AM, Castor D, Toth R,Blasco MA, Rouse J (2013). Localization-dependent and -independent roles of SLX4

    in regulating telomeres.Cell Rep4, 853-860.

    PATENT

    Blasco M, Gallardo M, Brkle A, Junk M, Ber-told M, Moreno-Villanueva M, Bernhardt J,Hoeijmakers HJ, Toussaint O, Grubeck-Loe-

    benstein B, Mocchegiani E, Collino S, Gonos

    reverse transcriptase synergizes with calorierestriction to increase health span and ex-tend mouse longevity. PLoS One8, e53760.

    Blasco MA (2013). Interview: Fightingdisease from the chromosome end.Epigenomics5, 483-485.

    Martnez P, Gmez-Lpez G, Garca F, Mer-cken E, Mitchell S, Flores JM, De Cabo R,Blasco MA (2013). RAP1 protects from obe-

    sity through its extratelomeric role regulatinggene expression. Cell Rep3, 2059-2074.

    MOLECULAR ONCOLOGY PROGRAMME | TELOMERES AND TELOMERASE GROUP

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 29

    Figure 2 TRF1 levels are highest in adultstem cell compartments of the skin and the

    small intestine. (A) eGFPTRF1 fluorescencein different compartments of tail-skin fol-licles of eGFPTRF1 mice decreases from theputative stem cell niches towards the moredifferentiated compartments. (B) Small

    intestine cell type definition.

    Figure 1 Chromosomal aberrations in cells from individuals with CLL expressingmutated POT1. (A) Telomeric FISH on metaphase spreads from POT1-mutated andcontrol CLL samples. (BD) Per-individual chromosomal aberrations for sister chro-

    matidtype end-to-end fusions (SCFs) (B), chromosome-type end-to-end fusions(C) and multitelomeric signals (MTSs) (D).

  • 5/19/2018 INFORME CNIO 3013

    32/248

    David Partida USA

    Beln Sanz ESP

    Marianna Trakala GRC

    Ana Martins PRT

    Manuel Eguren ESPElena Domnech ESPMara Salazar ESP

    Ignacio Prez de Castro ESPGuillermo de Crcer ESPMnica lvarez ESP

    Mara Sanz ESPAlejandra Gonzlez ESP

    Eva Porlan ESP

    Marcos Malumbres ESP

    Staff Scientists

    Mnica lvarez, Guillermo de Crcer,Ignacio Prez de Castro, Eva Porlan

    (since July)

    Marcos MalumbresGroup LeaderCELL DIVISION AND

    CANCER GROUP

    VICE-DIRECTION OF BASIC RESEARCH

    SCIENTIFIC REPORT 2013 30

  • 5/19/2018 INFORME CNIO 3013

    33/248

    OVERVIEW

    a Chromosome Region Maintenance 1 (CRM1)-dependentmanner before NEB. We postulate that, once at the cytoplasm,Greatwall inhibits the phosphatase 2 (PP2A)-B55 complexes

    to maintain the mitotic state (FIGURE 1).

    Our findings may have therapeutic implications since Greatwallacts by blocking the function of the PP2A phosphatase; atumour suppressor frequently altered in human cancer. Thisimplies that the inhibition of Greatwall could, at the same time,slow down cell division and reactivate tumour suppressorPP2A; a protein capable of inhibiting many of the oncogenicmolecular pathways involved in cancer development. We areactively working on this possibility.

    Greatwall: a new target for cancer therapy?

    Greatwall, also known as Mastl, is a recently identified kinase

    that regulates cell division. Until now, most studies on thisprotein were carried out in invertebrates. Our group, incollaboration with researchers from the National Centrefor Scientific Research (CNRS) in Montpellier, France, hasnow generated the first genetic mouse model of this protein.Using this conditional knockout model, we have shown thatGreatwall is essential for mouse embryonic development andcell cycle progression. This is due to mitotic collapse afternuclear envelope breakdown (NEB). We demonstrated thatGreatwall is exported from the nucleus to the cytoplasm in

    consequences of cell cycle deregulation in vivo. To characterise the function of microRNAs in cell biology

    and tumour development, as well as their potential usein cancer therapy.

    To understand how progenitor cells and cancer stem cellscontrol their self-renewal and proliferative properties.

    The main focus of our group is to understand the mechanismsby which mitosis is regulated. We are interested, not onlyin deciphering the control of chromosome segregationduring mitosis, but also in finding new ways to block tumourprogression through the inactivation of mitotic regulators.

    Using mouse models as a major research tool, we investigatemitotic kinases and phosphatases, as well as regulatorycomplexes involved in ubiquitin-dependent degradation ofproteins during mitosis. Other research areas of our groupinclude the study of microRNAs involved in the developmentand progression of hematopoietic tumours, as well as thecontrol of asymmetric cell division in progenitor/stem cellsand their relevance to development, tissue homeostasis andcancer.

    The Groups main objectives are the following:

    To understand the basic control mechanisms of the

    mammalian cell cycle. To characterise the physiological and therapeutic

    Technicians

    Marta Gmez de Cedrn (until July),

    David Partida (since February)

    Ana Martins (since October),Beln Sanz, Mara Sanz (since March),Marianna Trakala

    Graduate Students

    Elena Domnech, Manuel Eguren (until

    May), Alejandra Gonzlez,

    Post-Doctoral Fellow

    Mara Salazar

    MOLECULAR ONCOLOGY PROGRAMME | CELL DIVISION AND CANCER GROUP

    Our group has reported the invivorelevance of three mitoticregulators; namely, Greatwall,Aurora-A and Cdh1. Theseproteins are putative cancer

    targets and our results will becritical to better understandthe function of these proteinsand the effects of theirinhibition in cancer.

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 31

    RESEARCH HIGHLIGHTS

  • 5/19/2018 INFORME CNIO 3013

    34/248

    after nuclear envelope breakdown in

    mammals. Proc Natl Acad Sci USA110,17374-17379.

    Prez de Castro I, Aguirre-Portols C,Fernndez-Miranda G, Caamero M,Cowley DO, Van Dyke T, Malumbres M(2013). Requirements for aurora-A intissue regeneration and tumor devel-opment in adult mammals. Cancer Res73, 6804-6815.

    Gonzlez-Gugel E, Villa-MoralesM, Santos J, Bueno MJ, MalumbresM, Rodrguez-Pinilla SM, Piris M,

    cer: an epigenetics view. Mol AspectsMed34, 863-874.

    Eguren M, Porlan E, Manchado E, Gar-ca-Higuera I, Caamero M, Farias I,Malumbres M (2013). The APC/C co-factor Cdh1 prevents replicative stressand p53-dependent cell death in neural

    progenitors. Nat Commun4, 2880. Alvarez-Fernndez M, Snchez-Martnez

    R, Sanz-Castillo B, Gan PP, Sanz-FloresM, Trakala M, Ruiz-Torres M, Lorca T,Castro A, Malumbres M (2013). Greatwallis essential to prevent mitotic collapse

    Frangini A, Sjberg M, Roman-TruferoM, Dharmalingam G, Haberle V, Bartke T,Lenhard B, Malumbres M, Vidal M, DillonN (2013). The aurora B kinase and thepolycomb protein ring1B combine to

    regulate active promoters in quiescentlymphocytes. Mol Cell51, 647-661.

    Kim JA, Aberg C, de Crcer G, MalumbresM, Salvati A, Dawson KA (2013). Lowdose of amino-modified nanoparticlesinduces cell cycle arrest.ACS Nano7,

    7483-7494. Malumbres M (2013). miRNAs and can-

    PUBLICATIONS

    Kollmann K, Heller G, Schneckenleith-ner C, Warsch W, Scheicher R, Ott RG,Schfer M, Fajmann S, Schlederer M,Schiefer AI, Reichart U, Mayerhofer M,Hoeller C, Zchbauer-Mller S, Ker-jaschki D, Bock C, Kenner L, HoeflerG, Freissmuth M, Green AR, MorigglR, Busslinger M, Malumbres M, Sexl V(2013). A Kinase-independent functionof CDK6 links the cell cycle to tumor

    angiogenesis. Cancer Cell24, 167-181.

    VICE-DIRECTION OF BASIC RESEARCH

    SCIENTIFIC REPORT 2013 32

    Aurora kinases, biomarkers and cancer treatment

    Aurora-A, a major cell cycle regulator, is highly expressedin human tumours; it correlates with poor prognosis insome tumour types. Although the most important roles of

    this molecule have been studied in the past in other modelorganisms and in mouse embryos, the requirements of thiskinase in adult tissues or in young individuals remain unknown.Our group, in collaboration with T. Van Dyke and D. Cowleyat The University of North Carolina, has demonstratedthat inhibition of this kinase results in a premature ageingphenotype when applied to young individuals. This phenotypemainly results from an increase in the levels of senescentcells. These phenomena were accompanied with a significantincrease in the percentage of cells that accumulate high levelsof DNA content, indicating a defect in how cells segregate

    their DNA upon inhibition of this kinase. Our study alsohas important therapeutic consequences since the geneticelimination of Aurora-A efficiently inhibits the proliferationof tumours in mice (FIGURE 2). The fact that inhibition of

    Aurora-A also generated a significant amount of DNA damage is

    of special relevance in cancer therapy, since it implies that theinhibition of Aurora-A could sensitise tumours to anticanceragents that work better against cancer cells with high levelsof DNA damage.

    We hav e fou nd tha t Aur ora-A -defi cie nt tum our s arecharacterised by an accumulation of polyploid cells. Thesecells display a low proliferative potential, resulting in adefect in the ability of the tumour to progress. From theseresults, we propose that scoring the number of polyploidcells in patients treated with these compounds should be

    Figure 1 How to prepare a cell fornuclear envelope breakdown. Acti-

    vation and nuclear import of CycB/Cdk1 triggers Greatwall export to thecytoplasm, where it inhibits PP2A/B55. A defective inhibition of PP2A inearly mitosis would cause a defectivephosphorylation of Cdk substrates

    upon NEB leading to the mitotic col-lapse observed in Greatwall null cells(modified from Alvarez-Fernndez

    et al., 2013).

  • 5/19/2018 INFORME CNIO 3013

    35/248

    potentiation in the amygdala of adultmice. Learn Memory20, 11-20.

    Tuck C, Zhang T, Potapova T, MalumbresM, Novk B (2013). Robust mitotic entryis ensured by a latching switch. Biol Open2, 924-931.

    Alvarez-Fernndez M, Medema RH

    (2013). Novel functions of FoxM1: frommolecular mechanisms to cancer ther-apy. Front Oncol 5.

    Michel CI, Malumbres M (2013) MicroR-NA-203: tumor suppression and beyond.MicroRNA2, 118-126.

    Book Chapters

    Malumbres, M (2013). Cyclins and Cyclin-dependent kinases. In: Encyclopedia ofSystems Biology, (Dubitzky, W., Wolken-

    hauer, O., Yokota, H. & Cho, K.H., eds.),Springer, Heidelberg, in press.

    Malumbres, M (2013). Mitotic kinases. In:Encyclopedia of Systems Biology(Du-bitzky, W., Wolkenhauer, O., Yokota, H.& Cho, K.H., eds), Springer, Heidelberg,in press.

    Domnech E, Malumbres M (2013).Mitosis-targeting therapies: a trouble-shooting guide. Curr Opin Pharmacol13, 519-528.

    Trakala M, Fernndez-Miranda G, Prez

    de Castro I, Heeschen C, Malumbres M(2013). Aurora B prevents delayed DNAreplication and premature mitotic exitby repressing p21 (Cip1). Cell Cycle12,1030-1041.

    Pick JE, Malumbres M, Klann E (2013).The E3 ligase APC/C-Cdh1 is required forassociative fear memory and long-term

    Fernndez-Piqueras J (2013). Down-regulation of specific miRNAs enhancesthe expression of the gene Smoothenedand contributes to T-cell lymphoblasticlymphoma development. Carcinogenesis 34, 902-908.

    Parrillas V, Martnez-Muoz L, HolgadoBL, Kumar A, Cascio G, Lucas P, Rodr-guez-Frade JM, Malumbres M, CarreraAC, van Wely KH, Mellado M (2013). Sup-pressor of cytokine signaling 1 blocksmitosis in human melanoma cells. CellMol Life Sci70, 545-558.

    MOLECULAR ONCOLOGY PROGRAMME | CELL DIVISION AND CANCER GROUP

    Figure 2 Depletion of Aurora-A inhib-its the growth of oncogene-induced

    mammary tumours, whereas, controltumours significantly increase theirmetabolic activity measured by PET.Aurora-A null tumours did not increasetheir metabolic activity with time

    (Prez de Castro et al., 2013).

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 33

    highly informative for evaluating the efficiency of thesedrugs in clinical studies.

    The APC/C cofactor Cdh1, replicativestress and neural progenitors

    The requirements for the APC/C during mitotic exit haverecently been proposed as new therapeutic strategies againstcancer. One possible complication of these strategies residesin the possible undesired effect of inhibiting APC/C-Cdh1,since lack of Cdh1 activity may result in the accumulationof proliferative molecules such as cyclins, or oncogenes such as Pttg1/Securin or Aurora kinases that could driveincreased cellular proliferation. We have shown that geneticablation of Cdh1in the developing nervous system results inhypoplastic brain and hydrocephalus. These defects correlate

    with enhanced levels of Cdh1 substrates and increased entryinto S-phase in neural progenitors. However, cell divisionis prevented in the absence of Cdh1 due to hyperactivationof cyclin-dependent kinases, increased phosphorylationof H2AX, induction of p53, G2 arrest, and apoptotic death

    of these progenitor cells. This particular requirement forCdh1 during neurogenesis is related to the ability of Cdh1 toprevent replicative stress in progenitors of the developingbrain. Contrary to initial expectations, our genetic datasuggest that ablation of Cdh1results in replicative stress invivoand a general antiproliferative response that is not p53-dependent. Thus, putative APC/C inhibitors are unlikely togenerate proliferative responses, even in the case of unspecificinhibition of Cdh1 and with independence of the p53 statusof tumour cells. s

  • 5/19/2018 INFORME CNIO 3013

    36/248

    Julia Specks DEUFederica Schiavoni ITAMara Nieto ESP

    Sara Rodrigo ESP

    Marta E. Antn ESP

    Isabel Morgado ESPCristina Mayor ESPAndrs J. Lpez-Contreras ESPEmilio Lecona ESP

    Ariana Jacome PRTSergio Ruiz ESPMatilde Murga ESPscar Fernndez-Capetillo ESP

    Staff Scientists

    Matilde Murga, Sergio Ruizscar Fernndez-CapetilloGroup LeaderGENOMIC INSTABILITY

    GROUP

    VICE-DIRECTION OF BASIC RESEARCH

    SCIENTIFIC REPORT 2013 34

  • 5/19/2018 INFORME CNIO 3013

    37/248

    OVERVIEW

    During 2013, we have invested inthe implementation of technologyfor performing forward geneticscreenings in haploid mammaliancells, and discovered that the

    mechanisms that deal with thedissolution of inter-molecularDNA links are important for thesuppression of cancer and ageingin mammals.

    DNA damage is the source of pro-cancerous mutations butrecent evidence has suggested that the reverse connectionmight also exist; namely, that oncogenes can promote thegeneration of DNA damage. However, the nature of thedamage that is caused by oncogenes is still poorly understood.

    Our laboratory has centred its research on trying tounderstand how cells respond to replicative stress (RS); atype of DNA damage that arises, unavoidably, every time that acell replicates its DNA, and which is mainly prevented by ATRand Chk1 kinases. Unfortunately, the essential nature of thesekinases has significantly limited their study, particularly atthe organismal level. In order to overcome these limitations,a major part of our work these past years has focused on thedevelopment of cellular and animal tools for the study of ATRand Chk1. These tools include mice with enhanced or limited

    ATR-Chk1 function, cell systems in which the pathway can beactivated at will, and chemical inhibitors of the ATR kinase.Our studies have revealed the impact of RS on cancer and

    ageing, and have resulted in putative drugs that can be usedto test our conceptual approaches to cancer therapy. Overall,our main goal is to understand how genome maintenance issafeguarded particularly during replication and to exploitthis knowledge as a way to fight cancer.

    Technicians

    Marta E. Antn (since September),Sara Rodrigo, Rebeca Soria (until June)

    Federica Schiavoni (since September),Julia Specks, Enrico Tenaglia (until

    September)

    Graduate Students

    Cristina Mayor, ngela Monasor (untilJuly), Isabel Morgado, Mara Nieto,

    Post-Doctoral Fellows

    Ariana Jacome (until November), EmilioLecona, Andrs J. Lpez-Contreras

    MOLECULAR ONCOLOGY PROGRAMME | GENOMIC INSTABILITY GROUP

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 35

  • 5/19/2018 INFORME CNIO 3013

    38/248

    Stem Cell12, 88-100. Lecona E, Rojas L, Bonasio R, Johnston

    A, Fernndez-Capetillo O, Reinberg D(2013). Polycomb Protein SCML2 Regu-lates the Cell Cycle by Binding and Mod-ulating CDK/CYCLIN/p21 Complexes.PLoS Biol11, e1001737.

    Monasor A, Murga M, Lopez-ContrerasAJ, Navas C, Gomez G, Pisano DG,Fernandez-Capetillo O (2013). INK4a/

    Fernandez-Capetillo O, Nussenzweig A

    (2013). Naked Replication Forks BreakapRPArt. Cell155, 979-980.

    Marqus-Torrejn M, Porlan E, BanitoA, Gmez-Ibarlucea E, Lopez-ContrerasAJ, Fernndez-Capetillo O, Vidal A,

    Gil J, Torres J, Farias I (2013). Cyclin-dependent kinase inhibitor p21 controlsadult neural stem cell expansion byregulating Sox2 gene expression. Cell

    Callen E, Di Virgilio M, Kruhlak MJ,Nieto-Soler M, Wong N, Chen HT,Faryabi RB, Polato F, Santos M, StarnesLM, Wesemann DR, Lee JE, Tubbs A,Sleckman BP, Daniel JA, Ge K, Alt FW,Fernandez-Capetillo O, NussenzweigMC, Nussenzweig A (2013). 53BP1 me-diates productive and mutagenic DNArepair through distinct phosphoproteininteractions. Cell153, 1266-1280.

    PUBLICATIONS

    Barlow JH, Faryabi RB, Calln E, WongN, Malhowski A, Chen HT, Gutierrez-Cruz G, Sun HW, McKinnon P, WrightG, Casellas R, Robbiani DF, Staudt L,Fernandez-Capetillo O, Nussenzweig A(2013). Identification of early replicatingfragile sites that contribute to genomeinstability. Cell152, 620-632.

    VICE-DIRECTION OF BASIC RESEARCH

    RESEARCH HIGHLIGHTS

    SCIENTIFIC REPORT 2013 36

    Searching for players of the DNA damage response

    through forward genetics in haploid mammalian cells

    One of the key advantages of using yeast as a model system isthe capacity to grow it as a haploid organism, which has greatly

    facilitated genetic screenings based on gene-deletions. Inmammals, RNA interference (RNAi) emerged as a powerfulalternative, but unfortunately suffers from significant off-target effects and/or incomplete knockdowns that frequentlylimit its potential. The availability of human haploid cell lines(KBM7 and HAP1) isolated from a leukemic cell line, as wellas the capacity to generate primary mouse haploid embryonicstem cells (mESh), are rapidly shaking the field. During the lastyear we have invested in implementing the technology in ourlaboratory in order to perform forward genetic screeningsin human KBM7/HAP1 cells and mESh, using piggyBactransposons as mutagens. We have already performed some

    initial screenings that were directed at finding mutations that

    generate resistance against commonly used genotoxic drugs.One of these screenings was directed at exploring whetherresistances can arise against ATR inhibitors; compounds thatwe have generated in collaboration with the ExperimentalTherapeutics Programme, and which we believe might be

    particularly useful for the treatment of tumours with highlevels of replication stress. We have currently identifiedseveral mutant clones that can grow normally, even in thepresence of high doses of ATR inhibitors (FIGURE 1), andare working on the characterisation of the genes responsiblefor this resistance. Our next steps in this area include settingup a pipeline for the identification of mutants through NextGeneration Sequencing, as well as developing mEShlines atthe CNIO with the help of CNIOs Transgenic Mice Unit. Thein house generation of haploid lines will enable us to performsynthetic viability screens oriented to unmask genes that, whenmutated, enable the growth of cells lacking tumour suppressors

    that are otherwise essential in normal cells (i.e.BRCA1).

    Figure 1 Isolation of human cellsresistant to ATR inhibition.The picturerepresents the growth of a mutant cloneof human haploid cells at doses of ATRinhibitor that are toxic for wild typecells. This clone was isolated from alibrary of 125,000 clones mutagenised

    with a piggyBac transposon.

  • 5/19/2018 INFORME CNIO 3013

    39/248

    Fernndez-Capetillo O, Segrelles C,

    Paramio JM. p21 suppresses inflamma-tion and tumorigenesis on pRB-deficientstratified epithelia. Oncogene(in press).PMID: 24121270.

    PATENT

    Pastor J, Fernandez-Capetillo Oscar,Martinez S, Blanco C, Rico R, Toledo LI,

    Rodriguez S, Murga M, Varela C, LopezAJ, Renner O, Nieto M (2013). ChemicalEntities as ATR Inhibitors. EP13382089.

    AWARDS AND RECOGNITION

    Doctores Diz-Pintado National Awardfor Cancer Research, Spain.

    Mendez J, Muoz J, Fernandez-CapetilloO (2013). A proteomic characterizationof factors enriched at nascent DNA mol-ecules. Cell Rep3, 1105-1116.

    Article in press

    Saiz-Ladera C, Lara MF, Garn M, RuizS, Santos M, Lorz C, Garca-EscuderoR, Martnez-Fernndez M, Bravo A,

    ARF limits the expansion of cells suf-fering from replication stress. Cell Cycle

    12, 1948-1954. Juan D, Rico D, Marques-Bonet T,

    Fernndez-Capetillo O, Valencia A(2013). Late-replicating CNVs as a sourceof new genes. Biol Open2, 1402-1411.

    Lpez-Contreras AJ, Ruppen I, Nieto-Soler M, Murga M, Rodriguez- Acebes S,Remeseiro S, Rodrigo-Perez S, Rojas AM,

    MOLECULAR ONCOLOGY PROGRAMME | GENOMIC INSTABILITY GROUP

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 37

    A SUMO ligase involved in the dissolution of joint DNAmolecules suppresses cancer and ageing in mammals

    Most of our work focuses on understanding how cells areprotected from the accumulation of replication stress through

    a phosphorylation-based signalling cascade coordinatedby ATR and Chk1 kinases. However, there is emergingevidence that additional signalling pathways based on otherpost-translational modifications, such as SUMOylationor Ubiquitinylation, are also key for these responses. Inthis regard, we have focused our research on NSMCE2; aSUMO ligase that is part of the so-called SMC5/6 complex.This complex is similar to other SMC complexes such ascondensins and cohesins, but its real function remainsunclear. Studies in yeast have suggested that the complexplays a role in dissolving DNA linkages that arise betweensister chromatids. Through the generation of 3 independent

    mouse models of NSMCE2 ( genetrap, conditional knockout

    and a mutant strain lacking SUMO ligase activity) we havediscovered that this complex is essential for the suppressionof mitotic recombination, cancer and ageing in mice (FIGURE2). These (and other) phenotypes resemble those found ona human hereditary disease known as Blooms Syndrome.

    It is noteworthy that mice lacking the SUMO ligase activityof NSMCE2 do not show an obvious phenotype, so thatits role in the SMC5/6 complex seems to be independentof this activity. We are currently exploring the potentialrelationship between the SMC5/6 complex and BLM; theprotein that is defective in Blooms Syndrome patients. Inaddition, we are looking for genes that are particularly toxicto cells harbouring mutations in this complex; the aim beingto design future chemotherapeutic strategies that would beparticularly useful for tumours that present an accumulationof inter-molecular DNA links. s

    Figure 2 NSMCE2 deletion acceleratesageing in mice. (A)The picture exempli-fies the outcome of NSMCE2 deletion inadult mice. Mice develop a progeroidsyndrome with several features thatare reminiscent of Blooms Syndrome,such as (B) pigmentation problems or(C) the accumulation of micronuclei.

  • 5/19/2018 INFORME CNIO 3013

    40/248

    Miriam Rodrguez ESPSamantha Williams GBRMiguel Ruiz ESP

    Iva Krizaic HRVAleksandar Kojic SRBAna Cuadrado ESPAna Losada ESP

    Staff Scientist

    Ana CuadradoAna LosadaGroup LeaderCHROMOSOME

    DYNAMICS GROUP

    VICE-DIRECTION OF BASIC RESEARCH

    SCIENTIFIC REPORT 2013 38SCIENTIFIC REPORT 2013 38

  • 5/19/2018 INFORME CNIO 3013

    41/248

    OVERVIEW

    MOLECULAR ONCOLOGY PROGRAMME | CHROMOSOME DYNAMICS GROUP

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 39

    Graduate Students

    Aleksandar Kojic (since September), IvaTechnician

    Miriam RodrguezKrizaic, Silvia Remeseiro (until March),Miguel Ruiz, Samantha Williams

    Proper development of a multicellular organism entails 2major processes. One is proliferation; i.e. the cell duplicatesits genetic material and divides into 2 identical daughter cells.The other is differentiation; i.e. the specialisation of naiveprecursors into specific cell types. This is accomplished through

    the activation of tissue-specific transcriptional programmesthat establish cell identity. Higher order genome structure isa major determinant of such regulation of gene expression.Our research focuses on a protein complex named cohesinthat occupies a central position in both of these processes.On the one hand, cohesin mediates sister chromatid cohesionand thereby ensures faithful DNA repair by homologousrecombination and proper chromosome segregation duringcell division. On the other hand, cohesin contributes to thespatial organisation of the genome by promoting or stabilisingthe formation of chromatin loops. Mutations in cohesin andits regulatory factors have been identified in a group of humansyndromes, collectively known as cohesinopathies, and also in

    several tumour types. Our goal is to understand how cohesinworks, how it is regulated, and how its dysfunction contributesto cancer and other human diseases. In addition to the studyof cohesin, we are interested in the epigenetic inheritance ofcentromeres mediated by the histone H3 variant CENP-A;another essential aspect of chromosome segregation.

    SPANISH NATIONAL CANCER RESEARCH CENTRE, CNIO 39

    We have found that the cohesin-associated factors Pds5A andPds5B have non-redundant roles inembryonic development and cellproliferation. In particular, Pds5B is

    essential for centromeric cohesionand in its absence the cells oftenmis-segregate their chromosomesand become aneuploid.

  • 5/19/2018 INFORME CNIO 3013

    42/248

    Remeseiro S, Cuadrado A, Kawauchi S,Calof AL, Lander AD, Losada A (2013). Re-duction of Nipbl impairs cohesin loading

    locally and affects transcription but notcohesion-dependent functions in a mousemodel of Cornelia de Lange Syndrome.Biochim Biophys Acta1832, 2097-2102.

    Earnshaw WC, Allshire RC, Black BE,

    Bloom K, Brinkley BR, Brown W, Chee-seman IM, Choo KH, Copenhaver GP,

    Pds5B is required for cohesion estab-lishment and Aurora B accumulation atcentromeres. EMBO J32, 2938-2949.

    Haering CH, Losada A (2013). Under-standing chromatin and chromosomes:from static views to dynamic thinking.EMBO Rep14, 109-111.

    Remeseiro S, Cuadrado A, Losada A(2013) Cohesin in development and

    disease. Development 140, 3715-3718.

    RN, Tardn A, Chanock S, Heath S, Valen-cia A, Losada A, Gut I, Malats N, Real FX(2013). Recurrent inactivation of STAG2in bladder cancer is not associated withaneuploidy. Nat Genet45, 1464-1469.

    Remeseiro S, Losada A (2013). Cohesin,a chromatin engagement ring. Curr OpinCell Biol25, 63-71.

    Carretero M, Ruiz-Torres M, Rodrguez-Corsino M, Barthelemy I, Losada A (2013).

    PUBLICATIONS

    Balbs-Martnez C, Sagrera A, Carrillo-de-Santa-Pau E, Earl J, Mrquez M,

    Vazquez M, Lapi E, Castro-Giner F, Bel-tran S, Bays M, Carrato A, CigudosaJC, Domnguez O, Gut M, Herranz J,Juanpere N, Kogevinas M, Langa X,Lpez-Knowles E, Lorente JA, LloretaJ, Pisano DG, Richart L, Rico D, Salgado

    VICE-DIRECTION OF BASIC RESEARCH

    The specific functions of Pds5

    proteins in vertebrate cells

    In vertebrate somatic cells, cohesin consists of 4 subunits,Smc1, Smc3, Rad21, and either SA1 or SA2. Threeadditional factors, Pds5, Wapl, and Sororin bind cohesinand modulate its dynamic association with chromatin.There are 2 Pds5 proteins in vertebrates, Pds5A and Pds5B,but their functional specificity has remained elusive. Wehave generated conditional knockout alleles for the genesencoding Pds5A and Pds5B. Both genes are individuallyrequired for embryonic development although lethalityoccurs in late post-implantation stages. This has allowed

    us to obtain mouse embryonic fibroblasts in which we couldstudy the functions of the 2 proteins. Our results showthat Pds5 proteins have positive and negative effects onthe stability of cohesins association with chromatin. Inconcert with Wapl, Pds5 proteins promote cohesin releasefrom chromatin both during interphase and mitosis. Ininterphase, this dynamic association could be importantto facilitate chromatin processes like transcription andreplication. In mitosis, dissociation of most cohesin duringprophase allows sister chromatid resolution and therebyensures efficient chromosome segregation. Pds5 proteins haveadditional functions. They are required for Smc3 acetylationby the cohesin acetyl transferases (CoATs) Esco1/2 during

    S phase and for subsequent binding of Sororin; these arethe two key steps for cohesion establishment. While bothPds5A and Pds5B contribute to telomere and arm cohesion,Pds5B is specifically required for centromeric cohesion(FIGURE 1). In the Pds5B null cells, both acetylation ofcohesin by Esco2 and binding of Sororin at pericentromericheterochromatin are significantly decreased. Moreover,reduced accumulation of Aurora B at the inner centromereregion in mitotic chromosomes lacking Pds5B impairs its

    error correction function, promoting chromosome mis-

    segregation and aneuploidy. Decreased proliferation ofPds5B null cells could be explained by mitotic cell death andaneuploidy. Cells lacking Pds5A have a stronger proliferationdefect and Pds5A null embryos present an earlier lethality,but in this case cells display correct ploidy and no mitoticdefects. We speculate that the Pds5A null phenotypes maybe related to altered transcription. Future experiments willhave to address the genome-wide distribution of Pds5A andPds5B, as well as the effects of their ablation in gene expressionduring development.

    Analysis of cohesin functions in a mousemodel for Cornelia de Lange Syndrome

    Cornelia de Lange Syndrome (CdLS) is a genetic disorder thataffects around 1 in 30,000 newborns and is linked to mutationsin cohesin and its regulators. To date, it is unclear whichfunction of cohesin is more relevant to the pathology of thesyndrome. A mouse heterozygous for the gene encoding thecohesin loader Nipblgenerated by the group of A. Calof and A.Lander (University of California at Irvine, USA)recapitulatesmany features of CdLS. We have carefully examined Nipbldeficient cells and found that they have robust cohesion allalong the chromosome. DNA replication, DNA repair and

    chromosome segregation are carried out efficiently in thesecells. While bulk cohesin loading is unperturbed, binding tocertain promoters such as the Protocadherin genes in thebrain is notably affected and alters gene expression (FIGURE2). These results provide further support for the idea thatdevelopmental defects in CdLS are caused by deregulatedtranscription and not by malfunction of cohesion-relatedprocesses. s

    RESEARCH HIGHLIGHTS

    SCIENTIFIC REPORT 2013 40SCIENTIFIC REPORT 2013 40

  • 5/19/2018 INFORME CNIO 3013

    43/248

    Deluca JG, Desai A, Diekmann S, Er-hardt S, Fitzgerald-Hayes M, Foltz D,

    Fukagawa T, Gassmann R, Gerlich DW,Glover DM, Gorbsky GJ, Harrison SC,Heun P, Hirota T, Jansen LE, KarpenG, Kops GJ, Lampson MA, Lens SM,Losada A, Luger K, Maiato H, MaddoxPS, Margolis RL, Masumoto H, McAinshAD, Mellone BG, Meraldi P, Musacchio A,Oegema K, ONeill RJ, Salmon ED, Scott

    KC, Straight AF, Stukenberg PT, SullivanBA, Sullivan KF, Sunkel CE, Swedlow JR,Walczak CE, Warburton PE, WestermannS, Willard HF, Wordeman L, Yanagida M,Yen TJ, Yoda K, Cleveland DW (2013).Esperanto for histones: CENP-A, notCenH3, is the centromeric histone H3

    variant. Chromosome Res2, 101-106. Lpez-Contreras AJ, Ruppen I, Nieto-

    Soler M, Murga M, Rodriguez- Acebes S,

    Remeseiro S, Rodrigo-Perez S, Rojas AM,Mendez J, Muoz J, Fernandez-CapetilloO (2013). A proteomic characterizationof factors enriched at nascent DNA mol-ecules. Cell Rep3, 1105-1116.

    Article in press

    Baquero-Montoya C, Gil-Rodrguez M,

    Teresa-Rodrigo M, Hernndez-Marcos

    M, Bueno-Lozano G, Bueno-MartnezI, Remeseiro S, Fernndez-HernndezR, Bassecourt-Serra M, Rodrguez deAlba M, Queralt E, Losada A, Puisac B,Ramos F, Pi J. Could a patient withSM