pdf LRI Scientific report 2014
Transcription
pdf LRI Scientific report 2014
LONDON RESEARCH INSTITUTE Scientific ReporT 2014 Cover images (from the top, left to right) Ernest Bashford OBE, ICRF Director 1902-1914. Signalling microcluster formation in B lymphocytes responding to membrane-tethered ligands (Image courtesy of F Batista). Image of stem cells forming a sprouting blood vessel (Image courtesy of H Gerhardt). Sir Michael Stoker, ICRF Director 1968-1979. Dendritic cells (Image courtesy of C Reis e Sousa). Sir Walter Bodmer, ICRF Director 1979-1991, ICRF Director General 1991-1996. Sir Paul Nurse, ICRF Director General 1996-2002. Fluorescence image of the paths taken by vacciniainduced actin tails over a 5 minute period (Image courtesy of M Way). Tomas Lindahl, Director of the CRUK Clare Hall Laboratories 1986-2005. Live zebrafish embryos expressing GFP under the control of the ptc promoter. Cells in green are those that are responding to Hh signalling (Image courtesy of P Ingham). Richard Treisman, Director of the CRUK London Research Institute 2002-2015. Electron Microscopy of B Lymphocytes (rounded) binding antigen on the surface of an antigenpresenting cell (Image courtesy of F Batista). John Diffley, Director of the CRUK Clare Hall Laboratories 2005-2015. Scanning electron micrograph of lung cancer cells (Image courtesy of A Weston). SCIENTIFIC REPORT 2014 Contents FOREWoRD5 Research at lri laboratories – An 7 historical perspective Origins: The Imperial Cancer Research Fund8 LRI Laboratories Highlights 1- 20 10-48 INTRODUCTION TO THE 2014 LRI RESEARCH REPORT50 52 Francis Crick INSTITUTE report 54 Research Highlights Martin R Singleton Macromolecular Structure and Function Thomas Surrey Lymphocyte Interaction Axel Behrens 68 Mammalian Genetics Dominique Bonnet 70 Haematopoietic Stem Cell Dinis Calado 72 Immunity and Cancer Julian Downward 74 Signal Transduction Holger Gerhardt 76 Vascular Biology Nathan Goehring 78 Developmental Systems Adrian Hayday 80 Immuno Surveillance Caroline Hill 82 Developmental Signalling Nicholas Luscombe Charles Swanton Translational Cancer Therapeutics Nicolas Tapon Cancer Epigenetics Sharon Tooze Richard Treisman Scientific report 2014 LONDON RESEARCH INSTITUTE 114 Signalling and Transcription Frank Uhlmann 116 Chromosome Segregation Peter Van Loo 118 Cancer Genomics Folkert van Werven 120 Cell Fate and Gene Regulation Michael Way 122 Cell Motility Research Groups – Clare Hall Simon Boulton Peter Cherepanov 90 125 126 128 Peter Karran 92 136 138 Mechanisms of Gene Transcription Stephen C West 98 134 Cell Division and Aneuploidy Jesper Q Svejstrup 96 132 Mammalian DNA Repair Mark Petronczki 94 130 Chromosome Replication 140 Genetic Recombination Hasan Yardimci Single Molecule Imaging 2 112 Secretory Pathways John FX Diffley Tumour Cell Biology Paola Scaffidi 110 Cell Regulation Architecture and Dynamics of Macromolecular Machines Immunobiology Erik Sahai Takashi Toda 88 Protein Phosphorylation Caetano Reis e Sousa Epithelial Biology Alessandro Costa Cell Cycle Peter J Parker 108 Chromatin Structure and Mobile DNA Structural Biology Paul Nurse / Jacqueline Hayles 106 Apoptosis and Proliferation Control 86 Tumour Host Interaction Neil Q McDonald 104 DNA Damage Response 84 Computational Biology Ilaria Malanchi 102 Microtubule Cytoskeleton Barry Thompson Research Groups – Lincoln’s Inn FieldS 65 Facundo D Batista 66 100 142 TECHNOLOGY CORE FACILITIES Ni 145 147 Advanced Sequencing Aengus Stewart 148 Paul Bates 149 Biomolecular Modelling Ruth Peat 150 Cell Services Lucy Collinson RESEARCH PUBLICATIONS AND THESES RESEARCH PUBLICATIONS THESES 163 164 186 INSTITUTE INFORMATION ADMINISTRATION ACADEMIC PROGRAMME SEMINARS AND CONFERENCES EXTERNAL FUNDING INSTI TUTE MANAG EMEN T CONTACT DETAILS 187 188 190 192 194 196 IBC 151 Electron Microscopy Graham MG Clark 152 Equipment Park Gordon Stamp 153 Experimental Histopathology Derek Davies 154 Michael Howell 155 High-Throughput Screening Daniel Zicha 156 Light Microscopy Nicola O’Reilly 157 Bram Snijders 158 Protein Analysis and Proteomics Svend Kjaer 159 P Ian Rosewell 160 Transgenics Ali Alidoust 161 Francois Lassailly 161 In Vivo Imaging Stephane Mouilleron 162 Protein Structure Anne Vaahtokari 163 CONTENTS 3 SIM image of the actin cytoskeleton (black) in Drosophila hemocytes lacking a functional WAVE complex. Original image: Sven Bogdan, University of Münster (DE) from a collaborative study with the Cell Motility Group. 4 Scientific report 2014 LONDON RESEARCH INSTITUTE FOREWoRD Harpal Kumar CEO CRUK When Sir Henry Morris, Thomas Rudd, and others decided, in 1902, to create an institute to research the causes and treatment of cancer, few would have imagined how profoundly important and influential that institute would become over the ensuing years. In its various guises and forms, what is now known as the London Research Institute of Cancer Research UK has contributed immeasurably to our understanding of the causes and development of cancer in humans. Through a multitude of landmark discoveries, made by the brightest and best scientists from around the world, the LRI has contributed to the steady progress we have seen in recent years in outcomes from cancer. Over that time, the LRI has become widely known as one of the very best biomedical research institutes in the world, and consistently ranks in the top five. This reputation has been propelled as much by a constant stream of outstanding talent as by its inspirational leaders, most latterly including the outgoing Director Richard Treisman, and one of his predecessors, Sir Paul Nurse, the first Director of the Francis Crick Institute. translation, are critical factors that have led Cancer Research UK to conclude that the time is right for a new institute. The Francis Crick Institute will build from the very best that LRI has learnt and delivered over the years. It will be a global beacon of excellence and a magnet for the very best young scientists from across the world. It will also inherit the best scientists who have made the LRI what it is today. It is a truism of curiosity-driven fundamental science that its directions and outputs cannot reliably be predicted. LRI scientists, although notionally working in a cancer institute, have made profoundly important discoveries in a broad range of other disease areas, including HIV/AIDS, other infectious diseases, diabetes, and neurological illnesses, amongst others. I would like to offer my deepest and heartfelt thanks to all those scientists, technicians, officers and support staff who have, over many decades, contributed to the development and outputs of this great institute, the flagship institute of Cancer Research UK. Furthermore, as we understand more about the drivers of cancer, we are increasingly conscious of how much we do not, as yet, understand. Therefore the need for an institute that carries out discovery-oriented research has never been stronger. However, scientific methods change, and the need for updated facilities, as well as the opportunities afforded by multi-disciplinarity, cross-disease working, and an increased focus on This scientific yearbook, the last for the LRI, captures the extraordinary work that its latest cohort of scientists have undertaken over the past 12 months, work that will go on to benefit humankind in the years ahead. It is a fitting finale to the LRI and its predecessors, and an enticing taster of what we can expect from the Francis Crick Institute. Harpal Kumar March 2015. FOREWoRD 5 1963 1986 6 Scientific report 2014 LONDON RESEARCH INSTITUTE RESEARCH AT THE LRI LABORATORIES – AN HISTORICAL PERSPECTIVE This Report marks the end of Cancer Research UK’s operations at the Lincoln’s Inn Fields and Clare Hall laboratories. We have therefore devoted its first section to a review of research highlights over the sites’ long history – Lincoln’s Inn opened in 1963, Clare Hall in 1986 – to illustrate the achievements and introduce researchers who have contributed to the LRI’s reputation as a leading cancer research Institute. Over the 50-year lifetime of the LRI’s laboratories, our understanding of cancer and how to treat it has undergone a revolution. We now have a broad understanding of many of the cellular control mechanisms that are disrupted in cancer, and increasingly have the ability to design exquisitely specific therapeutic reagents. Next-generation DNA sequencing has revealed the variety of genetic lesions in individual tumours, allowing cancer and its evolution to be characterised in ever-increasing detail, both during its development and its response to therapeutic attack. We can now appreciate the challenges posed by the extraordinary genetic diversity of the disease: each patient’s cancer comprises competing subclones with distinct genotypes that we need to identify, understand, and counter. Patient profiling by sequencing, and the developing ‘-omics’ technologies, promises new ways to classify individual cancers and to monitor their response to therapy, and continues to identify new routes for potential therapeutic development. Increasingly, molecular analysis is complementing traditional pathology-based tools in diagnosis, with stratification of patients by cancer and birth genotypes guiding the use of molecularly-targetspecific molecular therapies. Alongside this, we are seeing improvements in our ability to image cells and tissues in vivo, enabling us to study how cells – both normal and cancerous – behave in the context of the living body. Nevertheless, in spite of our ability to identify targets for therapeutic interference, in most cases we do not yet know how to strike them effectively. We must do so in such a way that cancer cells are killed but their normal neighbours are spared, and that the evolutionary avenues through which therapeutic resistance develops are closed off: as yet, however, we simply do not understand cells, tissues and their cancerous development well enough. Moreover, our understanding of how cancers interact with host physiology in general, including the immune system, remains in its infancy, despite recent therapeutic advances. The Francis Crick Institute will provide an unparalleled environment to take cancer research to a new level, integrating interdisciplinary discovery research with the clinical and translational opportunities available through its university partners. This retrospective, written by Kathleen Weston, starts with a brief introduction to the former ICRF, the predecessor of Cancer Research UK, and follows with a set of twenty Highlights of research achievements from the LRIs laboratories at Lincoln’s Inn Fields and Clare Hall. More detail can be found in Jane Austoker’s ‘A History of the ICRF 1902-1986’ (Oxford Science Publications) and Kathleen Weston’s ‘Blue Skies and Bench Space’ (Cold Spring Harbor Laboratory Press). LRI Laboratories Highlights 7 Origins: The Imperial Cancer Research Fund 1901: “Little is at present known of cancer, but as an incurable disease” Cancer has been a scourge of humanity for thousands of years, but at the dawn of the 20th century, almost nothing was known about the cause or causes of the disease. Doctors were able to classify tumours by their appearance, and could relate appearance to clinical outcome, but treatments, where they existed, were brutal, frequently agonisingly painful, and generally useless. Cancer was seen as inexorable, relentless, and progressive, appearing almost at random and defying the restraints of normal growth control mechanisms. The advent of experimental microbiology in the 19th century led to a change in the nature of cancer research. The notion of the ‘cancer cell’, which multiplied to create tumours, was born, and it became possible to start to speculate about how cancers arose. Bacteriological research, showing that it was possible to transmit infectious diseases such as tuberculosis between animals, led to successful attempts to propagate tumours in mice and rats by analogous methods. Using such tumours as a source of material, it was now possible to start asking systematic scientific questions about the origins of cancer. Dr Ernest Bashford 8 With the methods and materials now at hand, research laboratories dedicated to the study of cancer emerged, with the first small laboratory opening in Buffalo, New York, in 1897. In Britain, an anonymous letter appeared in the St James’s Gazette in 1901, advocating that a subscription fund should be set up to found a “cancer klinik”, dedicated to studying all aspects of the disease with a view to finding a cure. The writer of the letter was revealed to be Thomas Rudd, a wealthy businessman who had made a fortune in South African mining ventures. Although Rudd became seriously ill, and died in early 1902, his idea was taken up by Sir Henry Morris, a prominent and very well-connected surgeon. Morris persuaded the Royal Colleges of Physicians and Surgeons to provide laboratory space, and a campaign to collect subscriptions for the new Cancer Research Fund was launched in The Times, attracting Scientific report 2014 LONDON RESEARCH INSTITUTE high-profile support from the Royal Family (the Fund was renamed the Imperial Cancer Research Fund in 1904), the traditional aristocracy, and the new monetary aristocracy of the City. In October 1902, the Executive Committee of the newlyfounded charity made one of the most important and far-sighted appointments in the entire history of the ICRF, by hiring the 29-year-old Dr Ernest Bashford as the first General Superintendent and Director. Ernest Bashford and the start of research at ICRF Ernest Bashford, an almost complete unknown, was on the face of it an unlikely choice by the rather crusty Executive Committee, composed as it was of surgical grandees and Edwardian worthies. His energetic, no-nonsense personality springs out from his Director’s summaries of research in the early ICRF Annual Reports, otherwise rather dusty documents. His manifesto, a ‘Scheme for Enquiring into the Nature, Cause, Prevention and Treatment of Cancer’ is a model of how to approach a major new research area, and was comprehensive, innovative, and multi-disciplinary. In the words of his contemporary Archibald Leitch, Director of what is now the Institute of Cancer Research: “As a Comet suddenly appears out of space, blazes gloriously across the sky, and fades rapidly out of sight, so did that forceful, brilliant and wayward personality impress us”. Bashford in turn made an inspired hiring decision in taking on James Murray, who was to succeed him as the second Director in 1914, and the two of them set up together in the new laboratories. Bashford set about turning what had been a haphazard, antediluvian discipline into a modern scientifically based venture. Using a network of suppliers comprising helpful vets, zoos, the more enlightened hospital cancer wards, and most spectacularly, the Medical Officers scattered throughout India and the British Colonies and Protectorates, he started to collect and classify cancer samples from every source he could find. In a few short years, he and his small group of colleagues debunked many theories, finding cancers in every vertebrate species, and in every nationality. They showed that cancer was not infectious, as had been feared, and by rational observation, demolished many claims of miracle cures. However, all this intensive work made one thing alarmingly clear: cancer was not, as had been hoped, a single disease triggered by one or a few factors, but a many-headed hydra, with multifactorial causes, and a huge multiplicity of subtypes. The optimism of the founders of the Staff of the ICRF 1909. Front row (l-r): W H Bowen, B R G Russell, M Haaland, E F Bashford, J A Murray, C Da Fano, F Medigrazianu, W H Woglom. Middle row: W J Milton, E G Miller, M Finerty, F G Hallett, Miss Anderson, A J Hall, A Chapman. Back row: Mrs Tuohy, W J Dunn, H L Smith, C Trott, H Thatford, L Sawer, A Storey, A J Sheene. Cancer Research Fund, who had hoped for a cure within a few years, changed to a grim resolve in the knowledge that many years of research would be required to achieve any sort of progress. Furthermore, decisions had to be made to prioritise certain areas. Bashford and Murray believed strongly that cancer had to be understood to be cured, and the only way to do this was by laboratory-based basic research. The focus of the ICRF changed, moving further away from the clinic towards finding out what caused cancers, what made them grow, and what inherent weaknesses might be exploited to kill them. Over the next decades, the small band of ICRF scientists, still numbering less than 10, produced a solid body of research into tumour biology, and in the fields of neoplasia and radiobiology, they made crucially important contributions. The foundations of modern cancer biology lie in these and other early experiments done in a handful of pioneering labs around the world. Neoplasia: cancers arise from normal cells, in stages The concept that neoplasia, the growth of a new cancer, begins by mutation of normal cells, and is a multistep process, arose out of studies in a number of labs in the first half of the 20th century, although it was not until the 1970s that the scientific community finally accepted the idea wholeheartedly. Magnus Haaland, a Norwegian working at the ICRF was one of the first to recognise, in 1911, that “the malignant transformation of tissue may take place by degrees and not necessarily in one step from the normal cell to the fully developed cancer cell” . However, his observations were so ahead of their time that they were forgotten for many years. Haaland aside, the ICRF made its most important contribution to early neoplasia research with the work of Leslie Foulds, a pathologist appointed by James Murray in 1929 to what must have been one of the first postdoctoral fellowships ever offered. Foulds’s temporary position was converted to a permanent one the following year, and he stayed at the ICRF until 1951. Foulds, building on the work of Haaland, Peyton Rous and others, was the first to clearly lay out the principles of tumour progression. He was the first to realise that the mixed populations of cells found in tumours were the outward sign of a dynamic progression from normality to cancer, and that the stages of progression were qualitatively different, with cells mutating through precancerous stages to become increasingly invasive and metastatic. His book ‘Neoplastic Development’ (1969) is a classic, describing his many decades’ worth of observations on mouse mammary neoplasia, and is still widely cited in today’s textbooks. Radiosensitivity of tumours Circumstantial evidence that ionizing radiation might cause cancer began to accumulate in the early years of the 20th century, and the study of the effects of radiation on rodents, and in the newly developed tissue culture systems, became a worldwide theme of early cancer research. William Cramer, a German immigrant, was the second scientist hired by Ernest Bashford, in 1903. He remained at the ICRF, with one short break, until 1939. In the 1930s, together with Herbert Crabtree, who had joined the ICRF in 1923, Cramer carried out a series of classic experiments looking at the radiosensitivity of tumours, and how ionizing radiation might be used as a therapeutic. Crabtree and Cramer’s observations, that the radiosensitivity of malignant cells was not a fixed inherent property, but varied within wide limits, and that, in conditions of low or absent oxygen, cells become radioresistant, were two of the most significant to emerge from the ICRF in the inter-war period. Their work stimulated a flurry of radiobiological research in the 1930s and 1940s, which has led directly to the development of high energy radiotherapy, still the treatment of choice for many cancers. Key references J Austoker (1988). A History of the Imperial Cancer Research Fund 1902-1986. Oxford Science Publications E F Bashford (1908). Draft Scheme for Enquiring into the Nature, Cause, Prevention and Treatment of Cancer. Scientific Report of the ICRF 3:441-7 LRI Laboratories Highlights 9 LRI Laboratories Highlight 1 DNA and RNA tumour viruses 1968: Sir Michael Stoker brings virology to ICRF. In the following decade the ICRF becomes a world-leading centre for tumour virus research. Background In the late 1960s, the ICRF, whilst financially viable and scientifically solid, was no longer the cuttingedge institution it had once been. Conflict between the fifth Director, Guy Marrian and the ICRF’s governing Council, mainly regarding scientific objectives, resulted in Marrian’s forced early retirement in 1968. The Council appointed Sir Michael Stoker as the new Director, ushering in a revolution in the ICRF’s scientific structure and direction which restored the ICRF to its former status in a remarkably short time. Stoker swept out the old hierarchical system, replacing it with a flat structure based on small independent research labs, and transformed the science by investing heavily in the new and exciting field of the molecular biology of tumour viruses. In 1911, Peyton Rous had shown that the chicken virus which today, as Rous Sarcoma Virus, bears his name, could be used to infect birds and cause cancer. Subsequently Shope demonstrated that rabbit papillomas (warts) were also likely transmitted by a viral agent. There was a flurry of interest in virally-mediated cancers, but due to various reasons the field fell into disrepute for some decades. Nevertheless by the early 1960s, the idea that viruses caused cancer in many animal Beverly Griffin 10 Scientific report 2014 LONDON RESEARCH INSTITUTE species was generally accepted and virology became an important discipline in cancer research. At the ICRF, tumour viruses became tools to try to understand the mechanisms by which a normal cell could turn into a tumour. Experiments were mainly done in tissue culture (‘in vitro’), using the new techniques developed by the fledgling molecular biology field to look at the behaviour of the DNA, RNA and proteins in cells. Almost all the equipment and reagents were laboriously home made, and methods were long-winded, temperamental and frequently quite dangerous. However, they opened a window into a sub-microscopic biological world where no-one had previously explored. The research The ICRF labs began studying two types of tumour virus: retroviruses, where the genetic material was RNA, and those with a DNA viral genome. Both camps were very successful. The retrovirologists, under Robin Weiss, John Wyke and Steve Martin, worked extensively on the mechanisms by which RNA viruses were able to infect cells, but their major contributions were probably in the study of how retroviruses cause cancer. In two instances, the discovery of the Fos oncogene, and how retroviruses work as insertional mutagens, ICRF scientists achieved world firsts, and these are described in more detail in Highlights 5 and 6. The DNA tumour virologists, led by Lionel Crawford, Mike Fried, Beverly Griffin, Bob Kamen and Alan Smith, not to be outdone by their colleagues, also led the world. They discovered the p53 protein (Highlight 4), the Guardian of the Genome, whose mutation we now know is a crucial factor in the development of the majority of human cancers, and showed that p53 overexpression was characteristic of transformed cells. Ed Harlow, an American PhD student with Lionel Crawford, later used the methodology he learnt in the Crawford lab to show in 1988 that the E1A transforming protein from another DNA tumour virus, adenovirus, bound and inactivated the Retinoblastoma (Rb) tumour suppressor protein, which is intimately involved in cell cycle control. Transcription is the process whereby the DNA of the genome is copied into RNA, which is then used to make protein. It is responsible for turning the DNA blueprint into the reality of a complete organism, and as might be expected, is hugely complicated. Today, we are still in the throes of working out the control mechanisms that determine which gene is transcribed when and where, with layer upon layer of regulation still being revealed. In the 1970s, the idea that “DNA makes RNA makes protein” had only been accepted for a few years, and most of what was known about transcription came from simple bacterial systems. Small DNA tumour viruses were seen as a way in to studying eukaryotic transcription. Their small, circular genomes, containing only a few genes, made them easy to purify away from the huge mammalian chromosomes in whose cells they lurked, and they were lytic, meaning that they replicated rapidly and prolifically, to give tractable amounts of DNA: in the primitive world of early molecular biology, they were incredibly useful reagents. At the ICRF, two viruses, polyoma and SV40, were studied extensively, and together with their close colleagues at Cold Spring Harbor Laboratory in the US, ICRF scientists were responsible for laying down some of the fundamental principles of eukaryotic transcription regulation. The work at ICRF determined the complete sequence of polyoma virus, the structures of its transcripts, and the proteins it encoded – only the second complete structure of a DNA virus to be determined. It was shown for the first time that it was possible for two or more different mRNAs, and hence proteins, to be made from the same piece of DNA. Different transforming functions for the overlapping genes of polyomavirus were identified, and they were shown to work cooperatively in cellular transformation, setting the scene for subsequent discoveries about oncogene cooperation (Highlight 6). In SV40, similar work led to the discovery of p53 (Highlight 4). Turning their attention to how transcripts were initiated on the DNA, the ICRF scientists showed that transcripts could have multiple start sites, and that there was a faraway regulatory element in the SV40 DNA which was needed for correct expression of the mRNA; this was the first indication that regulation of both transcription and DNA replication could be facilitated by remote DNA sequences. The SV40 enhancer, as the regulatory element came to be called, is the archetype of a universal control mechanism. The consequences These experiments were of enormous significance both for cancer biology and transcription. They form the bedrock of our understanding of both subjects, and are so embedded in the consciousness of modern scientists that it is hard to imagine a time without them. Where are they now? The roll call of leading scientists who have worked in tumour virology at the ICRF is very long. Confined solely to Institute Directors and/or Nobel Prize Winners, the list runs as follows: Renato Dulbecco received the Nobel Prize in 1975; David Glover became first Director of the MRC Laboratory of Molecular and Cell Biology, University of London. Yoshiaki Ito is Yong Loo Lin Professor & Director, Oncology Research Institute, Singapore. Sir David Lane is Scientific Director of the Ludwig Cancer Research and Chief Scientist, A*STAR Institute, Singapore. Ron Laskey is the Charles Darwin Chair of Animal Embryology in the University of Cambridge, former Director of the MRC Cancer Cell Unit in the Hutchison/MRC Research Centre, and former Joint Director of the Wellcome CRC Institute, Cambridge; Frank McCormick leads the NCI RAS initiative and was formerly Director of the UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, USA. Tony Pawson was a former Director, Samuel Lunenfeld Research Center, Mount Sinai Hospital, Toronto, and winner of the Kyoto Prize. Sir Bruce Ponder became the first Director of the CRUK Cambridge Institute; Alan Smith became Chief Scientific Officer, Genzyme Corporation, US; Richard Treisman became Director of the LRI and a Research Director at the Francis Crick Institute; Harold Varmus (Nobel Prize, 1989) became Director of the US National Institutes of Health, President of Memorial Sloane-Kettering Cancer Center, and is now Director of US National Cancer Institute; Robin Weiss, Professor of Viral Oncology, UCL, is a former Director of the Institute of Cancer Research, London; John Wyke is Director Emeritus of the Beatson Institute, Glasgow. LRI Laboratories Highlights 11 LRI Laboratories Highlight 2 Discovery of Fibronectin 1973: Richard Hynes and Nancy Hogg independently discover a protein, fibronectin, which is present on normal cell surfaces, but disappears when cells become cancerous. 2011: An entire field has grown around this discovery. The fibronectin family, and their partners the integrin receptors, turned out to be central to cell adhesion and migration, and many other processes. Numerous steps in the progression of cancer, including invasion and metastasis, involve altered adhesive properties of cells, and novel antiadhesive therapeutics are seen as a key weapon in the fight against cancer and other diseases. 12 Background In the early 1970s, before the advent of the molecular biology and genetics revolution that would transform biological research, almost nothing was known about the molecular structure of a cell’s surface. However, in cancer biology, there were hints that the surface of a cancerous cell was different from that of its normal counterpart in some way. Scientists reasoned that if they could find what this difference was, they would have the perfect marker for diagnosing cancer, and perhaps from there would be able to quickly find a cure. Richard Hynes, a postdoctoral fellow in Ian Macpherson’s Tumour Virology Laboratory, and Nancy Hogg, in Av Mitchison’s Tumour Immunology Unit at University College, both realised that work from other ICRF scientists studying DNA tumour viruses could lead them to the cancer cell marker. Their colleagues had seen that when normal cells growing in tissue culture were infected with tumour-causing viruses, they were ‘transformed’. The infected cells grew faster, heaped up into piles instead of growing in flat monolayers, required fewer nutrients, changed their appearance, and made tumours when injected into mice. It seemed very likely that some of these dramatic effects could be due to changes in what was on the surface of the transformed cells, and both Hynes and Hogg decided to find out what these changes might be. Nancy Hogg Richard Hynes Scientific report 2014 LONDON RESEARCH INSTITUTE The research Both researchers used a biochemical technique called lactoperoxidase labelling to tag cell surface proteins with radioactive iodine. They took sets of cell lines, some normal and some transformed, labelled them up, and separated the labelled protein products by size on fractionating gels. To their excitement, a very large protein was present in the normal cell controls but absent in the transformed cells. The protein was dubbed LETS (Large, External, Transformation-Sensitive) by Hynes, but was subsequently renamed fibronectin (from the Latin fibra meaning fibre, and nectere, meaning to bind or connect). The consequences Although the simplistic idea of a universally relevant cancer switch molecule was wrong, in the search for this holy grail, Hynes and Hogg were amongst the founders of a new field; the study of cell adhesion. Cell adhesion is essential in all multicellular organisms, determining how, when and where cells move, both during embryonic development and in normal and disease states. In cancer, the ability of a tumour cell to grow, invade new tissue, and metastasise all involve changes in cell adhesion, and increasing our understanding of this has been invaluable. Before fibronectin, nobody had realised the importance of the physical scaffolding, termed the extracellular matrix, in which most cells live. Fibronectin is part of this matrix, and studying its interactions with other proteins led to the realisation that all cells maintain an intimate, active conversation with their extracellular environment, and are exquisitively responsive to changes in it. Hynes and others went on to discover that fibronectin and other structural matrix proteins bind to the integrin family of receptors, which are of huge significance, as integrins are the main transmitters of signals between the matrix and the internal architecture and signalling systems of cells. Recently, interest in fibronectin has renewed, as certain members of the fibronectin family are found in large amounts in the tumour vasculature, the network of blood vessels which sustains growing cancers. Antibodies against these fibronectins have been used as anti-tumour and tumour imaging reagents, and understanding the role of fibronectins in tumour blood vessel development will be a key requirement in the development of anti-angiogenic cancer therapies. What happened next? Richard Hynes left the ICRF in 1975 to set up his own lab at the Massachusetts Institute of Technology (MIT), and has continued to work on cell adhesion, making seminal contributions to the field he founded. He is a former Head of the MIT Biology Department and Director of the Center for Cancer Research, and is now Daniel K Ludwig Professor for Cancer Research at MIT. He is a Fellow of the Royal Society, and a member of the American Academy of Arts and Sciences, the USA National Academy of Sciences and the Institute of Medicine. Awards include the Gairdner Foundation International Award, the Pasarow Medical Research Award, a Guggenheim Fellowship and a Research Career Development Award from the National Institutes of Health. He sits on the Board of Governors of the Wellcome Trust. Nancy Hogg moved from the Tumour Immunology Unit to the ICRF Lincoln’s Inn Fields site in 1982, and remained there as a Principal Scientist, running the Leukocyte Adhesion Laboratory until her retirement in 2011. She was elected to the Fellowship of the Academy of Medical Sciences in 2002 for her distinguished contributions to the field of leukocyte biology. Key references Hynes RO (1973). Alteration of cell-surface proteins by viral transformation and by proteolysis. Proc Natl Acad Sci USA. 70:3170-4 Hogg N (1974). A comparison of membrane proteins of normal and transformed cells by lactoperoxidase labeling. Proc Natl Acad Sci USA. 71:489-92 Hynes RO (2004). The emergence of integrins: a personal and historical perspective. Matrix Biology. 23:333-40 LRI Laboratories Highlights 13 LRI Laboratories Highlight 3 Personalising cancer medicine 1974: Mel Greaves realises childhood leukaemias can be classified according to their cell surface markers into different cancers of varying prognosis, work which revolutionised the diagnosis and treatment of childhood leukaemia and was the first step towards personalised cancer treatment. Background Mel Greaves trained as an immunologist with Ivan Roitt in the late 60s, and after some time at the Karolinska Institute in Sweden, and then at the National Institute for Medical Research, he wound up working in Av Mitchison’s ICRF Tumour Immunology Unit at University College London. He would probably have carried on as a basic immunologist had it not been for a life-changing event during his time there. In Mel’s own words: “I had a colleague working at Barts Hospital who took me round the wards at a time when my own children were three and four years old. I saw children the same age stricken with leukaemia and found it appalling. When I asked “What is leukaemia? What is the underlying problem here?” it was absolutely clear that ignorance was pretty rampant. We had no idea about the nature of the disease except that an expanding population of cells was damaging the bone marrow and children were dying. I felt that this must be a tractable problem, so I started asking simple biological questions such as “What sort of cell is involved?”” haemopoietic stem cell. Depending on cues they receive from their location in the body, the cells that surround them, and the demands made on them by external events such as infection, they mature and differentiate down particular lineages, leading to a multiplicity of cell types, each tailored for a particular purpose. Mel’s work in the ICRF Tumour Immunology Unit was on two of these cell types, the T and B white blood cells, which recognise and respond to infection as part of a normal immune response. As part of his work in classifying and studying the development and responses of T and B cells, Mel had developed a panel of antibodies, which could be used to specifically tag either type of cell, and could also distinguish between different stages of their development. By labelling his antibodies with a coloured chemical which fluoresced when hit by a laser beam, the cells binding the antibodies could be automatically detected and rapidly counted by a machine called a flow cytometer, one of the first commercial examples of which had recently been bought by the ICRF. The research All blood cells develop from a single cell type, the The most common type of childhood leukaemia is acute lymphoblastic leukaemia (ALL), which in the early 70s was classed as one homogeneous disease, with one treatment, a brutal sledgehammer of drugs and radiation, which effectively killed the cancer cells, but often had dreadful side effects including severe mental and physical impairment. From the viewpoint of a basic immunologist such as Mel, it seemed very obvious that the mutant ALL cells were likely to be derived from normal T and B cells, and that finding out how the cancers arose would be the key to studying and treating them. Most importantly, Mel was one of only two or three people in the world at the time who possessed the reagents (his antibody panel) and the technical know-how (his experience with flow cytometry) to do anything about this problem. Mel Greaves 14 Scientific report 2014 LONDON RESEARCH INSTITUTE Using his antibody panel, Mel and his colleagues began to analyse childhood ALLs, and as they had expected, found that they did not all look the same. They reclassified them into four subtypes: B-ALL; T-ALL; pro-B ALL, and mature B-ALL. By using their new classification to look at samples from ongoing clinical trials, they were also able to correlate the four types of ALL with different disease outcomes. The rarest subtypes, pro-B and mature B-ALL, had very poor prognoses. On the other hand, the commonest subtype, B-ALL, had a relatively good prognosis. Suddenly, a simple and rapid method existed to learn exactly which type of ALL a child had, and what their prognosis was. Each patient’s cancer could be treated in a targeted fashion, such that only the rare pro-B and mature B-ALLs with poor prognoses had to be hit hard, and the others could have less aggressive treatment. Leukemia Foundation. He is an EMBO member, an honorary MRCP, a Fellow of the Academy of Medical Sciences and a Fellow of the Royal Society. Mel’s most important work since leaving the ICRF has been to show that childhood leukaemia begins in the womb, and is a two-stage process. He realised that by studying pairs of identical twins, one with leukaemia and one without, he could look in the healthy twin for early precancerous changes. By this method, his lab showed that the initiating pro-leukaemic change takes place in the womb, and that due to shared placenta, identical twins both have pro-leukaemic cells. The lab extended this analysis to study how many babies were born with the initiating change, using blood extracted from blood spots stored on Guthrie cards (the blood samples taken at birth from all UK-born The consequences This work is a translational research classic – a basic babies). They found that 1 in 20 babies are born with a pre-leukaemic clone on board, but of these, biologist’s view of a clinical problem provided the new insight required for a fundamentally important only 1% go on to develop leukaemia. It is now accepted that childhood ALL develops in two change in diagnosis and treatment. Mel’s idea of stages. The first change, likely spontaneous, using panels of discriminatory antibodies coupled happens in the womb, and is frequent. The second to flow cytometry is now used throughout the change triggers development of leukaemia and world for the diagnosis and classification of happens in early childhood, with average age of leukaemia and many other cancers. The ICRF became the national immuno-diagnostic service for onset 2-5 years. This second change was suggested by Mel to be caused by an inappropriate response childhood ALL in the United Kingdom. Diagnosis of to infection, brought on by keeping babies in the ALL subtype meant that for the first time, developed world in surroundings that are too clean personalised cancer therapy, the dream of cancer – the hygiene hypothesis. The theory goes that the clinicians, was possible. immune systems of first world babies are not sufficiently exposed to pathogens, and therefore As well as their profound impact on the treatment are not properly educated in the vital months after of leukaemia, these experiments were the first birth. The unbalanced immune system may tentative steps taken into understanding the biology of childhood ALL. Once cancer cells could be respond in an exaggerated way to infection, triggering transformation of a pre-leukaemic cell categorised, they could be compared with normal into a leukaemia. blood cells, and the data Mel obtained suggested that leukaemias were a result of very immature Mel’s current research on the genetic blood cells somehow becoming arrested in their heterogeneity of cancer remains at the forefront of normal development. How this happens, what the leukaemic cancer stem cells are, and how the roots contemporary thinking about the causes and consequences of mutation in cancer, and how it can of the disease can be attacked and eradicated, are still the major preoccupations of today’s leukaemia be tackled therapeutically. His work in this field has influenced a new generation, including Charlie researchers. Swanton, whose work on tumour heterogeneity features in Highlight 20. What happened next? Mel Greaves left the ICRF in 1984 to become Director of the Leukaemia Research Fund Centre at Key references Brown G, Greaves MF, Lister TA, Rapson N, the Institute of Cancer Research. He remains there Papamichael M (1974). Expression of human T and as Professor of Cell Biology. For his work on B lymphocyte cell-surface markers on leukaemic leukaemia, Mel has won numerous awards and cells. Lancet. 7883:753-5 prizes, including the King Faisal International Prize Greaves MF (2008). A Scientist’s Journey. From for Medicine, the Gold Medal of the British Society of Haematology and the Jose Carreras Award of the White Blood: Personal Journeys with Childhood Leukaemia. ISBN: 978-981-279-039-2 European Hematology Association/Jose Carreras LRI Laboratories Highlights 15 LRI Laboratories Highlight 4 Discovery of p53 1979: David Lane and Lionel Crawford show that the oncogenic viral SV40 large T antigen binds a cellular protein, dubbed p53. p53, the ‘guardian of the genome’, is mutated in the majority of human cancers. Restoring normal p53 activity is the holy grail of cancer therapeutics. Background In the mid 1970s, the ICRF had entered a period of pre-eminence in virological research (Highlight 1). Part of this endeavour was the work on the DNA tumour virus, SV40. RNA tumour viruses, or retroviruses, had been shown in 1976 by Varmus and Bishop to contain genes, dubbed oncogenes, which were hijacked, mutant versions of cellular genes; when unleashed on a cell by viral infection, the oncoproteins encoded by the mutant oncogenes took over the cell’s metabolism, allowing it to grow uncontrollably. However, the oncogenes of DNA tumour viruses bore no relationship to cellular genes, and their tumourigenic mechanisms were a mystery. 16 three viral oncoproteins, unoriginally named large T-, small T- and middle T-antigens had been found; it seemed possible that SV40 might also have a hitherto undetected middle T-antigen, and many labs, including Lionel Crawford’s at the ICRF, began work to identify this putative new oncogene. David Lane arrived as a postdoc in the Crawford lab in 1975, fresh from a PhD with Av Mitchison at University College London, and his skills as an immunologist meant that he was ideally suited to a project involving the use of antibodies to detect novel, elusive proteins. Upon detecting infection by a DNA tumour virus, the host organism’s immune system was known to react by making antibodies against the viral proteins. Scientists realised that this antigenic (‘antibody-making’) response could be used as a tool to identify and monitor the viral proteins (dubbed ‘viral tumour antigens’) in cells. Conveniently, genetic studies showed that these viral tumour antigens were often encoded by the genes responsible for the oncogenic potential of the virus. In the case of SV40, the two viral proteins identified like this were called large T- and small T-antigen. For polyoma virus, one of the other DNA tumour viruses under intensive study at the time, The research As the antisera available at the time for detecting the SV40 tumour antigens were of extremely variable quality, David, in collaboration with research assistant Alan Robbins, spent the first part of his postdoc making antibody reagents that were specific for large and small T, no small feat at that time. He then used these improved antisera for immunoprecipitation experiments in SV40transformed cells. Immunoprecipitation is molecular fishing, using an antibody able to specifically recognise a target protein as the hook to fish out the protein of interest from the cellular soup. David saw, sure enough, that his antisera specifically brought down both large and small T, but in addition, he always saw a third protein, which he estimated to be about 53 kilodaltons (kD) David Lane Lionel Crawford Scientific report 2014 LONDON RESEARCH INSTITUTE in size. The mystery protein never appeared in his immunoprecipitates except in association with large T, but was not virally encoded, and was not a shortened, degraded version of either large or small T. David was left with no option but to suggest that the 53kD protein was virally induced, but cellular in origin, and that it was being immunoprecipitated because it was in a complex with large T antigen. He presented compelling data in his paper to indicate this was the case, and also showed that a protein, probably the same one, could also be found at high levels in Polyoma transformed cells. The fact that this same protein was involved in transformation by two separate tumour viruses led Lane and Crawford to propose that it was likely to play a crucial role in tumour formation. As is often the case when a discovery is ripe to be made, it transpired that four other groups had essentially the same observations; papers by Lane and Crawford, Pierre May’s group in France, Alan Smith’s group in London, and Linzer and Levine and the Carroll group in the US, were all published in 1979. The importance of p53 in cancer was underscored by another publication from the Crawford lab in 1982, showing that serum from 9% of all breast cancer patients contained antibodies that targetted p53. We now know this was the first hint of the avalanche of cancers in which p53 is found mutated. The consequences The first decade of p53 research was fairly confusing due to its initial categorisation as an oncogene in its own right. However, after the p53 gene was cloned from multiple cell lines, the field realised that all the oncogenic versions were actually mutated, and that normal p53 was in fact a tumour suppressor gene. Therefore, rather than p53 causing cancer, loss or mutation of the p53 gene was realised to be one of the contributory factors in tumour formation. In fact, p53 turns out to be possibly the most frequently mutated gene in human cancer, with functional changes found in the majority of all cases. More recent work has shown that p53 is a transcription factor, able to directly switch genes on and off, and that it is made by cells when they detect they have been damaged. In 1992, it was dubbed the ‘guardian of the genome’ by David Lane, due to this ability. Induction of p53 can halt the cell cycle, so damaged DNA can be repaired, or if the damage is too bad to be fixed, p53 forces the cell to commit suicide (apoptosis), or to become dormant (senescence). Its pivotal role in guarding against damage explains why it is so often put out of action during the evolution of a cancer, which can only grow if it overrides the normal cellular defence mechanisms. What happened next? David Lane remains one of the world’s most prominent cancer biologists, and was the second most highly cited medical scientist in the UK in the last decade. He has published in excess of 300 papers, and co-authored a successful immunochemical methods handbook, ‘Antibodies’, which has sold more than 40,000 copies. For his efforts in cancer research, he was knighted in 2000, and he has won many awards such as the Paul Ehrlich Prize in 1998, the Buchanan Medal in 2004, the Medal of Honor from the International Agency for Research on Cancer in 2005 and in 2008, the Royal Medal from the Royal Society of Edinburgh. He is a member of the European Molecular Biology Organisation (EMBO), a Fellow of the Royal Society and the Royal Society of Edinburgh, and a founder member of the Academy of Medical Sciences. Sir David was the first Chief Scientist of Cancer Research UK, leaving that position in 2010. After many years working in Dundee, latterly as Director of the Cancer Research UK Cell Transformation Research Group and Professor of Oncology, he is currently Chief Scientist of A*STAR in Singapore, and Scientific Director of Ludwig Cancer Research. Lionel Crawford worked at the ICRF labs in London until 1988, when he moved to run the ICRF Tumour Virus Group in the Pathology Department of the University of Cambridge until his retirement in 1995. He was elected a Fellow of the Royal Society of Edinburgh in 1970 and a Fellow of the Royal Society in 1988. In 2005, in recognition of his lifetime’s work on DNA tumour viruses, he was awarded the Gabor Medal of the Royal Society. Crawford was not only an internationally recognised leader in his field, but was also much appreciated for his extraordinarily unselfish attitude to nurturing younger scientists in his laboratory, many of whom besides David Lane went on to become ‘big names’ in their own right. In the words of a former colleague, he is “one of the unsung heroes of British biochemistry and molecular biology”. Key references Lane DP, Crawford LV (1979). T antigen is bound to host protein in SV40-transformed cells. Nature. 278:261-3 Crawford LV, Pim DC, Bulbrook RD (1982). Detection of antibodies against the cellular protein p53 in sera from patients with breast cancer. Int J Cancer. 30:403-8 Lane DP (1992). p53, Guardian of the Genome. Nature. 358:15-16 LRI Laboratories Highlights 17 LRI Laboratories Highlight 5 Discovery of Fos oncogene 1982: Tom Curran and Natalie Teich identify the v-Fos retroviral oncoprotein and associated protein p39 (later shown to correspond to the Jun oncoprotein). With US collaborators they clone and sequence the v-fos gene and show there is a cellular homologue, c-fos. Fos and its partner Jun were the first oncoproteins shown to directly regulate transcription, the switching on and off of genes. They are now known to participate in controlling growth, differentiation, cell death and activation of neurons. Their discovery led to the realisation that upregulation and downregulation of specific crucial target genes can cause cancer, and that all parts of the cellular machinery are vulnerable to cancer mutations. Background Peyton Rous’s 1911 isolation of a cancer-causing (oncogenic) tumour virus won him the 1966 Nobel Prize in Physiology or Medicine, and led to the identification of many other retroviruses with the same ability to cause cancer. In 1976, Harold Varmus and Mike Bishop made another Nobel Prize-winning discovery, that Src (pronounced ‘sarc’), the cancer causing oncogene in Rous Sarcoma Virus, had a cellular counterpart, the c-Src proto-oncogene. Varmus and Bishop’s work demonstrated that retroviruses could steal the genetic matter of their hosts, and mutate it into a superactivated form, which could then attack the host, causing uncontrolled proliferation, and hence cancer. Crucially, it also gave researchers a way in to the complex problem of how to detect other sleeping proto-oncogenes in the genome. Tom Curran 18 Scientific report 2014 LONDON RESEARCH INSTITUTE Post-1976, there was a flood of research to analyse the oncogenic proteins being made by retroviruses, and to try to match them to proteins in the normal genome. At the ICRF, Tom Curran, a young Scottish student in Natalie Teich’s lab, was given as his PhD project the task of finding the oncogenic component of the FBJ mouse osteogenic sarcoma retrovirus. The research Tom’s initial approach was to isolate the oncogenic part of the FBJ retrovirus by making transformed rat cells that were not also infected with a helper retrovirus (so-called non-producer cells). He then injected these cells under the skin of rats where they grew to form tumours. The rats made antibodies against the tumour cells in a vain attempt to stop them growing. Tom then took blood samples from the rats and used the sera to examine cells infected with the FBJ retrovirus using the technique of immunoprecipitation, in which an antibody in the sera is able to specifically recognise and capture proteins from the general cellular soup. Tom made a stock of antiserum (a mix of many antibodies) made from an animal inoculated with cells containing FBJ, together with some matching control antiserum from a normal animal. The FBJ-specific antiserum contained antibodies against FBJ proteins, whilst the control did not. When he used these two antisera to analyse multiple cell types, some infected with FBJ and some not, he was able to show that there were two specific and unique proteins in the FBJ-infected cells, one 55 kilodaltons (kD) in size and one 39 kD in size, termed p39. To find out which of these proteins was made by the FBJ virus, Tom purified viral RNA and used it to program a cell-free translation system that made all the proteins encoded by the virus. Remarkably, sera from rats with tumors specifically recognised and captured just the 55 kD protein showing that it was the product of the FBJ oncogene. Tom christened the new oncogene v-fos, (from FBJ-osteogenic sarcoma virus). Shortly before moving to Inder Verma’s laboratory at the Salk Institute in San Diego for postdoctoral work, under the tutelage of Gordon Peters (Highlight 6),Tom isolated and purified v-fos DNA. The collaboration continued after Tom moved to San Diego, ultimately demonstrating, that, like all other retroviral oncogenes, v-fos was a stolen and modifed copy of the proto-oncogene c-fos, found in the normal genome. The consequences In 1984, Tom established an independent laboratory at the Roche Institute of Molecular Biology. In an early collaboration with Rolf Müller and Rodrigo Bravo at the EMBL in Heidelberg, Tom showed that Fos was rapidly and transiently induced (turned on) when cells are exposed to growth factors. This work established Fos as an archetype of the set of genes known as cellularimmediate-early genes that function in signal transduction processes in response to extracellular stimuli. Furthermore, Tom, together with his long-standing collaborator Jim Morgan, showed that this response also happened when neurons are activated by neurotransmitters or electrical stimulation. This finding led to the model that changes in gene expression may underlie the long term adaptive modifications in neurons that contribute to learning and memory. In 1987, Jun, another retroviral oncogene which encoded a 39kD protein, was discovered by Peter Vogt’s lab. Jun protein was shown to be a component of the AP-1 transcription factor complex. AP-1 was at that time a very hot research topic in the transcription field, but had hitherto only been defined as a protein complex of unknown makeup which, in response to growth factor signals, could bind to DNA and activate neighbouring genes. Tom’s lab, in collaboration with Bob Franza at Cold Spring Harbor, showed that Fos also bound to the AP-1 DNA binding site. Shortly afterwards, in one hectic period spanning a few weeks in 1988, they went on to show that the mysterious 39kD cellular partner of Fos which Tom had defined six years before at ICRF, was actually the Jun protein, and that Fos was the other component of AP-1. These two oncogenes were the first to be identified as transcription factors, able to directly regulate gene transcription, and in their viral form switching on genes that would only normally be activated by strong growth signals. In addition to this fundamental insight into cancer biology, a flood of research into AP-1 in the next few years established many key concepts in transcription factor biology: the leucine zipper, a novel structural motif which zips together the Fos and Jun proteins to make the AP-1 complex, is now known to be ubiquitous, and the existence of Fos and Jun related proteins which can be mixed and matched showed that transcription factor families exist, and shed light on an extra level of regulation. Perhaps most importantly, the study of how AP-1 is regulated by growth factor signals, and how the complex can be modified to tune its activity precisely, has been in the forefront of research into understanding how a cell’s reaction to its environment is translated into a change in transcription of its genes, and hence its destiny. What happened next? Tom Curran is currently Deputy Scientific Director of the Children’s Hospital of Philadelphia Research Institute. He has won numerous awards, and is a past President of the American Association for Cancer Research. He was elected to the Royal Society in 2005 and the Institute of Medicine (IOM) of the National Academies of Science, USA in 2009. Natalie Teich retired from the ICRF in 1995 after 22 years as a lab head. Although her co-discovery of v-fos was her most significant contribution to cancer research, her work on blood cell development, exploring the potential of stem cells, was also influential. Key references Curran T, Teich NM (1982). Candidate product of the FBJ Murine Osteosarcoma Virus Oncogene: Characterization of a 55,000-Dalton Phosphoprotein. Journal of Virology. 42:114-22 Curran T, Peters G, Van Beveren C, Teich NM, Verma IM (1982). FBJ Murine Osteosarcoma Virus: Identification and Molecular Cloning of Biologically Active Proviral DNA. Journal of Virology. 44:674-82 Rauscher FJ 3rd, Cohen DR, Curran T, Bos TJ, Vogt PK, Bohmann D, Tjian R, Franza BR Jr (1988). Fos-associated protein p39 is the product of the jun proto-oncogene. Science. 240:1010-6 LRI Laboratories Highlights 19 LRI Laboratories Highlight 6 Insertional mutagenesis and oncogene cooperation 1983: Gordon Peters and Clive Dickson show MMTV causes tumours by insertional mutagenesis. 1986: Peters, Lee and Dickson discover first evidence of oncogene cooperation in an animal model. Both these discoveries form part of the conceptual foundations of our current understanding of cancer. Background Cancer-causing retroviruses are not all alike; in addition to those that have picked up a cellular gene and corrupted it into becoming an oncogene, there exists a second type. These other retroviruses still cause cancer in almost 100% of the infected hosts but do not contain an oncogene in their genomes. Until the 1980s, researchers were mystified as to how they might work, but in 1981, Bill Hayward, at the Rockefeller Institute in New York, provided one possible answer, when he solved the puzzle of how Avian Leukosis Virus (ALV) was able to cause leukaemia. The answer lies in the life cycle of a retrovirus. As the name implies, retroviruses reproduce by copying the information in the viral RNA into double-stranded DNA, the exact opposite of the normal process of gene expression. The resulting double-stranded DNA ‘provirus’ is then inserted into the host cell’s DNA, becoming a passenger in the genome that is copied each time the cell divides. Each infected cell produces large amounts of viral RNA and protein, packages it into new retroviral shells, and dispatches it into the extracellular soup to infect more cells. When Hayward examined where in the genome the ALV provirus had integrated in leukaemic cells, he Gordon Peters (left) and Clive Dickson (right) 20 Scientific report 2014 LONDON RESEARCH INSTITUTE found that it had inserted itself next to a growth controlling gene called c-Myc. c-Myc had already been identified as the oncogene captured by another virus, MC29, and it was later shown that many human tumours make too much (overexpress) c-Myc protein. This latter is exactly what happens when the ALV proviral DNA is inserted nearby. The viral DNA contains powerful transcriptional control elements that are designed to maximise viral RNA production, but their effects can often spread to host genes that happen to be in the vicinity of the provirus. If by chance the adjacent gene is a potential oncogene, such as c-Myc, its inappropriately high expression can enable that particular cell to outgrow its neighbours and develop into a tumour. Hayward christened this new method of tumourigenesis ‘insertional mutagenesis’. Clive Dickson and Gordon Peters, in Robin Weiss’s RNA Tumour Virus section at the ICRF, were studying a virus called Mouse Mammary Tumour Virus (MMTV), then considered to be the best available animal model for human breast cancer. Like ALV, MMTV did not carry its own oncogene. Clive and Gordon realised that Hayward’s discovery of ALV causing cancer by insertional mutagenesis might also apply to MMTV - perhaps it, too, was integrating into the genome next to a potential oncogene and switching it on. If their hunch was right, it was likely that Hayward had discovered a general principle of viral tumourigenesis, and also, that new oncogenes of great importance in breast cancer might be discovered. The research Whereas the oncogene targeted by ALV was already known and easy to recognise as c-Myc, there were no obvious clues about the likely targets of MMTV. However, Clive and Gordon reasoned that if they found where the MMTV provirus had integrated in different tumours, they might find the same region of genomic DNA cropping up again and again next door to the provirus. The first step was to clone fragments of DNA that spanned the junctions between proviral and cellular DNA and to use these to prepare probes that would identify the site of integration within the genome. By using these probes to analyse the DNA from a series of MMTV-induced tumours, they found that in almost half of the tumours there was a provirus in the same region of the host genome which did not seem to correspond to a known oncogene. At the same time as this work was underway, Roel Nusse and Harold Varmus in San Francisco had been conducting similar experiments and discovered a different MMTV integration site, which they called int-1. Gordon and Clive’s potential oncogene was therefore dubbed int-2. Interestingly, at both common insertion sites, the MMTV proviruses were clustered on either side of a host gene that was switched off in normal mammary cells but switched on in tumour cells by MMTV. The following year, both groups succeeded in cloning the respective int-1 and int-2 genes and began to ask what these new oncogenes might do. One of the surprises of this research was that despite doing very similar experiments, the two groups had discovered completely different genes. Could there be a third or fourth gene waiting to be discovered? If so, the obvious place to look would be in tumours where int-1 and int-2 were not involved but, in searching for such an example, Gordon and Clive came upon another surprise. In many of the tumours they analysed, proviruses had integrated at both int-1 and int-2, not just one or the other, and both genes were turned on. This fitted well with the persuasive theory doing the rounds at the time that the transformation of a normal cell into a tumour cell requires the cooperative action of more than one oncogene. The idea was initially formulated from studies on mouse and rat cells grown in tissue culture, in which the ICRF DNA tumour virus groups had played a major role, but Gordon and Clive’s 1986 publication in Nature was the first to show that oncogene cooperation occurs in real tumours in an animal model. The consequences As a technique for probing the molecular biology of tumours, insertional mutagenesis has been invaluable, for a long time remaining the tool of choice for identifying new oncogenes, and also for examining which oncogenes are able to cooperate with each other. While in some cases there is a rational explanation for co-operativity (such as the role of c-Myc in apoptosis described in Highlight 10), the reason for co-activation of int-1 and int-2 remains unresolved. int-1 proved to be the prototype of the Wnt family of growth factors that have pivotal roles in development and cancer, and some other members of the Wnt family were later found to be activated by MMTV insertion. The int-2 gene product turned out to be a member of the Fibroblast Growth Factor family, and is now known as Fgf3. Interestingly, Fgf4 which is right next to Fgf3 in the genome, is also activated by MMTV, reinforcing the idea of co-operativity between the two families of growth factors. Following initial excitement that the human FGF3 and FGF4 genes are amplified (present in extra copies) in many human breast cancers, Clive and Gordon subsequently found that both genes remain switched off in these tumours. However, this observation suggested that there might be another previously unknown oncogene within the region that is co-amplified with FGF3 and FGF4, so they set about mapping the region of amplification. It quickly emerged that the likely culprit was cyclin D1, a pivotal regulator of the cell cycle. What happened next? Gordon Peters and Clive Dickson remained at the LRI and continued to work closely together for most of their careers. Gordon retired in 2013, and Clive in 2004. Both were elected Fellows of the Academy of Medical Sciences for their work in cancer biology and transcription. Key references Peters G, Brookes S, Smith R, Dickson C (1983). Tumorigenesis by Mouse Mammary Tumour Virus: Evidence for a Common Region for Provirus Integration in Mammary Tumours. Cell. 33:369-77 Dickson C, Smith R, Brookes S, Peters G (1984). Tumorigenesis by Mouse Mammary Tumour Virus: Proviral Activation of a Cellular Gene in the Common Integration Region int-2. Cell. 37:529-36 Peters G, Lee AE, Dickson C (1986). Concerted activation of two potential proto-oncogenes in carcinomas induced by mouse mammary tumour virus. Nature. 320:628-31 LRI Laboratories Highlights 21 LRI Laboratories Highlight 7 Growth factors and receptors can be oncogenes 1983: Mike Waterfield and colleagues show that the human gene encoding Platelet Derived Growth Factor (PDGF), and the cancercausing sis oncogene, found in a tumour virus, are closely related. 1984: Julian Downward discovers that another tumour virus has hijacked the human Epidermal Growth Factor Receptor (EGF-R) and converted it into the v-erb-b oncogene. These two discoveries showed that oncogenes can cause tumours because they encode mutationally activated components of normal cellular growth control mechanisms. The work transformed our understanding of how growth is regulated in normal cells, and what goes wrong in cancer. 22 Background The existence of soluble proteins that could stimulate cells to proliferate was first discovered in the late 1950s by Rita Levi-Montalcini, Giuseppe Levi and Stan Cohen. Levi-Montalcini and Cohen received the 1986 Nobel Prize in Physiology or Medicine for their discovery, which showed that the immensely complicated process of growing a whole organism from a single fertilised egg might be understood by looking at what was happening to individual cells at the molecular level. In the years that followed, many more growth factors, as the proteins came to be called, were discovered, and they were shown to be the drivers of normal cell growth, providing essential signals for growth, differentiation (the process of developing into functionally specialised cells) and eventually, death. By the late 1970s, whilst it was relatively easy to purify growth factors and find out what they did, it was much harder to identify them as specific genes; protein sequencing, the determination of amino acid sequence, was extremely difficult and costly, and databases containing sequence information (where one could check for potential sequence similarity to known proteins), were small and hard to access. Mike Waterfield, however, was in a very good position to tackle this problem. He had been a postdoc at Caltech in the lab of Leroy Hood, a protein sequencing pioneer, and when he set up his own lab at ICRF, he brought with him the expertise to build what was for a time the world’s most sensitive protein sequencer. He decided to attempt to sequence PDGF, a potent growth factor that stimulates wound healing. Mike Waterfield Julian Downward Scientific report 2014 LONDON RESEARCH INSTITUTE The research Waterfield obtained purified human PDGF from colleagues in Sweden and the US, and, using his state-of-the-art protein sequencer, obtained some partial amino acid sequences from the PDGF protein. After some fruitless searches of public databases, the PDGF sequence was compared to a tape of the most up-to-date database available, posted from San Diego by the evolutionary biologist Russell Doolittle. There was an immediate match, to the v-sis oncogene. V-sis, found in a tumour virus causing sarcomas, was therefore making a powerful growth factor normally only induced by wounding, which meant that any cell infected by its parent virus would immediately start proliferating abnormally, one of the classic hallmarks of cancer. At the same time, Julian Downward, a graduate student in Waterfield’s lab, and Yossi Yarden, a visiting Israeli scientist, with collaborators Axel Ullrich and Jossi Schlessinger, were working on a separate project, to determine the first ever full length nucleic acid sequence of a growth factor receptor. To do this, they purified human Epidermal Growth Factor Receptor (EGFR), obtained some partial amino acid sequences, and then used these sequences to make probes to fish out DNA corresponding to the coding region of the EGFR gene. Late in December 1983, Downward compared his EGFR sequences to an in-house database of oncogene sequences compiled by a postdoc, Geoff Scrace, and discovered that EGFR matched up perfectly with the viral oncogene v-erb-B, whose host virus causes acute leukaemia. Tumour viruses had therefore not only stolen growth factors, but also their receptors from the organisms in which they could cause cancer, equipping themselves with mutant versions of the most powerful weapons in a normal cell’s armoury. The consequences These findings were the first to definitively show at the molecular level that growth factor signalling was central to carcinogenesis. A new discipline, the study of signal transduction, exploded into life as many other researchers realised that it was possible to purify, sequence and identify not only a multitude of growth factors, but all the components of the chain of command stretching from the growth factor outside the cell right into the cell’s nucleus. In cancer, there are now many examples of mutated or overactive growth factors and receptors causing particular human tumours, and numerous drugs have been developed which target such molecules. Mutations in the EGFR have been found in lung, pancreatic and colorectal cancers amongst others, and a number of effective EGFR-inhibiting drugs are now in use in the clinic. One of the EGFR’s close relatives, HER-2, is overexpressed in about 30% of breast cancers, and is associated with poor prognosis, but again, herceptin, a HER-2 inhibitor, is now a standard and effective part of first-line breast cancer treatment. Tony Burgess, a long-time colleague of Mike Waterfield put these findings in this context: “All scientists strive to participate in a discovery that will change the direction of the world. Most of us work for a lifetime admiring the great achievements of a few of our colleagues and occasionally we brush with one great discovery. In the seven months from June 1983 to January 1984, Mike Waterfield led two great teams which changed our understanding of cancer biochemistry and biology.” What happened next? Mike Waterfield moved from ICRF in 1986 to head the London University College branch of the Ludwig Institute for Cancer Research. In 1991, he became the Courtauld Professor of Biochemistry and Molecular Biology at UCL, and from 1991 to 2002, was the Head of UCL’s Department of Biochemistry and Molecular Biology. Prof. Waterfield has also received numerous academic awards, including being made a Fellow of the Royal Society. His work post-ICRF centred around the study of a family of enzymes, the phosphoinositide-3 kinases (PI3Ks), which are downstream components of many growth factor signalling pathways. Alterations affecting PI3K signalling are found in many cancers, and in 2002 Waterfield and two Cancer Research UK-funded scientists, Peter Parker and Paul Workman, set up a company, PIramed, to develop PI3K-targeted therapies for cancer. The company was acquired by Roche in 2008, and the PIramed inhibitor, GDC-0941, is currently in early clinical trials. Mike Waterfield retired in 2008. After postdoctoral training with renowned cancer researcher Bob Weinberg at the Whitehead Institute in Boston, Julian Downward was recruited back to the ICRF and started his own laboratory there in 1989 (Highlight 14). He became LRI Associate Director in 2005, and is now an Associate Research Director at the Francis Crick Institute. He was elected to EMBO in 1995, became a fellow of the Royal Society in 2005, and a fellow of the Academy of Medical Sciences in 2009. Key references Waterfield MD, Scrace GT, Whittle N, Stroobant P, Johnsson A, Wasteson Å, Westermark B, Heldin CH, Huang JS, Deuel TF (1983). Platelet-derived growth factor is structurally related to the putative transforming protein p28sis of simian sarcoma virus. Nature. 304:35-9. Downward J, Yarden Y, Mayes E, Scrace G, Totty N, Stockwell P, Ullrich A, Schlessinger J, Waterfield MD (1984). Close similarity of epidermal growth factor receptor and v-erb-B oncogene protein sequences. Nature. 307:521-7 LRI Laboratories Highlights 23 LRI Laboratories Highlight 8 Conservation of the cell cycle 1987: ICRF Cell Cycle Control laboratory, led by Paul Nurse, shows for the first time that the cell cycle works the same way in all eukaryotic cells, a discovery with relevance for many diseases, but especially cancer. 2001: Paul Nurse, fellow ICRF lab head Tim Hunt, and Leland H. Hartwell are awarded the Nobel Prize in Physiology or Medicine “for their discoveries of key regulators of the cell cycle”. Background The process of reproduction is a central property of life, and this is seen in its simplest form in single cells. As a graduate student, Paul Nurse reasoned that studying the cell cycle, the mechanism driving the reproduction of cells, was important; in particular, the control of the cell cycle would be crucial. He realised that the best way to study such a complex process was by using genetics in a very simple organism. Over the next few years, Paul, first as a post-doctoral worker and then as a lab head in his own right, made his name by identifying many of the key regulators of the cell cycle in Schizosaccaramyces pombe (fission yeast). His work was dogged by the scepticism of many outside the yeast genetics community, who thought that his findings were irrelevant to more complex organisms such as mammals, but Tim Hunt and Paul Nurse, Nobel Prize Ceremony, 2001 24 Scientific report 2014 LONDON RESEARCH INSTITUTE fortunately, Walter Bodmer, the then director of the ICRF, was sufficiently farsighted that in 1984, he recruited Paul to the Lincoln’s Inn Fields laboratories. It was there that Paul’s lab confounded his detractors by proving that the fundamental components of the cell cycle were conserved between yeast and man. in 1987, a postdoctoral fellow in his lab, Melanie Lee, showed that one of the most important cell cycle regulators in yeast, a gene called cdc2, had a human counterpart, so establishing that the cell cycle in humans was likely to be regulated in the same way as in yeasts. The research Lee used a mutant version of fission yeast in which the yeast’s own cdc2 gene could be inactivated by raising the temperature at which the yeast was growing. Loss of cdc2 activity is lethal, so growing the mutant yeast at the higher temperature resulted in its death. This strain could now be used to hunt for genes able to substitute for cdc2 and rescue the yeast from dying. Lee introduced into the mutant strain a library in which all human genes were represented; in such a screen, each gene from the library is able to enter a small number of cells in the total population. If the gene is able to rescue (‘complement’) the lethal mutation, the cell in which it resides is saved, and keeps growing, making it easily recognisable in a field of its dead and dying compatriots. Amazingly, one human gene was able to save its host yeast cell, and when its DNA was rescued and analysed, it turned out to be very close in sequence to yeast cdc2; it was in fact the human version of the gene, conserved throughout millions of years of divergent evolution between yeast and us. The consequences This pioneering work led to an exponential leap forward in mammalian cell cycle research, as many of the other genes already known to be important in the yeast cell cycle also turned out to be conserved. The mammalian cell cycle community was therefore presented with a basic blueprint showing them how cells reproduced, and saving them years of arduous research. Lee and Nurse’s publication laid the foundations for much of our understanding of how the cell cycle is regulated in both normal cells, and in diseases such as cancer where the cell cycle is no longer controlled properly. Tim Hunt’s 1982 autoradiograph: the first sighting of cyclin What happened next? Paul Nurse became Director General of ICRF in 1996, and in 2002 oversaw its merger with the Cancer Research Campaign to create Cancer Research UK, the largest cancer research organisation in the world outside of the USA. He led Cancer Research UK as its first Chief Executive, before moving to Rockefeller University as its President in 2003. In 2010, he became President of the Royal Society, and in 2011 moved back to London as the first Chief Executive of the Francis Crick Insitute. Knighted in 1999 for services to cancer research, in 2010 he was voted Britain’s most influential scientist. Melanie Lee left the Nurse lab in 1988 for a highly successful career in the pharmaceutical industry. She is currently Chief Executive Officer of NightstarRx Ltd, a company established to pursue gene therapy treatment of the degenerative eye disease Choroideremia. She was formerly CEO of Syntaxin, and President of New Medicines and Executive Vice President R&D with UCB, having served as R&D Director at Celltech before its merger with UCB. Melanie Lee currently chairs Cancer Research Technology, the early drug R&D and technology licensing company of CRUK. Key references Lee M G, Nurse P (1987). Complementation used to clone a human homologue of the fission yeast cell cycle control gene cdc2. Nature. 327:31-5 Nurse P (2010). Sir Paul Nurse – Autobiography. http://nobelprize.org/nobel_prizes/medicine/ laureates/2001/nurse.html LRI Laboratories Highlights 25 LRI Laboratories Highlight 9 Why can’t a woman be more like a man? The race for the male sex-determining gene 1990: Peter Goodfellow’s lab show that the testis-determining factor, which specifies maleness, is encoded by the SRY gene. Background Humans normally have 46 chromosomes, comprising 22 pairs of autosomes and 1 pair of sex chromosomes, XX for females, and XY for males. Amazingly, the Y chromosome’s function as a determinant of maleness has only been known since 1959, when it was shown that the presence of a Y chromosome overrides any number of X chromosomes, and is necessary and sufficient for testis development and maleness. But what was it on the Y chromosome that specified maleness? By painstaking analysis of patients with jumbled sex chromosomes lacking pieces of Y, or where the X and Y had managed to mix together, scientists were able to narrow the region containing the so-called Testis Determining Factor (TDF) to a chunk of the Y-specific part of the Y chromosome, near the boundary with the pseudoautosomal region (the part of the Y chromosome which is able to pair with the X). But this still meant that many hundreds of thousands of base pairs of DNA had to be searched for the TDF gene. Peter Goodfellow, fanatical Arsenal fan, sometime poet, and all round eccentric, was recruited by Walter Bodmer in 1979, in his drive to bring human genetics to the ICRF. Peter’s expertise lay in gene mapping, pinpointing genes in the vast human Peter Goodfellow 26 Scientific report 2014 LONDON RESEARCH INSTITUTE genome, at that time a complicated and esoteric procedure. He had started his career mapping components of the immune system as a PhD student with Walter Bodmer in Oxford, and, after a postdoc at Stanford (during which he attended the last Sex Pistols concert and had the honour of being spat on by Sid Vicious), he started a lab at the ICRF to do the same thing for genes encoding tumourspecific antigens. By coincidence, one of these mapped very near the putative TDF locus. The unsolved problem of male sex determination sparked Peter’s interest, and, equipped as he was with the expertise to find genetic needles in genomic haystacks, he decided to identify and clone the elusive TDF gene. The research Things progressed very well to begin with. The Goodfellow lab’s advanced methodologies allowed them to produce a very good genetic roadmap of the TDF region, and in 1987, they published a paper in Nature which narrowed the region to be searched to a manageable 50 - 150 kilobase (50,000 - 150,000 basepairs) piece of DNA abutting the pseudoautosomal boundary. Then, what looked like disaster struck; the nightmare of every scientist working on a hot problem is to be scooped by the competition, and, in a Cell paper in 1987, David Page, at the Massachusetts Institute of Technology, announced the cloning and sequencing of the gene corresponding to Peter’s candidate locus. This gene, which Page called ZFY, was claimed, with much trumpeting, to be the long-awaited TDF. After some time spent sitting under his desk writing poetry and drinking too much coffee, Peter emerged to continue working, when he realised that the Page paper had not truly nailed the problem. To be the TDF, ZFY had to fulfil a number of criteria. The first, that it should be on the Y-specific part of the Y chromosome, was clearly fine, and that it appeared to be the only gene in a rather empty area of DNA was also in its favour. However, one significant problem was that the X chromosome carried a highly homologous gene, called ZFX. This was odd, as the two genes were so closely related that it would be hard for them to have different functions, as they must if ZFY, but not ZFX, determined maleness. Secondly, both Page and Goodfellow contacted an Australian expert on marsupial genetics, Jenny Graves, and asked her to probe the marsupial genome to check that marsupial Zfy was also on the Y chromosome. To everyone’s surprise, it was not; it lay on an autosome, which was very unexpected, as maleness in marsupials is also specified by the presence of a Y chromosome. This was a significant nail in the coffin, but ZFY’s short life as the TDF truly ended after the Christmas 1989 edition of Nature, which carried two papers, one from Peter’s lab together with the Fellous lab in Paris, and one from Peter’s collaborator Robin Lovell-Badge; the first described a number of sex-reversed XX men whose genomes did not contain ZFY, and the second showed that murine Zfy was not found in the cells specifying maleness. more ancient specifier of maleness than SRY; it exists in multiple non-mammalian species, in contrast to SRY, which is solely mammalian. The race was back on again, and this time, the Goodfellow lab won. Andrew Sinclair, the PhD student in Jenny Graves’s lab who had shown that marsupial Zfy was autosomal, moved to London to do a postdoc with Peter, and in 1990, was first author on a Nature paper describing the positional cloning and correct identification of SRY as the gene for determining maleness. The SRY gene was 150kb away from ZFY, and had been missed before because it was tiny, a mere 1000 bases long. What happened next? In 1992, the flamboyance quotient of the ICRF was reduced to a depressingly normal level when Peter Goodfellow moved to the University of Cambridge to become the Balfour Professor of Genetics. He still carries the distinction of being the only head of the Cambridge University Science Department to have a ponytail. He left academia to become Senior Vice President at Smith Kline Beecham in 1996, and became Senior VP, Discovery Research upon Smith Kline’s merger with Glaxo in 2001. He remained at Glaxo Smith Kline until his retirement in 2006, and now acts as a biotech consultant. His work on sex determination won him the 1995 Louis-Jeantet Prize, which he shared with Robin Lovell-Badge and three others, and, with Lovell-Badge and David Page, the 1997 Francis Amory Prize of the American Academy of Arts and Sciences. He was elected to the Royal Society in 1992, and in 1998, became a founding member of the Academy of Medical Sciences. SRY was subjected to the same scrutiny as ZFY, but passed with flying colours. The Goodfellow lab published another paper using the Fellous lab’s clinical material, showing that the three XX men described in the 1989 paper had picked up the part of the Y chromosome containing SRY; and as final proof that SRY was the real deal, Peter published yet another paper in Nature in 1991 with Robin Lovell-Badge, showing that female mice engineered to carry the SRY gene developed as males. This latter paper, with its memorable front cover image of Randy the sex-reversed mouse, caused a media storm at the time, and with its appearance, signalled an end to one of the highest profile scientific races ever. The consequences SRY proved to be a difficult protein to study. After many years of effort, we now know that it is a rather weedy transcription factor, whose sole purpose as a testis determining factor is to switch on a second gene, SOX9 (also cloned in Peter’s laboratory), which then does all the rest of the work in establishing maleness. Interestingly, SOX9 is an autosomal gene, and appears to be a much In a recent twist to the story, the long-held dogma that femaleness was a default state, and to be male one simply had to activate SRY and SOX9, was overturned in 2009 by Robin Lovell-Badge in a collaborative study which showed that in mice the autosomal gene FoxL2 specifies femaleness, and when present, overrides Sox9 and prevents ovaries changing into testes. Loss of FoxL2 in adult female mice upregulates Sox9, causing reprogramming of some ovarian cell types to those found in testes. Remarkably, as in life, the Sox9/FoxL2 story shows that maleness and femaleness appear to be established by what some might view as a balancing act, and others as a war. Key references Sinclair AH, Berta P, Palmer MS, Hawkins JR, Griffiths BL, Smith MJ, Foster JW, Frischauf AM, Lovell-Badge R, Goodfellow PN (1990). A gene from the human sex-determining region encodes a protein with homology to a conserved DNAbinding motif. Nature. 346:240-4 Berta P, Hawkins JR, Sinclair AH, Taylor A, Griffiths BL, Goodfellow PN, Fellous M (1990). Genetic evidence equating SRY and the testisdetermining factor. Nature. 348:448-50 Koopman P, Gubbay J, Vivian N, Goodfellow P, Lovell-Badge R (1991). Male development of chromosomally female mice transgenic for Sry. Nature. 351:117-21 LRI Laboratories Highlights 27 LRI Laboratories Highlight 10 Myc causes apoptosis 1992: Gerard Evan and co-workers show that the Myc oncogene is a double-edged sword, not only instructing cells to proliferate, but also causing programmed cell death (apoptosis). The balance between proliferation and apoptosis is now a universally recognised mechanism of growth control; the default pathway for all cells is death, and cancers are as rare as they are because it is hard to escape this default when things go wrong. Background As a postdoc in San Francisco in the lab of Mike Bishop (co-winner of the 1989 Nobel Prize in Physiology or Medicine), Gerard Evan developed an interest in the molecular mechanisms of cell growth and cell death, focusing particularly on the biology of the enigmatic Myc protein, a powerful engine of cell proliferation that is aberrantly expressed in most human cancers. Gerard continued to work on Myc upon his return to the UK and, after a period in Cambridge, was recruited to the ICRF in 1988. It had been known for some time that when tissue culture cells were forced to express large quantities of Myc protein, they divided uncontrollably but, in addition to this, researchers had noticed that there was a lot of death in the cultures. This phenomenon, known colloquially as ‘sick of myc’, had been dismissed either as a tissue culture artefact, perhaps brought on by exhaustion of the serum in the growth medium in which the cells were bathed, or as a Gerard Evan 28 Scientific report 2014 LONDON RESEARCH INSTITUTE toxic side-effect of Myc driving cells into division without giving them time to get properly prepared. Gerard, always the iconoclast, decided to find out what exactly was going on. The research Gerard and his coworkers introduced the Myc protein into a cell line, Rat-1 fibroblasts, and verified that in conditions where the cells should normally arrest their growth, too much Myc caused the cells to continue to divide. As expected, there was also a huge amount of cell death. To look more carefully at the death, instead of simply counting the cells or looking at them down a microscope, Gerard used a different technique, time-lapse video microscopy, which allowed the cultures to be filmed. What he saw was entirely unexpected. The cells were dying, not in a disorderly manner, but in a highly stylised fashion: the cells lost contact with their neighbours in culture; their nuclei condensed; their outer membranes started to bulge and then break away in small droplets (blebbing), and finally their cytoplasms condensed and the cells blew up spectacularly. When DNA from the dying cells was analysed on size-fractionating gels, it ran as a ladder of distinct bands. This sequence of programmed cell death, christened apoptosis in 1972, had been described nearly 150 years previously, and was well known, but in a different context: as well as having a role in shaping immunity, it is the main mechanism by which embryos are refined; for example, all human foetuses have webbed feet and hands, but the webbing is removed in the womb through regulated apoptosis. Gerard’s discovery that Myc, thought to be solely a growth-promoting oncogene, was also able to cause death as part of its normal function, was a great surprise. The consequences The Myc paper, together with work on the prosurvival oncogene bcl2, was in the vanguard of an explosion of research on apoptosis, which previously had been considered a minority interest even amongst immunologists and developmental biologists. An average of about 40 papers a year on apoptosis were published in the 20 years between the coining of the phrase in 1972 and 1992, when the Evan paper was published; the average is now close to 10,000 a year. Apoptosis has been shown to be universal, and to proceed by highly regulated control mechanisms, which are now well understood. The notion that cells have inbuilt suicide genes, and that when they cannot detect the appropriate signals for growth, the default pathway is apoptosis, now underpins modern cell biology. The unmasking of Myc as a bringer of death as well as life led to a complete reevaluation of theories as to how cancers develop. Prior to 1992, the prevailing view was that cancer was a disease of uncontrolled growth but, as growth promoting mutations are frequent, there was no good explanation for why, if this were the case, we do not develop many more cancers. The Evan paper on Myc showed that if cells are forced to divide uncontrollably by a mutated oncogene, the same signal, by default, pushes the cells to die: cancer is as rare as it is due to the intrinsic ability of our cells to suppress uncontrolled growth by committing suicide. If cancer is to develop, the default suicide pathway has to be switched off by another, cooperative, mutation. In more recent work, the Evan lab has shown that in cancer cells transformed by Myc, anti-apoptotic mutations have also occurred and, if these mutations are reversed, the cells die. Drugs attacking this weakness are now in Phase III clinical trials. What happened next? Gerard Evan was appointed Napier Professor of the Royal Society in 1996, whilst still at the ICRF. In 1999, upon his move to the University of California San Francisco Cancer Center, Gerard became Gerson and Barbara Bass Bakar Distinguished Professor of Cancer Biology. He returned to the UK in 2009 and is currently the Sir William Dunn Professor and Head of the Biochemistry Department at Cambridge University. He is an EMBO member, and a Fellow of both the Academy of Medical Sciences and the Royal Society. Gerard’s current research is focused on understanding the processes responsible for genesis and maintenance of cancers, in particular cancers of the pancreas, colon, brain, skin and liver. Understanding the molecular mechanisms that underlie the cell suicide machinery and how it can be manipulated therapeutically are still the overarching aims of his laboratory, and he continues to be one of the most productive and thought-provoking cancer biologists working today. Key references Evan GI, Wyllie AH, Gilbert CS, Littlewood TD, Land H, Brooks M, Waters CM, Penn LZ, Hancock DC (1992). Induction of Apoptosis in Fibroblasts by c-myc Protein. Cell. 69:119-28 LRI Laboratories Highlights 29 LRI Laboratories Highlight 11 DNA repair 1980s-2000s: The Lindahl lab identifies and dissects the base and nucleotide excision repair machinery. 1995: Rick Wood reconstitutes mammalian nucleotide excision repair in vitro. Background For as long as the DNA double helix has been around, environmental and cellular stresses have conspired to inflict damage upon it. Heat-induced breakages, ultraviolet light, ionizing radiation, chemicals, and the inbuilt frailty of the DNA molecule itself all contribute to the daily toll that life takes upon our genetic material. So it’s no surprise that even the simplest organisms use a spectacular battery of error-detection and repair mechanisms to keep their precious DNA as fault-free as possible. To begin with, all the work on DNA repair happened in single-cell organisms, where one could use biochemistry to purify components of the repair machinery, and genetics to look at function. However, this began to change when it was realised that many of the ancient enzymes discovered in bacteria were conserved in complex multicellular organisms such as ourselves. Furthermore, it also became clear that the DNA repair processes in humans were clinically important: firstly, mutations in human DNA repair enzymes started to crop up as the causative factors in some inherited diseases, and secondly, where the DNA repair machinery was working efficiently, it was opposing the action of the anti-cancer drugs and therapies which worked by damaging tumour cell DNA. Clearly, working on DNA repair was a fitting topic for a cancer research institute. Rick Wood 30 Tomas Lindahl Scientific report 2014 LONDON RESEARCH INSTITUTE In the 1970s, the ICRF took its first steps into the DNA repair field via the brilliant molecular biologist John Cairns, who was recruited from Cold Spring Harbor to be Director of the ICRF unit at Mill Hill. On his departure for Harvard and a MacArthur Genius Award in 1980, he was replaced by a major biochemical star who, in addition to his fundamental work on DNA repair, was to shape and guide the ICRF Clare Hall Laboratories as their first Director: Tomas Lindahl. The research As a postdoc with Jacques Fresco at Princeton in the mid-1960s, Tomas was the first person to realise that DNA was inherently unstable, and could be damaged without any requirement for exogenous mutagens. By their very nature, some of the chemical bonds formed inside the bases, and between the bases and the sugar-phosphate backbone, are relatively susceptible to breakage. Cytosine, for example, can be readily deaminated, that is, stripped of its amino group, which turns it into the simpler base, uracil. Uracil can pair comfortably with adenine, rather than with cytosine’s proper partner guanine, leading to errors in replication and transcription similar to those induced by chemically- and radiationinduced damage. Base Excision Repair The discovery of DNA’s instability defined Tomas’s career. In tracking down how the cytosine-to-uracil error was fixed, Tomas and his lab unmasked an entirely new class of repair mechanism: base excision repair. Uracil is detected and chopped out of the DNA chain by a glycosylase, an enzyme able to cleave the bond between the errant base and the backbone, leaving the double-stranded backbone intact. A gang of other enzymes then take over, chopping out the sad remains of the original nucleotide together with its nearest neighbours, mending the gap using the opposite strand as a template, and finally stitching up the break. Over the next decades, Tomas and his lab identified many of the weird and wonderful enzymes and mechanisms involved in multiple types of DNA repair, including the suicide inactivator O6methylguanine-DNA methyltransferase, which can restore mutated O6-methylguanine bases to health by removing the methyl groups and disposing of them by methylating itself – the enzymatic equivalent of falling on a live grenade (remarkably, Tomas found that O6-methylguanine-DNA methyltransferase is also a transcription factor, activating several repair-essential genes, including itself). And in 1994 and 1996, returning to Tomas’s first love, his lab achieved a biochemical triumph: the purification and reconstitution of base excision repair in vitro, using respectively the E.coli and human enzymes, some of which were remarkably conserved across the yawning evolutionary gulf. Xeroderma pigmentosum In 1985, Tomas hired a new American postdoc, Rick Wood, to work on xeroderma pigmentosum (XP). XP is a recessive autosomal inherited disease whose sufferers exhibit extreme sensitivity to ultraviolet light and have a strong predisposition to skin cancer. Its genetics are complex – there are at least ten different genes implicated – but the root of the XP problem had been known since the 1960s: XP patients were unable to repair UV-induced DNA damage properly. However, in the nearly 20 years since the defect had been defined, no one had managed to determine the mechanism involved. The problem was the lack of a cell-free system in which to do the fiddly biochemistry required to identify and purify the UV-repair machinery. Repair of UV damage in E.coli takes place via a process called nucleotide excision repair, one of the first repair mechanisms discovered. In a paper published in 1988, Rick, Peter Robins and Tomas showed that a similar process was occurring in humans. Using extracts made from human cells growing in tissue culture, they succeeded in developing a cell-free system capable of repairing UV-damaged DNA in a test tube. If extract made from the cells of XP patients was added to the assay, repair no longer happened; the XP defect appeared to affect the detection and nicking of damaged DNA before the repair process to remove dud bases and replace them with the correct ones had even kicked in. Rick was clearly too valuable to lose, and in 1988, he started his own laboratory at Clare Hall, to study the biochemistry of inherited syndromes, with a particular interest in XP. In 1995, Tomas’s faith in him paid off when the Wood lab, together with collaborators in France, Switzerland, Finland and the US, published a completely reconstituted mammalian nucleotide excision repair system. About 30 separate proteins had been identified, purified and characterised, a formidable achievement in the field of DNA repair. The consequences Correct DNA repair is the bedrock on which healthy cells thrive. Cells have an elaborate system of checks and balances in their transcriptional and cell-cycle machinery to detect DNA damage, assess its seriousness, and decide whether to fix it or to self-distruct. The medical consequences of defective repair are legion: for example, nucleotide excision and mismatch repair genes are essential for the prevention of cancer and neurological disease. With our increasing understanding of defects in DNA repair and checkpoint control in cancer, there is also extensive interest in targeting particular DNA repair and checkpoint pathways for cancer therapy. What happened next Tomas Lindahl remained as Director of the Clare Hall Laboratories until 2005, retiring in 2009. He is still one of the world’s foremost authorities on DNA damage. For his many landmark discoveries in DNA repair, he has been awarded many prizes and honours. He is an EMBO member, a Fellow of the Royal Society and the Academy of Medical Sciences, and has won both the Royal Society’s Royal Medal and Copley Medal. The latter is the oldest scientific award in the world, whose other recipients include Charles Darwin, Michael Faraday and Stephen Hawking. Rick Wood returned to the US in 2001, and is currently Grady F. Saunders Distinguished Professor in Molecular Biology at The University of Texas MD Anderson Cancer Center. He is a Fellow of the Royal Society and an EMBO member, and in 1998 won the Meyenburg Prize for Cancer Research, one of Germany’s most prestigious science awards. Key references Lindahl T (1996). The Croonian Lecture, 1996. Endogenous Damage to DNA. Phil Trans R Soc Lond B. 351:1529-38 Wood R, Robins P, Lindahl T (1988). Complementation of the Xeroderma Pigmentosum DNA Repair Defect in Cell-Free Extracts. Cell. 53:97-106 Aboussekhra A, Biggerstaff M, Shivji MKK, Vilpo JA, Moncollin V, Podust VN, Protic M, Hubscher U, Egly J-M, Wood, RD (1995). Mammalian DNA Nucleotide Excision Repair Reconstituted with Purified Protein Components. Cell. 80:859-68 LRI Laboratories Highlights 31 LRI Laboratories Highlight 12 Regulation of DNA replication 2015: John Diffley’s group reconstitutes regulated DNA replication origin firing using purified proteins. This work marks the culmination of over twenty five years’ study of the mechanism of replication initiation and its control by the cell cycle and DNA damage response machinery. Background Every time a cell divides, it has to make a complete and accurate copy of its genome. If it does not, the result is disastrous, resulting in at best, a crippled existence likely to end in early death, and at worst, mutation into a cellular psychopath with the potential to cause cancer and a multitude of other diseases. DNA replication, the process whereby the genome is copied, is therefore one of the most important and basic processes a cell undertakes. The great biochemist Arthur Kornberg began the study of DNA replication in the mid-1950s, by looking at the enzymes required for replication in basic organisms such as bacteria. Bacteria, with their small genomes, start replicating from one particular point in their DNA, known as the origin of replication, and just continue until they have gone full circle. This is a relatively simple process. Eukaryotes, however, have a far larger complement of DNA, which is broken up into separate chromosomes, and therefore they need multiple origins of replication in order to carry out the process on each chromosome, and within a reasonable timescale; in the case of mammalian genomes, origins of replication number in the tens of thousands. The problem of how all these origins are able to fire just once in every cell cycle to John Diffley 32 Scientific report 2014 LONDON RESEARCH INSTITUTE orchestrate the complete replication of the genome has preoccupied researchers for the last 40 years. In the late 70s and early 80s, Bruce Stillman’s lab at Cold Spring Harbor, Long Island, NY, was one of the dominant forces in the study of eukaryotic DNA replication, using a biochemical approach to laboriously purify and then identify the proteins involved in viral DNA replication. John Diffley came to the Stillman lab at Cold Spring Harbor as a postdoc in 1985, where he initiated work on DNA replication in yeast, the simplest eukaryotic organism there is. Yeast is easy to grow and manipulate, and can be used not only for biochemistry but also for genetic studies, giving scientists more versatility in how they approach problems. Crucially, what holds for yeast is frequently broadly true for higher eukaryotes, and therefore it is a great model organism. During his time with Stillman, John found the first ever protein known to bind to eukaryotic replication origins, and on the back of his outstanding postdoctoral work, he was recruited to the Clare Hall Laboratories of the ICRF in 1990. The research John Diffley studies how replication begins, the mechanisms that ensure replication is triggered only once during each cell cycle, and what happens when replication is not initiated properly. His work on all of these issues has been of seminal importance. ORC: After his arrival at Clare Hall, John began work to identify the proteins bound at yeast replication origins in a region of DNA known to be essential for the origins to fire properly. In 1992, his lab and Stillman’s defined these proteins as the Origin Replication Complex (ORC), and the following year, in collaboration with Kim Nasmyth’s lab, the Diffley group, along with several others cloned Orc2, the first gene of the six-member complex, and showed by genetics that Orc2 was essential for replication. PreRC and Licensing Factor: The identification of ORC created a great deal of interest, but it was unlikely to be what determined the timing of replication during the cell cycle, as it was bound to DNA almost all of the time. The lab decided to look for other replication origin proteins which bound only at particular points during the cell cycle, reasoning that they might play a role in replication timing. They found that one particular DNA sequence in replication origins was only occupied by proteins during a phase of the cell cycle called G1, which occurs immediately before S phase, the point where the genomic DNA is replicated. In 1994, they published evidence that this protein complex, dubbed the Pre-Replicative Complex (preRC), was very similar to Xenopus Licensing Factor, an activity in frog eggs required for initiation of replication. That similar complexes were found in two such dissimilar organisms suggested, much to everyone’s excitement and relief, that the control of replication regulation might be broadly the same in all eukaryotes, and this has indeed turned out to be the case. In 1996, the Diffley and Nasmyth labs again collaborated to identify the first preRC complex protein, Cdc6, and the Diffley lab subsequently demonstrated that Cdc6 was required to recruit other components, the MCM helicase proteins, into the complex. Last year, 18 years on, they reconstituted the entire preRC step in a test tube using purified proteins, a scientific and technological tour de force. DNA replication is inextricably linked to the cell cycle: Having found that assembly of the replication complex was cell cycle-regulated, the next step was to find out how. In this again, the Diffley and Nasmyth labs were, in 1996, the first to show that the reason the preRC is only present on DNA during G1 phase is that a class of enzymes called cyclin dependent kinases (CDKs) modifies and inhibits the preRC; G1 is the only time during the cell cycle that CDKs are absent. Put together with work from other researchers, including Diffley’s Nobel Prize winning colleague at Clare Hall, Tim Hunt, these results provided an elegantly simple solution to the puzzle of how replication only happens once during the cell cycle. First, the preRC binds to replication origins in G1 phase, readying the DNA for replication. Then, in S phase, CDKs are made, and the MCM helicase is activated, allowing the DNA to be unwound and then copied. The CDKs switch on the initiation of DNA synthesis, the genome is replicated, and the cells go through S phase and into mitosis, or cell division. When G1 of the next cell cycle comes round again, CDKs are degraded, and the whole process repeats. In a further flourish, the Diffley lab has shown exactly how the CDKs inhibit the preRC, and, in 2007, that they trigger S phase DNA replication by modifying just two proteins, Sld2 and Sld3. How DNA damage halts replication: The Diffley lab has also worked extensively on how replication in S phase is halted in response to DNA damage. The so-called DNA damage checkpoint shuts down the firing of late replication and this works via modification of two key initiator proteins, Sld3 and Dbf4. Again, CDKs turned out to be intimately involved. There are also other mechanisms at work too; the checkpoint can stabilise stalled replication forks by inhibiting a crucial enzyme, the flap endonuclease Exo1. The ubiquity of CDKs in controlling when and whether DNA replication occurs has major consequences in many cancers, as CDKs are frequently deregulated, switched on at the wrong times, and/or overactive. Misfiring of the replication origins leads to genome instability, and if the normal damage limitation procedures are not in place, the genesis of a tumour. Diffley’s work is of extreme importance both in terms of basic science, and also for the development of new anticancer therapeutics targeting DNA damage checkpoints. What happened next? John Diffley became Deputy Director of the London Research Institute and Director of the Clare Hall laboratories in 2005. He is currently an Associate Research Director of the Francis Crick Institute. He was elected to EMBO in 1998, FRS in 2005, and elected a Fellow of the American Association for the Advancement of Science in 2007. He won the Paul Marks Prize for Cancer Research in 2003. Key references Diffley JFX, Cocker JH, Dowell SJ, Rowley A (1994). Two steps in the Assembly of Complexes at Yeast Replication Origins In Vivo. Cell. 78:303-16 Santocanale C, Diffley JF (1998). A Mec1- and Rad53-dependent checkpoint controls late-firing origins of DNA replication. Nature. 395:615-8 Tercero JA, Diffley JF (2001). Regulation of DNA replication fork progression through damaged DNA by the Mec1/Rad53 checkpoint. Nature. 412:553-7 Zegerman P, Diffley JF (2007). Phosphorylation of Sld2 and Sld3 by cyclin-dependent kinases promotes DNA replication in budding yeast. Nature. 445:281-5 Yeeles JT, Deegan TD, Janska A, Early A, Diffley JF (2015). Regulated eukaryotic DNA replication origin firing with purified proteins. Nature. 519:431-5 LRI Laboratories Highlights 33 LRI Laboratories Highlight 13 Into the nucleus: regulation of transcription 1992: Identification of SAP-1, the first growth factor-regulated transcription factor. 1995: Demonstration that proteins involved in controlling cell shape and structure can also regulate transcription, leading to the later demonstration that transcription can be directly regulated in response to cytoskeletal dynamics. Background At the end of the 1970s, it was becoming evident that there was a relationship between growth factors, oncogenes, and the activation of gene expression: cells required gene transcription to be activated in order to respond to growth signals, so there must be a signalling pathway leading from outside the cell, where growth factors docked, right across the cytoplasm and into the nucleus. What this was, and how it caused gene transcription, was the next big challenge in the transcription field. All genes are regulated by specific DNA control sequences often found in front (upstream) of the gene. It was clear that the secret of how signals were transmitted to genes lay in these regulatory sequences, but it was a mystery how this worked. Whilst at the MRC Laboratory of Molecular Biology in Cambridge, Richard Treisman begun working on how the Fos gene, cloned by Tom Curran at the ICRF (Highlight 5), was regulated. Fos is an immediate early gene, switched on almost instantly when a growth signal is received by a cell, and Richard showed very quickly that this response was controlled by a short DNA sequence, the Serum Response Element (SRE) in the Fos Richard Treisman 34 Scientific report 2014 LONDON RESEARCH INSTITUTE upstream regulatory region, to which was bound a protein, the Serum Response Factor (SRF). After his initial work, he and others found SREs in the control regions of many other genes, demonstrating this was a widespread mechanism of rapid gene induction. However, there were still huge holes in the pathway; although the SRE and SRF were necessary for Fos gene induction, the method by which the signal to fire up transcription reached them was still unknown. In 1988, lured by the presence of a substantial community working on the control of eukaryotic transcription, Richard returned to the ICRF, where he’d been a PhD student, bringing this puzzle with him. The research Work on SRF continued in the Treisman lab, but soon hit a block; SRF did not seem interested in responding to any growth factor signals, and did not work very well in assays where it was asked to activate growth-factor responsive transcription by itself. However, others in the transcription field had descended in force on the SRE, and Peter Shaw and Alfred Nordheim in Germany showed in 1989 that SRF bound the SRE with a partner protein, which they called TCF (ternary complex factor). Steve Dalton, an Australian postdoc, used a genetic screen to identify a protein called SAP-1 as a TCF; and Nordheim’s group produced Elk-1, another member of the same protein family as SAP1, as a second TCF candidate. But was the SAP-1 and Elk-1 family the link to growth factor control of the SRE? While this work was proceeding, the study of a set of enzymes in the cell known as the MAP kinases was rapidly merging with studies of proto-oncogenes, leading to the discovery of a pathway whereby growth factor receptors activated the Ras oncogene, which then signalled to a kinase called Raf, which in turn activated another kinase called ERK (Highlight 14). Kinases act by putting phosphate groups onto other proteins at particular amino acids, and excitingly, both SAP-1 and Elk-1 contained the correct amino acid sequences that might allow them to be direct targets of ERK. Richard Marais, another Treisman postdoc, spent much of the summer of 1992 in the lab, and was rewarded for his lost holidays by data showing definitively that ERK was able to phosphorylate Elk-1, and that this was the activating signal switching on SRE-regulated transcription. For the first time, a complete pathway leading from outside the cell into the heart of the nucleus had been delineated. Although it was clear that growth factors switched on the SRE through the SRF-TCF interaction, something about transcriptional regulation by the SRE was still very puzzling. The Serum Response Element, as its name suggests, is activated by serum, a rich liquid mix of nutritional goodies used for growing cells in tissue culture. However, there was something in serum capable of activating the SRE that was not a protein growth factor. Caroline Hill, another postdoc, worked out that this extra something was lysophosphatidic acid, or LPA, and that for LPA induction, TCF proteins were not required. Furthermore, the signal coming to SRF was not transmitted by the MAPK pathway, but by a totally different route, by Rho family proteins, whose normal function is in remodelling the actin cytoskeleton. The actin cytoskeleton is the main structural framework of a cell, giving it both shape and strength, and it must be continually remodelled in response to the cell’s need to move, to change shape, and to adhere to the extracellular matrix and to other cells. The observation that the enzymes involved in cytoskeletal control are also activating transcription has supplied an entirely new perspective on a cell’s ability to fine tune all its processes in response to a stimulus. The consequences This work was extremely important to the nascent field of transcriptional regulation as it laid down some basic principles not only in terms of the mechanics of regulation itself but also in its use of innovative experimental techniques: Richard and his coworkers adapted and invented methodology which became universal tools in transcription research. That the lab won some very hard-fought contests in a highly competitive field is a tribute to Richard’s unerring instinct for discerning and pursuing what was interesting, and discarding what was not, and his recruitment of a cohort of very good scientists who were as unafraid of hard work as he was himself. The Treisman lab has continued its work on SRF and its protein partners, and is now studying not only basic transcriptional mechanisms but also the biological readout of the different interactions and signalling pathways that intersect at SRF. Their work on how the actin cytoskeleton controls SRF is particularly interesting. The targets of Rho family signalling have been identified as Myocardin-related Transcription Factors (MRTFs) called MAL and Mkl2. MRTFs bind SRF, and the SRF-MRTF complex performs an analogous role to SRF-TCF as the terminus for actin cytoskeleton-regulated signalling. Rho signalling to SRF-MRTF works by an entirely novel mechanism; there are fluxes in the amount of MRTF protein free to regulate transcription, depending on how much is bound by actin, and this itself is regulated very tightly by what the cell is doing at any given time. Rho signalling is strongly implicated in cancer cell invasion and metastatic tumour spread, and the lab has now shown that these processes require transcription regulation via the Rho-actin-MRTF-SRF signalling pathway. What happened next? Richard Treisman became Director of the LRI in 2000, and is now a Research Director at the Francis Crick Institute. He is an EMBO member, and a fellow of the Royal Society and the Academy of Medical Sciences. For his work on growth factor regulation of transcription, he was awarded the 1995 EMBO Gold Medal, and the 2002 Louis-Jeantet Prize. Steve Dalton is Chair of Molecular Biology at the University of Georgia. His lab works on the molecular biology of stem cells. Caroline Hill has been a group leader at the LRI since 1998, working on the Transforming Growth Factor family of signalling molecules. She was elected an EMBO member in 2002. Richard Marais is now Director of Cancer Research UK’s Manchester Research Institute. He is a member of EMBO and the Academy of Medical Sciences. Key references Dalton S, Treisman R (1992). Characterization of SAP-1, a protein recruited by serum response factor to the c-fos serum response element. Cell. 68:597612 Marais R, Wynne J, Treisman R (1993). The SRF accessory protein Elk-1 contains a growth factorregulated transcriptional activation domain. Cell. 73:381-93 Hill CS, Wynne J, Treisman R (1995). The Rho family GTPases RhoA, Rac1, and CDC42Hs regulate transcriptional activation by SRF. Cell. 81:1159-70 Miralles F, Posern G, Zaromytidou A-I, Treisman R (2003). Actin dynamics control SRF activity by regulation of its coactivator MAL. Cell. 113:329-42 LRI Laboratories Highlights 35 LRI Laboratories Highlight 14 Upstream and downstream of the RAS oncoprotein 1990s: Julian Downward shows that signals from outside the cell can activate the Ras oncoprotein, and goes on to demonstrate that growth factor regulation of Ras controls the Raf-MAPK pathway. Background In 1981, Bob Weinberg’s lab at the Whitehead Institute in Boston demonstrated for the first time that there were human genes that could cause cancer when mutated. Using the new technique of DNA transfection, whereby foreign DNA could be introduced directly into cells, they showed that a fragment of DNA from a human tumour was able to transform mouse NIH3T3 tissue culture cells. Other groups began to see the same results using DNA from other tumour types, and together, they realised that the fragments of human DNA they were isolating all contained the H-ras gene, the cellular homologue of v-H-ras, one of two closely related retroviral oncogenes from Rat Sarcoma viruses. By the late 80s, Ras family proteins had been shown to be attached to the inside of the cell membrane, and to belong to a larger protein family, the GTP binding proteins. Such proteins are able to bind guanine triphosphate (GTP), a product of cellular metabolism, and convert it to guanine diphosphate (GDP). It was known that the mutations that converted normal Ras proteins into oncogenes caused them to stick in the GTP-bound activated state, so they were permanently switched on. However, Ras proteins existed in a knowledge vacuum: nobody knew what proteins normally lay upstream of them to cause the switching from GTP to GDP binding, or what lay downstream. Julian Downward, who as a PhD student at ICRF had shown that the Epidermal Growth Factor Julian Downward 36 Scientific report 2014 LONDON RESEARCH INSTITUTE Receptor gene EGFR had been picked up by a retrovirus and converted into the erb-b oncogene (Highlight 6), had gone off in 1986 to do a postdoc in Bob Weinberg’s lab. He ended up spending quite a lot of his time there unsuccessfully trying to work out what the signal might be, which triggered the activation of normal Ras proteins. In 1989, he returned to the ICRF as a lab head, bringing with him the problem he had been working on in Boston. The research Ras can be activated by extracellular stimuli: At the ICRF, Julian met Doreen Cantrell, a leading T cell immunologist. As an experimental tool for studying signal transduction, T cells are extremely attractive, as they can be easily purified, and fooled in vitro into activating themselves if they are fed mock antigens. Doreen suggested to Julian that her T cell system would be an ideal place to look at what was activating Ras. The shift in perspective, as can often happen in scientific research, broke the logjam. In four incredibly productive months, the two labs showed for the first time that the Ras protein could be activated very rapidly in response to extracellular signals stimulating the T cell receptor, the cell surface molecule on T cells to which antigen binds. Upstream of Ras: With this fundamental observation, that the mechanism of normal Ras activation was via a signal from outside the cell, the race was on to try to fit together the gaps in the signalling pathway between the receptor on the cell surface and Ras. After the first observation in T cells, many other receptors were shown to respond to extracellular stimuli by switching on Ras, including Julian’s old friend EGFR. Julian and his postdoc Laszlo Buday realised that work done genetically dissecting Ras signalling in flies could be put together with biochemical clues from mammalian cells to produce two likely candidates for the missing links, the Sos and Grb2 proteins. Sos was known to act downstream of the fly EGFR gene and upstream of fly Ras, and there was also good reason to think that it was a guanine exchange factor, able to catalyse rapid exchange of GDP for GTP, and therefore a prime candidate for activating Ras. Grb2 had been proposed to be an adaptor protein, acting as a link between the EGFR and intracellular signalling components, and there was indirect evidence implicating it in Ras activation. Julian and Laszlo used antibodies to immunoprecipitate Ras protein along with Grb2 and Sos, and found that the interactions they detected resulted in Ras becoming activated. Their efforts successfully showed that the EGFR, when activated by EGF binding, recruited Grb2 to its intracellular tail, that Grb2 in turn bound Sos, that Sos had hold of Ras, and that all together, this enabled Ras to be activated. The missing upstream links had been defined. But what lay downstream of Ras, between it and the genes controlling the cell cycle? Part of the answer lay with another proto-oncogene, cRaf. cRaf protein is a kinase (an enzyme catalysing the addition of phosphate groups to proteins), and there was indirect evidence linking it with activation of Ras: disruption of cRaf activity had been shown to block Ras action. The Downward lab used a piece of cRaf as a biochemical bait to see whether it was able to pull down Ras protein, and showed that indeed, Ras and cRaf interacted, but only when Ras was in its activated, GTP-bound state. This was a crucial interaction to map, as downstream of cRaf lay the mitogen-activated protein kinase (MAPK) cascade. Kinases belonging to the MAPK family are used throughout evolution to control the cellular responses to external signals such as growth factors, nutrient status, stress or inductive signals, and, as shown in the previous year in Richard Treisman’s lab at the ICRF, MAPK could regulate transcription factors (Highlight 13). The Ras signalling pathway had reached the nucleus. There was one further twist to the Ras story, as it became clear to Julian that not all of the things Ras did could be explained by it simply activating the MAPK pathway. Another protein, phosphatidylinositol-3-kinase (PI3K), had been shown in 1992 to interact with Ras in vitro, and PI3K was known to be activated in response to the same stimuli as Ras. Julian’s lab, in collaboration with his old mentor, Mike Waterfield, demonstrated in 1994 that activated, GTP-bound Ras bound directly to PI3K, and that this interaction was likely to result in PI3K being activated in turn. The Downward lab’s work had placed Ras at the apex of two downstream signalling cascades, both of which are vitally important for cell growth. The consequences The mechanisms of Ras signalling have been fleshed out in the intervening years such that we now have a very detailed map of signal transduction in both normal and cancerous cells. Pathways turn out to be interconnected, and the molecular wiring diagrams for different cell types are varied and complex. Scientists are turning towards systems biology to model how perturbations in one part of a cell’s wiring changes outcome for the cell, and signal transduction research is becoming rigorously quantitative. Mutations in Ras genes account for some 20% of all human cancers. More work from the Downward lab has shown that mice with mutations in PI3K that block its ability to interact with RAS are highly resistant to lung tumours induced by KRAS, and skin tumours induced by HRAS. Interaction of RAS with PI3K is needed for normal growth factor signalling and also for RAS-driven tumour formation. What happened next? Julian Downward became LRI Associate Director in 2005, and is now an Associate Research Director at the Francis Crick Institute. He was elected to EMBO in 1995, became a fellow of the Royal Society in 2005, and a fellow of the Academy of Medical Sciences in 2009. He is one of the top 20 most cited European cell biologists. Doreen Cantrell CBE moved from the LRI in 2002 to become Professor of Cell Biology at the University of Dundee, where she is now Vice Principal and Head of the College of Life Sciences. She is an EMBO member and a Fellow of the Academy of Medical Sciences, the Royal Society of Edinburgh and the Royal Society. Laszlo Buday returned to his native Hungary, and is now Director of the Institute of Enzymology of the Hungarian Academy of Sciences in Budapest. Pablo Rodriguez-Viciana runs a lab at the UCL Cancer Institute, and is still working productively on the RAS family of proteins. Key references Downward J, Graves JD, Warne PH, Rayter S, Cantrell D (1990). Stimulation of p21ras upon T-cell activation. Nature. 346:719-23 Buday L, Downward J (1993). Epidermal Growth Factor Regulates p21ras through the Formation of a Complex of Receptor, Grb2 Adapter Protein, and Sos Nucleotide Exchange Factor. Cell. 73:611-20 Warne PH, Rodriguez-Viciana P, Downward J (1993). Direct Interaction of Ras and the aminoterminal region of Raf-1 in vitro. Nature. 364:352-5 Rodriguez-Viciana P, Warne PH, Dhand R, Vanhaesebroeck B, Gout I, Fry MJ Waterfield MD, Downward J (1994). Phosphatidylinositol-3-OH kinase as a direct target of Ras. Nature. 370:527-32 LRI Laboratories Highlights 37 LRI Laboratories Highlight 15 Interferon signalling and the JAK-STAT pathway 1992: Interferons are shown to signal through a novel pathway involving the JAK and STAT protein families. 38 Background In 1957, Alick Isaacs and Jean Lindenmann, working at the National Institute for Medical Research (NIMR) in Mill Hill, described a new phenomenon, ‘virus interference’. Chicken cells infected with influenza virus produced and secreted a factor which made non-infected cells resistant to viral attack, not just from ‘flu viruses, but also from a myriad of other virus types. The mysterious antiviral factor was a small protein, which Isaacs and Lindenmann named ‘interferon’. During the 1960s and 1970s, excitement mounted when it became clear that interferons, by then shown to be a family of proteins, did not just inhibit viral infection, but also had antitumour activity. By 1981, purified interferons were entering clinical trials as treatments for both viral infection and cancer. the end, from the genes being regulated in the nucleus. George and Ian’s labs, working closely together, went for the latter strategy, searching for genes that were switched on when the interferon family members IFN-α, IFN-β and IFN-γ were added to cells. Like Richard Treisman (Highlight 13), they reasoned that these genes must be activated by the binding of sequence-specific, interferon-inducible transcription factors. If they could identify the DNA sequences to which the factors bound, they could bootstrap their way to the factors, and thence to the rest of the proteins in the pathway. By 1988, they, and Jim Darnell’s lab in the US, had identified short stretches of DNA to which interferonregulated transcription factors might bind. The hunt to identify these transcription factors, and the signalling cascade that led to them, was on. In 1980, to boost its activity in the expanding interferon field, the ICRF Director Walter Bodmer hired a new faculty member, Ian Kerr. Ian had made his name some years previously by discovering one of the major antiviral defence mechanisms activated by interferon, the 2-5A system, and it was a major coup for the ICRF to have bagged such a big star of the interferon field. Matters improved still further in 1983 when Ian’s long-time friend and collaborator, George Stark, also moved to the ICRF, to work more closely with Ian on the problem of exactly how interferon signalled into cells. To define a signalling pathway, one can start at the beginning, at the cell surface, or work back from The research In 1989, Ian and George’s PhD student Trevor Dale published a paper describing an interferoninducible DNA binding factor, dubbed ISGF3. Attempts to purify ISGF3 in London were preempted by Jim Darnell’s lab, which, after some heroic biochemistry, showed in 1992 that ISGF3 was a multiprotein complex, containing three proteins eventually named Interferon Responsive Factor 9 (IRF9), STAT1 and STAT2 (for Signal Transducer and Activator of Transcription). Darnell’s lab also found that in response to an interferon signal, the STAT proteins are phosphorylated on a particular amino acid, tyrosine. How all this fitted into a complete pathway was unclear, though, and might have remained so for some time but for an Italian George Stark Ian Kerr Scientific report 2014 LONDON RESEARCH INSTITUTE postdoc in George’s lab, Sandra Pellegrini, who used a genetic approach to solve the problem. Sandra had arrived in George’s lab in 1986, and was given the task of mutating cells so that they would no longer respond to INF-α stimulation. The idea was that once she had made a mutant cell line, she could introduce a library of normal genes into it, and find which one was able to rescue (‘complement’) the mutation. Sandra developed a cunning drug selection strategy whereby only those mutant cells that did not respond to interferon would survive. It took a while, and was incredibly labour-intensive, but in the end, all the patience and toil paid off: Sandra published a paper in 1989 containing details of the mutant cell line, before leaving London to set up her own lab at the Institut Pasteur in Paris. She subsequently cloned TYK2, a member of the Janus Kinase (JAK) family, which phosphorylate proteins on tyrosine residues when cells are stimulated with interferons. Following Sandra’s success, Diane Watling, working with Ian and George, used a cell surface marker – in place of Sandra’s drug–based selection – to establish that IFN-γ, of intense interest in the immune response, also signals through a JAKdependent pathway. In all, eight cell lines defective in interferon-inducible signalling were isolated. They proved incredibly powerful tools; between them, they covered the entire signalling pathway, including the STAT proteins that the Darnell lab had already identified, and could be used for verification of any genes thought to encode pathway components. The cell lines could also be used for functional studies on the pathway proteins, defining the domains of the proteins required to carry out particular tasks and to contact partners in the signalling cascade. By 1994, when Ian, George and Jim Darnell jointly published an influential review on the subject, the basics of the pathway were well understood: when interferons bound to their receptors, the JAK kinases, which were associated with the inward facing parts of the receptors, were activated; they then bound and phosphorylated STAT proteins, allowing them to move into the nucleus, find their DNA binding sites, and fire up the interferoninducible genes. In a few hectic years, an entire novel signalling pathway had been defined. The consequences Interferon’s early therapeutic promise has been partially fulfilled: synthetic interferons are used to treat multiple sclerosis, hepatitis B infection and several different types of cancer, particularly kidney cancer, malignant melanoma, multiple myeloma and some types of leukaemia. Interferons belong to a larger class of proteins called cytokines, which are vitally important in cell signalling, particularly in the immune system. JAK-STAT pathways are used by most cytokines and a large number of other extracellular signalling molecules, and JAK and STAT family members are used in a myriad of different combinations to elicit different effects in different biological contexts. What happened next? Ian Kerr remained at the LRI until his retirement in 2005. He is an EMBO member, and a Fellow of the Royal Society and the Academy of Medical Sciences. He shared the Milstein Prize of the International Society for Interferon and Cytokine Research in 1993 and won it again, with George Stark and Jim Darnell, in 1997. In 1999 the trio, together with two others, won the William B Coley Award for Distinguished Research in Basic and Tumour Immunology. In 2003, Ian received the Feldberg Foundation Prize for Outstanding Work in Medical or Biological Sciences. George Stark returned to the US in 1992 to become Chair of the Lerner Research Institute at the Cleveland Clinic Foundation. He remains there as a Distinguished Scientist, with a joint appointment as Emeritus Professor of Genetics at Case Western Reserve University. In addition to the Milstein Prize in 1997 and Coley Award in 1999 he has been elected to the US National Academy of Sciences, to the Royal Society, and to the US Institute of Medicine. By 2012, on the 20th birthday of the JAK-STAT pathway, the Kerr and Stark labs’ cell lines had been used by 500 laboratories worldwide. Key references Dale TC, Imam AM, Kerr IM, Stark GR (1989). Rapid activation by interferon alpha of a latent DNAbinding protein present in the cytoplasm of untreated cells. Proc Nat Acad Sci USA. 86:1203-7 Pellegrini S, John J, Shearer M, Kerr IM, Stark GR (1989). Use of a selectable marker regulated by alpha interferon to obtain mutations in the signaling pathway. Mol Cell Biol. 9:4605-12 Velazquez L, Fellous M, Stark GR, Pellegrini S (1992). A protein tyrosine kinase in the interferon alpha/beta signaling pathway. Cell. 70:313-22 Darnell JE Jr, Kerr IM, Stark GR (1994). Jak-STAT pathways and Transcriptional Activation in Response to IFNs and Other Extracellular Signaling Proteins. Science. 264:1415-21 LRI Laboratories Highlights 39 LRI Laboratories Highlight 16 The Hedgehog signalling pathway 1993: Phil Ingham’s lab clones the zebrafish hedgehog homologue, and predicts its importance as a morphogen. The hedgehog signalling pathway is the key to development of the vertebrate embryo. Multiple cancers exhibit hedgehog pathway activation and drugs that inhibit signalling through the pathway are in clinical trials. Background In 1980, Christiane Nüsslein-Volhard and Eric Wieschaus published a paper in Nature detailing the results of a screen they had devised in the fruit fly Drosophila Melanogaster to identify every gene required for early pattern formation in the Drosophila embryo. This work, for which, together with the American geneticist Ed Lewis, they were awarded the 1995 Nobel Prize in Physiology or Medicine, signalled the advent of modern fly developmental biology; researchers pounced on the many lines of mutant flies that had been generated, knowing that each had a mutation in just one gene, which it was now possible to isolate and clone by the new molecular biology techniques. Excitement spread when it was realised that many of the genes being cloned in Drosophila had vertebrate homologues, and that these similar genes likely had a similar role in the patterning of vertebrate embryos. Phil Ingham had done a PhD with Robert Whittle in Sussex, and by the mid-80s, was a postdoc with David Ish-Horowicz at the ICRF labs at Mill Hill, working, amongst other things, on fly segment polarity genes, which set the anterior/posterior Phil Ingham 40 Scientific report 2014 LONDON RESEARCH INSTITUTE (front to back) axis of each segment of a fly’s body. In 1987, in the new ICRF Developmental Biology Unit in Oxford, Phil started his own Molecular Embryology lab, and in the early 90s, his interest turned from flies towards vertebrate models, specifically, the zebrafish Danio rerio. He began a collaboration with Harvard scientists Andy McMahon, a mouse geneticist, and Cliff Tabin, who worked on chick limb development, to look at what a segment polarity gene named hedgehog was doing in vertebrates. In common with all other aspects of early development, segment polarity is achieved by a series of inductive interactions, which are of fundamental importance for the development of all multicellular organisms. Inductive interactions work rather like a very complicated domino run the preceding domino must fall correctly to trigger the next one, and so on, until the developmental process, such as the generation of a segment, or an organ, or a limb, or indeed a whole organism, is complete. They require cells to be able to signal to each other, either by direct contact between their proteins, or by using secreted messenger proteins (morphogens) to move between them, sometimes over long distances. In fly segment polarity, hedgehog (hh) seemed very likely to be a key gene; it was expressed in exactly the right place (the cells at the front edge) to specify the front-to-back organisation of each segment, and its protein product was secreted, meaning that it could move from one cell to another to transmit a signal. The research To isolate vertebrate hh, Phil and his collaborators began by screening molecular libraries containing all the genes from their respective organisms, and soon found that there was a small family of three vertebrate Hh genes which they christened Desert hedgehog (Dhh), Indian hedgehog (Ihh), and Sonic hedgehog (Shh). Phil’s lab showed that Shh turned out to be the most similar to fly hh, able to substitute for it in flies lacking their own hh gene. Zebrafish embryo The three collaborators then turned to analysing where Shh was switched on (expressed) during early vertebrate development, and discovered, to their great excitement, that Shh’s expression pattern made it very likely to be involved in two processes of extreme developmental importance, induction of floor plate, and ZPA signalling. Floor plate induction is responsible for the patterning of the neural tube, which eventually forms the spinal cord, and the ZPA, or Zone of Polarising Activity, is the region which specifies the correct development of limbs. Further experiments forcing expression of Shh protein in inappropriate places or times during development confirmed that it was very likely that Shh was the key morphogen being secreted both from the ZPA and from the notochord, the region responsible for inducing floor plate. The consequences A great deal of our knowledge of the mechanisms of early development stem from this work. The genes acting in both floor plate induction and the ZPA had been an almost complete mystery which had perplexed workers for many years, and the Shh work was the first chink of molecular light to be shed on the processes. In 1991, Phil had published an important paper predicting the interactions between Drosophila Hedgehog and its receptor, the Patched protein, and he and others built on this work to show that in vertebrates too, the Hh signal is relayed into the cell by the surface proteins Patched (PTCH) and Smoothened (SMO). In the absence of Hh, PTCH acts as a suppressor of SMO, and no signals are sent. Upon Hh binding, SMO is released, and activates a transcription factor called GLI, which moves into the nucleus and turns on Hh target genes. This work also has important implications for cancer treatment. Today, we know that there are few parts of the vertebrate body plan that are not in some way influenced by a Hh signal, but normally, Hh is almost completely silenced after birth. When Hh signalling is accidentally reactivated, it can cause cancer, sometimes by mutations in far downstream components of the pathway, but sometimes, in tumors such as colon, pancreatic, ovarian and basal cell carcinomas, because of mutation in the Hh ligands PTCH and SMO. So, if PTCH function is lost, or SMO is mutated in such a way that it is always on, the Hh signalling pathway fires even in the absence of Hh protein, causing cell overgrowth, and cancer. Basal cell carcinomas, where loss of PTCH is common, are highly treatable if caught early, but if not treated, progress to an advanced metastatic stage which has very poor prognosis and no good treatment. Development of small molecule inhibitors of SMO was begun by Ontogeny, a biotech start-up company whose inception was based on licensing the Shh patents filed by ICRF and Harvard following the discovery of Shh. Vismodegib (Erivedge), a drug inhibiting SMO activity developed by Curis Inc (Ontogeny’s successor) & Genetech, was licenced in 2012 to treat metastatic basal-cell carcinoma, and is in clinical trials as treatment for multiple other solid tumours. What happened next? Phil Ingham is currently Vice Dean, Research, and Toh Kian Chui Distinguished Professor at the Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore. Phil is a member of EMBO, a Fellow of the Academy of Medical Sciences and the Royal Society, and an Honorary Fellow of the Royal College of Physicians. He received the Medal of the Genetics Society of Great Britain in 2005 and the Waddington Medal of the British Society for Developmental Biology in 2014. He continues to work on the zebrafish hedgehog signalling pathway and its relevance to human disease. Key references Krauss S, Concordet JP, Ingham PW (1993). A functionally conserved homolog of the Drosophila segment polarity gene hh is expressed in tissues with polarizing activity in zebrafish embryos. Cell. 75:1431-44 Riddle R, Johnson R, Laufer E, Tabin C (1993). Sonic hedgehog mediates the polarizing activity of the ZPA. Cell. 75:1401-16 Echelard Y, Epstein DJ, St-Jacques B, Shen L, Mohler J, McMahon JA, McMahon AP (1993). Sonic hedgehog, a member of a family of putative signaling molecules, is implicated in the regulation of CNS polarity. Cell. 75:1417-30 LRI Laboratories Highlights 41 LRI Laboratories Highlight 17 DNA double-strand break repair, Holliday junctions, and BRCA2 1994: Steve West and colleagues purify human RAD51, a protein that plays a key role in DNA double strand break repair. West’s search for the elusive human Holliday junction resolvase that processes recombination intermediates finally succeeds in 2008, with the identification of GEN1. West’s work has been instrumental in understanding the implications of DNA double strand break repair both for tumourigenesis and for cancer therapy. Background Chromosomal double-strand breaks (DSBs) are one of the most dangerous forms of DNA damage. They can be induced by external agents, such as ionizing radiation or anti-cancer drugs, or by the cell itself, as in the case of the damage caused by free radicals generated from oxidative metabolism. Efficient double-strand break repair is essential for the survival of each cell because unrepaired breaks can lead to chromosome fragmentation and cell death, and improperly repaired breaks lead to mutations, chromosomal translocations and cancer. To preserve genomic integrity, we now know that DNA double-strand breaks are repaired in two ways: homologous recombination (HR) and non-homologous end joining (NHEJ). However, 25 years ago, very little was known about how these processes occurred in mammalian cells. In bacteria and yeast, matters were slightly more advanced, as it had been recognised that HR occurred, and several of the enzymes responsible had been identified. To study the mechanism of HR, the goal was to purify proteins and study their activities with DNA molecules in the test tube. To do this, Steve West 42 Scientific report 2014 LONDON RESEARCH INSTITUTE biochemical expertise is a key skill. As a graduate student in 1975 in Newcastle, Steve West identified the bacterial E. coli RecA protein, a central player in homologous recombination, and during his postdoc in Yale, he produced some classic papers on RecA, showing how it promoted the pairing of homologous strands of DNA and subsequent DNA strand exchange. These studies underpin much of our present understanding of the molecular mechanism of HR. The research RuvC: Steve West was recruited by Tomas Lindahl to the brand new ICRF Clare Hall Laboratories in 1985, and on his arrival, began working to biochemically purify the enzymes known as the Holliday junction resolvases, the proteins that catalyse the final resolution step in HR that allows the separation of recombinant chromosomes. He won the race to identify the E. coli enzymes that process Holliday junctions, christened RuvA, RuvB and RuvC, and characterised how they worked in a series of in vitro experiments using purified proteins and artificial DNA templates. Armed with their expertise in bacterial recombination, the focus of the West lab changed towards addressing the far more complex puzzle of identifying the enzymes involved in eukaryotic recombination. In the case of RuvC, this turned out to be a slightly knottier problem than anyone at the time could have anticipated, and it took until 2008 to identify and clone the human and yeast versions of RuvC, called GEN1 and Yen1 respectively. Unfortunately for the lab members involved, despite the functional similarity between the bacterial and eukaryotic resolvases, there was absolutely no sequence homology; in the case of human GEN1, brute force biochemical techniques had to be applied to harvest the tiny amounts of GEN1 in each cell, whilst yeast Yen1 was finally identified by genetic screens. RAD51 and BRCA2: In relation to West’s PhD project protein, bacterial RecA, things moved rather faster, and his lab was the first to purify the mammalian homologue of RecA, called RAD51, in 1994. Here, they found some remarkable similarities, showing that the RAD51 recombinase formed nucleoprotein filaments almost indistinguishable from those made by bacterial RecA. The subsequent discovery that RAD51 is both positively and negatively regulated by the BRCA2 protein in response to DNA damage has brought this work to centre stage in cancer biology. BRCA2 belongs to a class of genes known as tumour suppressors, as its normal function is essential in guarding the cell from possibly oncogenic mutations. It is defective in about 10% of inherited breast cancers, and when it is unable to work, double strand breaks cannot be repaired by homologous recombination, a defect that ultimately results in tumour formation. Biochemically, BRCA2 is a nightmare, as it is a huge protein nearly 400kD in size, which makes working with it very challenging due to the difficulty in purifying the full-length protein and its propensity to fall apart. As a result, little was known about the precise mechanisms by which it worked. However, a 2010 publication from the West lab, in which BRCA2 was purified and studied in complexes by electron microscopy has now shown that it is responsible for controlling the ability of RAD51 to bind DNA. These observations have at long last provided the molecular explanation for the role of BRCA2 in the maintenance of genome stability. Recombination and disease: Not surprisingly, defects in DNA repair lead to a number of rather unpleasant and crippling diseases, and the West lab has been at the forefront of research into the causes of two of these, Bloom’s Syndrome and Fanconi Anaemia. Recently, work from the West group has defined the molecular defect associated with a neurological disorder known as Ataxia with Oculomotor Apraxia-1 (AOA1). AOA1 is a progressive disease, first manifesting in apparently healthy children at about 7 years of age. It begins with difficulty in coordinating movement (ataxia), and many sufferers also develop an inability to move their eyes to look sideways (oculomotor apraxia). Symptoms increase in severity such that after ten years, most sufferers have atrophied muscles and nerves, and sometimes limb deformities, meaning that they are typically wheelchair bound. One gene, APTX, encoding Aprataxin, is defective in AOA1 patients, and the West lab showed that Aprataxin protein is the proofreader which detects abortive DNA ligations, the process whereby two pieces of broken DNA are sewn back together. All cells, especially neuronal cells, are subjected to high levels of oxidative stress resulting in the formation of DNA strand breaks. When these breaks are repaired by a DNA ligase, it is not uncommon for the reaction to stall at an intermediate stage, such that abortive ligation intermediates accumulate. It turns out that Aprataxin specifically interacts with these intermediates, allowing them to be resolved. When no Aprataxin is present, unrepaired nicks accumulate in the DNA, and eventually, there are so many that the machinery responsible for transcription (turning the DNA into RNA transcripts, ready to make protein) is unable to work properly and breaks down. Without efficient transcription, the neuronal cells die. Interestingly, while the AOA1 defect is present in most tissues, it is only a problem in the brain. The reason is that brain neurons are post-mitotic, that is, they no longer replicate, and so they lack all the replication-associated DNA repair mechanisms which are able to sort out stalled ligases by other means; with the loss of Aprataxin activity, they lose their only safeguard. What happened next? Steve West is currently the Deputy Director of the LRI Clare Hall Laboratories. He is an EMBO member and a Fellow of the Royal Society and the Academy of Medical Sciences. His awards include the 2002 Leeuwenhoek Prize of the Royal Society, the 2007 Louis-Jeantet Prize for Medicine, the 2008 Novartis Medal and Prize from the Biochemical Society, and the 2010 GlaxoSmithKline Medal and Prize of the Royal Society. Key references Dunderdale HJ, Benson FE, Parsons CA, Sharples GJ, Lloyd RG, West SC (1991). Formation and resolution of recombination intermediates by E. coli RecA and RuvC proteins. Nature. 354:506-10 Baumann P, Benson FE, West SC (1996). Human RAD51 protein promotes ATP-dependent homologous pairing and strand transfer reactions in vitro. Cell. 87:757-66 Ip SCY, Rass U, Blanco MG, Flynn HR, Skehel JM, West SC (2008). Identification of Holliday junction resolvases from humans and yeast. Nature. 456:357-61 Thorslund T, McIlwraith MJ, Compton SA, Lekomtsev S, Petronczki M, Griffith JD, West SC. (2010). The breast cancer tumour suppressor BRCA2 promotes the specific targeting of RAD51 to single-stranded DNA. Nat Struct Mol Biol. 7:1263-5 LRI Laboratories Highlights 43 LRI Laboratories Highlight 18 Cancer genetics 1979: Walter Bodmer establishes the ICRF as a major centre for cancer genetics. 1987: Walter Bodmer and Ellen Solomon identify the FAP locus. 2007 and 2008: Tomlinson lab publishes collaborative studies identifying loci in genome associated with susceptibility to breast, colorectal and prostate cancer. 44 Background In 1979, when Sir Walter Bodmer succeeded Sir Michael Stoker as Director of the ICRF, his appointment signalled the start of a new initiative at the lab, in human genetics. One of Bodmer’s primary research interests was the study of the genetic basis of cancer, the notion that at the molecular level, cancer is caused by changes in genomic DNA. These changes can be induced by damage or mutation, but can also be hereditary; families in which a particular sort of cancer is common have a defect in a gene or genes making them susceptible to the disease. Finding out what these susceptibility genes are has been a major cancer research goal since the 1980s. Their identification allows for genetic testing of potential carriers, enhances understanding of the basic biology of cancer, and often sheds light on how sporadic (non-hereditary) tumours form. The ICRF has provided two particularly good examples of this progression from gene identification to biological understanding. Bodmer’s lab, together with that of Ellen Solomon, mapped the location of the gene responsible for a hereditary type of bowel cancer, familial adenomatous polyposis (FAP), in 1987. The APC gene which lay within the FAP locus was cloned by Kinzler and colleagues in 1991, and is mutated in some 80% of sporadic colon cancers. Work from many laboratories has shown that APC lies at the centre of a network of molecules, many of which are also implicated in colon and other cancers. In a somewhat different arena, Peter Goodfellow, another human geneticist recruited by Bodmer, in 1990 identified the SRY gene (Highlight 9), which is responsible for determining the sex of males during embryonic development. Walter Bodmer Ian Tomlinson Scientific report 2014 LONDON RESEARCH INSTITUTE Ian Tomlinson, after postdoctoral work in the Bodmer lab and at the Institute of Cancer Research, started his own lab at the ICRF in 1998, and in the next decade, was involved in a collaborative effort searching for common genes which increase the risk of cancer. His lab contributed to studies to identify loci contributing to breast and prostate cancer, and his work on colorectal cancer produced a series of high profile publications in 2007 and 2008. The research Working in a field – cancer genetics – in which big screens for new disease genes are the norm, Ian Tomlinson’s work required the collection of tens of thousands of patient samples, representing huge efforts for several members of his laboratory and the laboratories of collaborators. The main developments that made this venture possible were technological and financial, owing to the willingness of Cancer Research UK to fund what remains very expensive work. Ian’s group, together with colleagues at the Institute of Cancer Research and in Edinburgh, started searching for common alleles that increased the risk of developing colorectal cancer, in the hope that by fully understanding the genetic architecture of cancer, it would be possible to predict individual risk and thus tailor effective cancer prevention measures to those at relatively high risk. A secondary justification was to obtain information about disease biology, by identifying some of the functional pathways that might usefully be targeted by, for example, new chemopreventive agents. The project design was a relatively simple association study, based on comparing genotype frequencies in cases and controls and searching for significant differences at individual loci. 10 tagSNPs (genomic markers) associated with differential risks of disease were identified. In one case, the SNP was also associated with increased risks of prostate and ovarian cancer, but the other 9 SNPs seemed unique to colorectal cancer. In all cases, the effects on risk were relatively modest, with typically a 1.1-1.3-fold differential risk per allele (although these estimates are conservative). These SNPs seemed to act independently of one another, with no instances of gene-gene interactions and little deviation from additive or log-additive effects on risk. Perhaps most intriguingly, there was some evidence that several of the colorectal cancer SNPs acted in the same functional pathway, namely bone morphogenetic protein (BMP) signalling. Inhibition of BMP signalling was already proposed to have an essential function in maintaining the stem cell niche at the bottom of the colonic crypt. If, as is thought, cancer arises from normal stem cells, it is therefore possible that the colorectal cancer SNPs affected stem cell numbers, and hence the number of cells that could potentially give rise to cancer, thereby increasing cancer risk. Intriguingly, several colorectal cancer SNPs lay in regions where genes in the conventional sense were absent; thus, their most likely function was in regulation of gene expression, potentially over hundreds of thousands of kilobases. The consequences Such genome-wide association studies have taken genetics into new areas in which the science has become much more similar to epidemiology than ‘classical’ genetics, and individual effort is pooled for the good of a greater whole. Whilst intellectually less satisfying, requiring project management rather than purely scientific skills, such studies are essential to make the advances in clinical research necessary to treat many diseases. However, part of their attraction is also to provide seedcorn for functional studies on the genes identified, so that it will be possible to work out exactly what the differences are between individuals that cause variation in disease risk, and to work out how these differences have their effects. What happened next? Walter Bodmer became ICRF’s first Director General in 1991. In 1996 he left the ICRF to become Principal of Hertford College Oxford, from which he retired in 2005. He is currently Head of the Cancer and Immunogenetics Laboratory in the Weatherall Institute of Molecular Medicine at the University of Oxford. Ellen Solomon moved to King’s College London in 1995, where she is Professor of Cancer Genetics in the Division of Genetics and Molecular Medicine. Ian Tomlinson moved to the Wellcome Trust Centre for Human Genetics in Oxford in 2008. He continues to work on the genetics of colorectal and other cancers, with an emphasis on cancer predisposition. For his contributions to the field, he was elected to the Academy of Medical Sciences in 2009, and in 2013 won the UEG Research Prize – an award of €100,000 for excellence in basic science or clinical and translational research. Key references Tomlinson I, et al., (2007). A genome-wide association scan of tag SNPs identifies a susceptibility variant for colorectal cancer at 8q24.21. Nat Genet. 39:984-8 Houlston RS, et al., (2008). Meta-analysis of genome-wide association data identifies four new susceptibility loci for colorectal cancer. Nat Genet. 40:1426-35 Tenesa A, et al., (2008). Genome-wide association scan identifies a colorectal cancer susceptibility locus on 11q23 and replicates risk loci at 8q24 and 18q21. Nat Genet. 40:631-7 Tomlinson IP, et al., (2008). A genome-wide association study identifies colorectal cancer susceptibility loci on chromosomes 10p14 and 8q23.3. Nat Genet. 40:623-30 LRI Laboratories Highlights 45 LRI Laboratories Highlight 19 Innate immunity Caetano Reis e Sousa has made multiple key discoveries concerning how dendritic cells of the innate immune system recognise pathogen associated molecular structures or normal cell constituents generated by dying cells. Background The vertebrate immune system fights infection using two closely interlinked defence networks. Adaptive immunity, mediated by T- and B-lymphocytes, learns from previous battles fought with enemy antigens, honing their specificity and response time so that the body is never taken unawares if reinfected. However, generation of T and B cell responses relies upon signals from the far more ancient innate immune system, which is found in all plants and animals, and from which adaptive immunity evolved. Central to the whole process are the dendritic cells, a heterogeneous family of white blood cells that integrates innate information and conveys it to lymphocytes. activate the T cell branch of the adaptive immune system. Different threats lead to differences in the mix, producing a precisely tailored coded message telling the microbe-specific T cells how they have to react, or differentiate, to combat the incoming microbes. Infection by different types of micro-organism creates distinctive tell-tale clues, or patterns. The various dendritic cell subtypes work by recognising these patterns, using a network of so-called pattern recognition receptors and other innate immune receptors which are programmed to recognise deviations from cellular normality: for example, molecules that look right, but are in the wrong place, or dead cells cropping up where they should not, are all detected by different classes of dendritic cells. Once the dendritic cell has worked out exactly what the threat is, it produces a mix of cytokines and other factors, which are able to The research Right face, wrong place: Flu and other similar viruses are recognised by dendritic cells and other cells, which set up an antiviral response, triggering rapid production of type 1 interferons and activating the adaptive arm of the immune system, as well as potentiating innate antiviral resistance. However, both the viral clues being detected, and the identity of the molecular sensors on the dendritic cells were unknown. Caetano and collaborators, in a series of papers published between 2004 and 2010, shed light on these mysteries, and established the concept that mislocalisation of an ostensibly innocent molecule is seen by dendritic cells as an extremely suspicious event. They discovered that dendritic cells have evolved several mechanisms for distinguishing viral RNA from cellular RNA, relying on the fact that viral RNA crops up in places where cellular RNA would never normally be found. For example, two of the detectors, the TLR7 and TLR3 receptors, live in the endosomes, the vesicles that capture material at the cell surface and transport it around inside the cell. As cellular RNA is never found in endosomes, the cell can be certain that any RNA found there is suspect, and this provides a mechanism by which dendritic cells can detect the presence of virally infected cell corpses. RNA is also the key to the third detection system, which is cytoplasmic, and uses the RIG-I protein which recognises RNAs with a particular modification (a 5’ triphosphate group) Caetano Reis e Sousa 46 Scientific report 2014 LONDON RESEARCH INSTITUTE Work from Caetano Reis e Sousa’s lab has helped define what some of the most important microbial signatures are, how dendritic cells recognise these signatures, and how they then transmit this information to the adaptive immune system. His work has important implications for tumour immunotherapy, where the body’s immune system is used to destroy cancers. at one end. Again, this mark is diagnostic of viral RNAs and provides a way by which viral infection can specifically activate RIG-I. Disease and death; a Syk story: One of Caetano’s long standing interests was to crack the cytokine code - to work out the different mixes of cytokines elicited by different types of microbial infection. Whilst doing this, he realised that when he gave dendritic cells yeast as the microbial agent, they produced cytokines as expected, but intriguingly, did so independently of known TLR family members. His lab began hunting for this unknown signalling pathway, and in 2005, published a landmark paper identifying the Syk tyrosine kinase as the key player. Syk is activated by Dectin-1, which recognises carbohydrates uniquely present on fungal cell walls. The lab also showed that a second receptor, Dectin-2, could also signal via the Syk pathway, and that together, Dectin-1, Dectin-2 and the Syk pathway are necessary for warning the immune system about all types of fungal infection. Interestingly, some patients with chronic mucocutaneous candidiasis turn out to have mutations in components of this novel pattern recognition pathway, highlighting its importance in human defence from fungal infection. The Syk pathway is important for more than just fungal infection. Another common sign of infection or injury is the presence of cells dying by necrosis, a form of death occurring during trauma. Caetano’s lab has recently characterised another C-type lectin family member called DNGR-1 as the bridge between recognising necrosis, and alerting the T cells of the adaptive immune system. DNGR-1’s discovery is provocative as it resurrects the concept of an innate ‘danger receptor’, a molecule able to sense abnormal cell death and trigger an adaptive immune response, first proposed by the American immunologist Polly Matzinger. The consequences Dendritic cells are a very hot topic in terms of cancer therapeutics, as if it were possible to harness their power to activate the adaptive immune system, and use it to attack and destroy tumours, the body would be able to assist in ridding itself of cancers. The fact that DNGR1 is only found on one particular sort of mouse dendritic cell, the CD8a+ subtype, makes it a potentially exciting target for cancer therapeutics, as CD8a+ dendritic cells are very efficient at presenting antigens to the killer T cells of the mouse adaptive immune system. Caetano’s lab has shown that if antigens derived from melanomas are artificially fused to antibodies against DNGR-1, these hybrid molecules bind to DNGR-1 on CD8a+ dendritic cells, and generate a strong response against melanoma, shrinking hard-to-treat tumours to almost nothing in experimental mouse models. Vexingly, CD8a+ dendritic cells had not been identified in species other than mouse. Caetano’s lab recently used the specificity of DNGR-1 expression to identify the human equivalents of mouse CD8a+ dendritic cells, bringing the translational potential of DNGR-1 targeting one step closer to the clinic. What happened next? Caetano Reis e Sousa has become a world leader in the field of innate immunity. He was elected to EMBO and to the Fellowship of the Academy of Medical Sciences in 2006, won the Liliane Bettencourt Life Sciences Award in 2008, and the ESCI Award for Excellence in Basic/Translational Research in 2011. Key references Schulz O, Diebold SS, Chen M, Naslund TI, Nolte MA, Alexopoulou L, Azuma Y-T, Flavell RA, Liljestrom P, Reis e Sousa C (2005). Toll-like receptor 3 promotes crosspriming to virus-infected cells. Nature. 433:887-92 Pichlmair A, Schulz O, Tan CP, Naslund TI, Liljestrom P, Weber F, Reis e Sousa C (2006). RIG-I–Mediated Antiviral Responses to SingleStranded RNA Bearing 5’-Phosphates. Science. 314:997-1001 Robinson MJ, Osorio F, Rosas M, Freitas RP, Schweighoffer E, Gross O, SjefVerbeek J, Ruland J, Tybulewicz V, Brown GD, Moita LF, Taylor PR, Reis e Sousa C (2009). Dectin-2 is a Syk-coupled pattern recognition receptor crucial for Th17 responses to fungal infection. J Exp Med. 206:2037-51 Sancho D, Joffre O, Keller A, Rogers NC, Martinez D, Hernanz-Falcón P, Rosewell I, Reis e Sousa C (2009). Identification of a dendritic cell receptor that couples sensing of necrosis to immunity. Nature. 458:899-903 Rehwinkel J, Tan CP, Goubau D, Schulz O, Pichlmair A, Bier K, Robb N, Vreede F, Barclay W, Fodor E, Reis e Sousa C (2010). RIG-I detects viral genomic RNA during negative-strand RNA virus infection. Cell. 140:397-408 Ahrens S, Zelenay S, Sancho D, Hanč P, Kjær S, Feest C, Fletcher G, Durkin C, Postigo A, Skehel M, Batista F, Thompson B, Way M, Reis e Sousa C, Schulz O (2012). F-actin is an evolutionarily conserved damage-associated molecular pattern recognized by DNGR-1, a receptor for dead cells. Immunity. 36:635-45 LRI Laboratories Highlights 47 LRI LABORATORIES Highlight 20 Tumour heterogeneity 2012: Charlie Swanton and colleagues show that tumours are composed of a continually evolving mixed population of cancer cells, explaining why advanced cancer is so hard to treat. Cancer heterogeneity arises through Darwinian evolution, and nipping the evolutionary process in the bud by early diagnosis and detection will be the key to successfully combatting the disease. The background In the first decade of this century, our understanding of the molecular mechanisms underlying many human cancers started a revolution in personalised cancer treatment. Tumours could be biopsied to determine which of the cancer-causing oncogenes and tumour suppressor genes were mutated, and targeted drugs against the offending molecules could be deployed to attack the root of the cancer. There was a wave of optimism in the cancer community, but very quickly, it became clear that all was not well; whilst there were some startling successes, with patients plucked from the jaws of death by tailored therapies, the grim reality was that after weeks or months, the therapies would fail as the tumour fought back, with fatal consequences. The question of why advanced cancers were so lethally good at acquiring resistance to drugs was not a new one, and an answer had in fact been proposed in 1976 by Peter Nowell, who suggested that cancer was an evolutionary disease: as tumours were genetically unstable, the selective pressure applied by aggressive therapy resulted in subpopulations of tumour cells acquiring resistance, thereby flourishing anew. However, Charlie Swanton 48 Scientific report 2014 LONDON RESEARCH INSTITUTE whilst biopsies of advancing disease showed correlations between new chromosomal abnormalities and drug resistance, the limited techniques of the time meant that finding out what was going on at the molecular level was impossible. By 2010, technology had advanced enormously and several groups, one led by former ICRF star Mel Greaves (Highlight 3), published landmark papers on the molecular archaeology of cancer. Mel’s group, looking at acute leukaemia, proposed that instead of proceeding inexorably from normality to fatality by a series of linear mutational events, leukaemias exhibited branched evolution, just as in Darwin’s iconic evolutionary tree diagram. Two other publications showed this was also the case in solid tumours: branched evolution meant that primary tumours and metastases were genetically distinct. Together, the three papers confirmed Nowell’s theory, and suggested that tumour heterogeneity might be more extensive than previously suspected. Charlie Swanton’s double life as an oncology consultant and a scientist interested in translating research into effective anti-cancer therapeutics meant that he was perfectly placed to tackle the problem of tumour heterogeneity head-on. Following his arrival as a group leader at the LRI in 2008, Charlie and a group of collaborators decided to look at tumour evolution in unprecedented detail. There was a real urgency for this research: if Charlie’s experiments showed that there was diversity between biopsies within the same primary cancer, the implication for treatment and predicting outcome using genomics-based diagnosis in solid tumours was alarming: the long-established practice of taking only a single biopsy of a tumour, and basing therapeutic decisions on that one sample, might mean that clinicians were prescribing targeted drugs that would kill only a small subset of cancer cells, leaving the rest of the tumour unharmed. The research Charlie and his collaborators took multiple biopsies from different regions of primary kidney cancers and metastases, sequenced the tumour DNA, and also looked at chromosomal abnormalities and changes in gene expression. The sequencing alone was a huge job: to ensure total accuracy, each tumour DNA sample had to be sequenced many times, and the LRI sequencing facility worked solidly for 4 months to read around 140 billion bases of DNA code. The results exceeded everyone’s worst suspicions: even before extensive treatment had commenced, primary tumours were already heterogeneous; no two samples were the same, even if they’d come from adjacent regions of one tumour. In the primary tumours, only about a third of the mutations detected were common to all the samples. Even more worryingly, genetic signatures known to be associated with either a good or a poor prognosis could be picked up in different regions of the same tumour. The stark reality was that a single biopsy would never provide a picture of the mutational landscape of the entire tumour, and frighteningly, could be extremely misleading. Analysis of the mutated genes did provide one encouraging fact; although there were only one or two driver mutations common to all the samples, there were quite a few examples of parallel evolution, in which different tumour samples from the same tumour had mutated the same genes, but in different ways. It looked as though progression to kidney cancer absolutely required the mutation of certain cellular pathways, and some of these key pathways were already therapeutic targets. What happened next? There was a degree of scepticism in the cancer community following the publication of Charlie’s paper, with some disbelief that intratumour heterogeneity was so extreme. However, his results have been validated in most solid tumours, and what was once heterodox has now become an accepted and rather obvious fact. After the initial gloom prompted by the realisation that their enemy was even more devious than suspected, cancer biologists have returned to the fray with renewed vigour and better plans. In depth analysis of as many cancer types and tumours as possible will eventually give the field a clear idea of which mutations lie in the ‘trunk’ of a cancer’s evolutionary tree, and also which pathways exhibit parallel evolution. Once identified, therapies directed at such driver mutations will be far more likely to succeed than those against less ubiquitous molecules, and more reliable prognostic tests can also be developed, using the tens of thousands of biomarkers already known to be associated with cancer. The recognition that cancers evolve, and that more and more mutations accumulate as time goes by, has also emphasised the importance of early detection of disease. Catching and containing cancer before it goes out of control is becoming a major priority, as has defining exactly how diversity is generated. In 2014, Charlie’s lab showed that in colon cancer, following a genome doubling event, tetraploid colon cancer cells became tolerant of ongoing chromosomal instability. These tetraploid supergenomes were strongly associated with both poor prognosis and in the laboratory evolved much faster than their diploid progenitors. Such extravagantly mutated cells are an extreme example of chromosome instability, a common and high risk feature of cancer, and working out the mechanisms by which the instability arises is an ongoing preoccupation for many labs around the world, including Charlie’s.Whilst the basic research continues, Charlie is trying to put his findings to clinical use by running TRACERx, a multi-millionpound multi-centre collaborative trial for lung cancer, following 850 patients through therapy. By sampling tumours at all stages of treatment, he can map the ebb and flow of the different variants in the tumours by DNA sequencing. The speed of sampling and sequencing means that what he learns can then be used to inform the next stage of treatment, keeping clinicians one step ahead of the cancer. In addition to this immediate therapeutic benefit, Charlie will use the sequencing data from the project to understand the evolutionary biology of lung cancer, and to link the more common types of therapeutic drug resistance to particular genes, making it even faster and easier to monitor changes in a patient’s response to treatment. Key references Gerlinger M, et al., (2012). Intratumor Heterogeneity and Branched Evolution Revealed by Multiregion Sequencing. N Eng J Med. 366:883-92 Gerlinger M, et al., (2014). Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet. 46:225-33 Dewhurst SM, et al., (2014). Tolerance of wholegenome doubling propagates chromosomal instability and accelerates cancer genome evolution. Cancer Discov. 4:175-85 LRI Laboratories Highlights 49 INTRODUCTION TO THE 2014 LRI RESEARCH REPORT 2014 saw the last full year of operation of the London Research Institute before our coming incorporation into the Crick in 2015. The Research Highlights section attests to how the Institute continues to conduct its science largely as usual, but increasingly, Institute staff have been contributing to preparations for the transfer of operations to the new Crick laboratories. Richard Treisman Peter Van Loo, Cancer Genomics Fields to returned to his native Germany, taking up a position at the Max Delbrück Center for Molecular Medicine (MDC) in Berlin. The same month, Mark Petronczki left Clare Hall to joined Boehringer Ingelheim in Vienna, also a return to his Austrian roots. We wish both of them all success in the future. Group leader recruitment to LRI has wound down as the move to the Crick approaches, but we continue to see both new arrivals, and departures as established group leaders move on to take up new research opportunities. The past year saw the arrival of two new group leaders, hired under the Crick’s ‘6+6’ Junior Group Leader scheme. Peter Van Loo joined us in September from the Sanger Institute. A bioinformatician, Peter is especially interested in genetic variation in cancer, and pursues genetic approaches to elucidate the evolutionary history of tumours and their metastases. His work will complement the increasing interest in cancer genomics and evolution at LRI. In contrast, Guillaume Salbreux, who joined the Institute in February 2015, is a soft-matter physicist. This area is particularly relevant to understanding the movement of cells, whether during tissue and organ development or migration of individual cells. In September we said farewell to Holger Gerhardt, who left Lincoln’s Inn While these new arrivals reinforce the continued renewal and growth of the Institute, our last year as LRI has been also touched by the deaths of two of its senior members, who will be sorely missed. In April, Julian Lewis succumbed to prostate cancer, which he had fought valiantly for several years. Julian’s kind and modest personality was combined with intellectual rigour, great clarity of expression, and broad interests, both personal and scientific. The fluency and elegance of his writing was displayed in his beautifully written papers, and in his contributions to ‘The Molecular Biology of the Cell’, of which he was a long-serving Julian Lewis Francois Lassailly Guillaume Salbreux, Theoretical Physics of Biology 50 Scientific report 2014 LONDON RESEARCH INSTITUTE Sally Leevers, LRI Academic Director Donna Brown, NIMR Director of Studies co-author. He had continued to write following his retirement from LRI in 2012, and his last paper appeared only weeks before his death. Julian was remembered by scores of colleagues and friends at a memorial gathering held in July at Balliol College Oxford, of which he was a fellow. As these words were being finalised in January 2015, we were shocked by the death of Francois Lassailly in a traffic accident. Francois was instrumental in the introduction of non-invasive animal imaging methods to LRI, and was working hard to establish the new Crick Institute animal imaging facility; many researchers at LRI will have experienced Francois’ good nature, enthusiasm, and skill at first hand. Research activity and academic life at LRI has seen both continuity and new developments over the last year. Outstanding research achievements by Institute staff continue to be recognised by our peers. Erik Sahai joined the ranks of LRI EMBO members in Spring, while Charlie Swanton was awarded the 2015 Laura Ziskin prize for translational cancer research from the Stand up to Cancer in the US. Congratulations to them both. Marcus Wilson from Jesper Svejstrup’s group was awarded the 2013 Pontecorvo prize for the best CRUK-funded PhD thesis, while Andy Filby from the Flow Cytometry facility was chosen as an Emerging Leader by the International Society for the Advancement of Cytometry. The last-ever LRI retreat was held at the Oxford Examination Schools, at which Barbara Schraml presented the Hardimon-Redon lecture. Presentation of the 2014 Hardiman-Redon Prize will form part of the handover ceremony on the LRI’s last day at the end of March 2015. Finally two very enjoyable Crick Sophie Acton, winner of the 2014 Hardiman-Redon Prize group leader retreats introduced LRI scientists to future colleagues, from GSK, at London Zoo in July, and from the Wellcome Trust Sanger Institute, at Keble College Oxford in September. Over the past few years the profile of the Crick amongst our junior trainees – graduate students and postdocs – has become more and more prominent. Development and implementation of the Crick Graduate programme has been a major activity for Sally Leevers and her opposite number at NIMR, Donna Brown. Much hard work on their part led to the formal finalisation of the regulations governing the Crick graduate student programme with the Crick’s University partners. As a result, the 2014 graduate student intake is the first to be admitted entirely under Crick auspices. The Crick held its third annual postdoc retreat at the British Library in June, bringing together some 200 postdocs from LRI, NIMR and the Crick partners, and featuring for an interactive day of science, career advice and networking, including a stimulating discussion about open-access science publishing. The opening of the new Lincoln’s Inn Fields laboratories in 1963 followed an appeal for a million pounds by the ICRF, CRUK’s predecessor. It is sobering to note that 50 years on, the Create the Change campaign is seeking 100 million pounds to fund the CRUK capital contribution to the Crick project, and as I write the campaign is well on track towards this target. In closing I would like to thank not only Charles Manby and the CtC Board, but also Antonia Newman, Russell Delew and their team for all their hard work, without which the Crick would not have been possible. Antonia Newman and Russell Delew, Create the Change DIRECTOR'S INTRODUCTION 51 Francis crick institute report During 2014, the Crick Laboratories attained their final profile, and the main emphasis on the building project has moved towards the fitting-out of what will be one of the most technologically complex buildings in Europe. As the building project moved steadily closer to completion, 2014 saw a broad range of activities aimed at establishing the Crick as a functional research organisation in readiness for our migration to the new building in late 2015. David Roblin There have been a number of important changes in the Crick’s leadership during the year in preparation for first year of operations, which will be on the old LRI and NIMR laboratory sites. David Roblin took over as COO and Director of Scientific Translation in September, with John Cooper moving to Projects Director. Two new committees, the Science Strategy and Policy Committee and the Operations Management Committee, were established to lead on research and operational matters respectively, chaired respectively by Paul Nurse and David Roblin. In readiness for the research operation, three new associate research Directors were appointed in December: Julian Downward, leading on STPs and BRF; John Diffley, leading on Junior Researchers and Training; and Anne O’Garra, leading on Group Leader Development. Andy Smith Francis Crick Institute, February 2015 52 Scientific report 2014 LONDON RESEARCH INSTITUTE Michael Schuitevoerder and his team have worked tirelessly during the year to develop the migration plan for occupation of the new building. The challenge here was to develop a robust plan by which the science of the founding institutes can be moved from three laboratory sites into the Crick laboratories without significant down-time. Following the building handover on November 3rd 2015, central support functions will move in first, and IT and animal house facilities will be made ready. The STPs will move over Christmas, with some being phased to allow operations to continue at the former Institute sites pending complete transfer to St Pancas. Research laboratories will move in starting early 2016, and the move will be complete shortly after the centenary of Francis Crick’s birth on June 8th. Alongside this planning, work to bring the NIMR and LRI mouse populations Francis Crick Institute atrium, February 2015 to a uniform Crick health status, guided by Kathleen Mathers and Gary Childs, proceeded steadily during the year. This will allow animal transfer to the new building to occur without delay once the new animal facilities are commissioned. Over the year several important transitional appointments were made, critical for the integration of laboratory operations and science technology platforms and migration of research activities to the new laboratories following the Crick’s initial period of multi-site operations. In January Simon Caidan, David Hudson, Jo Payne and Nigel Peat were appointed to lead on Health and Safety, STP operations, Lab infrastructure and logistics, and Lab Operations respectively. Later in the year, transition science technology platform leads were appointed from each of the two Institutes to lead integration of support technologies. The year’s end saw the appointment of the Laboratory Operations Managers, who will oversee the operation of each of the research floors of the new laboratories, each appointee looking after the laboratories on one sector of each floor. At the time of writing they are working with the transition team to finalise equipment lists and placings, and develop the logistics of the migration. The nascent Crick Institute’s academic life has continued to develop. Late in 2014 the Crick issued a pilot call for the first wave of university researchers to join the Crick, with a special focus on interdisciplinarity. It is anticipated that these will include research groups applying for secondment to the Crick laboratories, smaller ‘satellite’ groups that will be attached to Crick research groups for specific collaborative work, and sabbatical placements. These groups will join the Crick upon the completion of the initial migration. Joining then will be Akhilesh Reddy, a clinican scientist working on circadian rhythm biology, who will join UCL and the Crick from Cambridge. Ak will hold one of three Crick Clinical Professorships at the partner universities, in his case at UCL. Finally, a new interim Crick Faculty Committee met for the first time in November. This committee provides a way for the Crick’s management to consult with the academic staff concerning science policy, initiatives, and strategy. It will operate in interim form, comprising representatives of the founding institutes until the migration is complete, when it will be reconstituted to include members from the Crick’s partner universities. Finally the Crick continued to engage with its staff and the public through its programme of Crick scientific symposia, staff away-days and diverse public engagement events organised by the Comms team. Particularly notable amongst these was a Crick-led ‘Science Museum Lates’ event in February. The event gave visitors the chance to mingle with researchers from the Crick’s partners to take part in a range of experiments and activities at the Museum. A record attendance of 6900 people took advantage of the various opportunities, which included photographing developing zebra fish on their smartphones, creating and drinking a DNA cocktail, knitting a blood vessel and meeting some of the twins taking part in a long-term study in epigenetics. It is now only weeks before the Crick starts operations: our first year will be one of tremendous excitement, with our move of our laboratories to the new building. The following years will surely be more exciting still as we begin to implement the vision of what promises to be the most significant development in UK biomedical science in a generation. DIRECTOR'S INTRODUCTION 53 RESEARCH HIGHLIGHTS Every year the Report features highlights of Institute research, summarising the findings in terms accessible to the non-specialist scientific reader, and each year the Hardiman-Redon Prize is awarded to a junior researcher who has made outstanding contributions to one of these highlighted publications. Our final year has seen an excellent output of remarkable papers, and the 2014 HardimanRedon Prize is awarded to Sophie Acton from the Immunobiology Group headed by Caetano Reis e Sousa. Dendritic cells control fibroblastic reticular network tension and lymph node expansion. Acton SE, Farrugia AJ, Astarita JL, Mourão-Sá D, Jenkins RP, Nye E, Hooper S, van Blijswijk J, Rogers NC, Snelgrove KJ, Rosewell I, Moita LF, Stamp G, Turley SJ, Sahai E, Reis e Sousa C. Nature. 2014; 14(7523):498-502 Lymph node swelling is the hallmark of an adaptive immune response and is driven by an influx of lymphocytes from the blood followed by their eventual antigen-driven proliferation. Throughout the expansion phase, the lymph node must maintain the integrity of the stromal network that acts as the organ’s scaffold and marks the ‘paths’ on which immune cells travel and meet one another. In this paper, we show that a major stromal cell type in the T cell areas of lymph nodes, the fibroblastic reticular cells (FRCs), stretch rapidly in response to signals from dendritic cells (DCs) and thereby permit organ expansion driven by lymphocyte influx. Mechanistically, DCs provide Figure 1 Fluorescence microscope image showing insulin-positive β cells (red) in pancreatic ducts (green) in mice lacking Fbw7. 54 Scientific report 2014 LONDON RESEARCH INSTITUTE a transmembrane protein known as CLEC-2 that engages another known as podoplanin and which is expressed by FRCs. Podoplanin ligation by CLEC-2 causes it to re-distribute on the FRC membrane and stops its signalling via RhoA/C and ROCK to the cells’ actin-based contractile machinery. The net result is a relaxation of the FRC actomyosin cytoskeleton that permits cell stretching. Notably, CLEC-2 is primarily expressed at high levels on those DCs that immigrate into lymph nodes from sites of inflammation and deliver antigens and co-stimulatory signals to T cells. This study indicates that DCs additionally contribute to adaptive immunity by delivering the key signals for lymph node remodelling. Loss of Fbw7 reprograms adult pancreatic ductal cells into α, δ and β cells. Sancho R, Gruber R, Gu G, Behrens A. Cell Stem Cell. 2014; 15(2):139-153 The hormone insulin regulates blood glucose levels and is produced in the pancreas by the β cells. In Type 1 diabetes, the β cells are destroyed and so patients need to control their blood glucose levels with insulin injections. A better approach would be to generate replacement β cells that could be transplanted into the patient to improve blood glucose regulation in the long term. However, β cells do not regenerate in the adult, and so, finding a ‘progenitor cell’ that can produce new β cells is an important goal of diabetes research. In this study by Rocio Sancho, our group has shown that rare pancreatic duct cells can be reprogrammed into β cells in mice by inactivation of a single gene, Fbw7. These new β cells respond to glucose and release insulin in the same way as ordinary β cells do. This study shows that rare duct cells can act as pancreatic progenitor cells, raising the possibility that they could be used to generate replacement β cells for therapy (Figure 1). Control mice Usp28-deleted mice Figure 2 Survival curve of mice showing that Usp28 deletion in established intestinal tumours prolongs survival (green line). % Survival 100 50 0 0 50 100 150 Days post Usp28 deletion The ubiquitin protease Usp28 controls intestinal homeostasis and promotes colorectal cancer. Diefenbacher ME, Popov N, Blake SM, Schülein-Völk C, Nye E, Spencer-Dene B, Jaenicke LA, Eilers M, Behrens A. Journal of Clinical Investigation. 2014; 124(8):3407-3418 The lining of the gut is constantly renewed by cell division and new specialised cells mature to replace the cells lost. These processes are normally tightly controlled so that the gut lining is maintained in balance. In colorectal cancer, however, this balance goes awry and too much cell division creates a mass of cells that becomes a tumour. In this study by Markus Diefenbacher, our group has shown that an enzyme called Usp28 helps to promote the division of new cells in the healthy gut. However, Usp28 is often present at higher levels in colorectal tumours where it stabilises several proteins known to promote cancer. When the gene for Usp28 is deleted, mice genetically predisposed to develop intestinal cancer get fewer tumours. Even in mice with established tumours, deleting the gene for Usp28 reduces cell division, and so, tumours grow more slowly and the animals live longer. This study suggests that developing drugs to inhibit Usp28 could be a promising strategy for future colorectal cancer therapy (Figure 2). Origin licensing requires ATP binding and hydrolysis by the MCM replicative helicase. Coster G, Frigola J, Beuron F, Morris EP, Diffley JFX. Molecular Cell. 2014; 55(5):666-677 Loading of the six related Minichromosome Maintenance (MCM) proteins as head-to-head double hexamers at replication origins is crucial for ensuring once-per-cell-cycle DNA replication in eukaryotic cells. This reaction requires the Origin Recognition Complex (ORC), Cdc6 and Cdt1. ORC, Cdc6 and MCM are members of the AAA+ family of ATPases, and MCM loading requires ATP hydrolysis; but it was unclear so far which proteins need to hydrolyse ATP. In this paper we showed that ORC and Cdc6 mutants defective in ATP hydrolysis were still competent for MCM loading. However, ATP hydrolysis by Cdc6 was required for ‘proofreading’ non-productive licensing intermediates. We showed that ATP binding stabilises the wild type MCM hexamer. Moreover, by analysing MCM containing mutant subunits, we showed that ATP binding and hydrolysis by MCM are required for Cdt1 release and double hexamer formation. This work fundamentally changed our view of how ATP hydrolysis promotes this key reaction. Prereplicative complexes assembled in vitro support origin-dependent and independent DNA replication. On KF, Beuron F, Frith D, Snijders AP, Morris EP, Diffley JFX. EMBO Journal. 2014; 33(6):605-620 Eukaryotic DNA replication initiates from multiple replication origins. To ensure each origin fires just once per cell cycle, initiation is divided into two biochemically discrete steps: the Minichromosome Maintenance (MCM) helicase is first loaded as an inactive double hexamer by the origin recognition complex (ORC), Cdt1 and Cdc6; the helicase is then activated by a set of firing factors. We had previously reconstituted this first reaction with purified proteins (Remus et al., Cell. 2009; 139(4): 719-730). In the current paper we showed that plasmids containing MCM loaded with purified proteins supported complete and semi-conservative replication in extracts from budding yeast cells overexpressing firing factors. Replication requires cyclin-dependent kinase (CDK) and Dbf4- dependent kinase (DDK). DDK phosphorylation of MCM did not by itself promote separation of the double hexamer but was required for the recruitment of firing factors and replisome components in the extract. Plasmid replication did not require a functional replication origin; however, in the presence of competitor DNA and limiting ORC concentrations, replication became origin-dependent in this system. These experiments showed that MCM double hexamers are precursors of replication and provided insight into the nature of eukaryotic DNA replication origins. A Ctf4 trimer couples the CMG helicase to DNA polymerase α in the eukaryotic replisome. Simon AC, Zhou JC, Perera RL, van Deursen F, Evrin C, Ivanova ME, Kilkenny ML, Renault L, Kjaer S, Matak-Vinković D, Labib K, *Costa A and *Pellegrini L. Nature. 2014; 510(7504):293-297 *Joint corresponding authors Genome duplication requires tight coordination between DNA unwinding and synthesis within the RESEARCH HIGHLIGHTS 55 for multivalent interactions, illustrating a mechanism for the concomitant recruitment of proteins that act together at the replication fork. Figure 3 The Ctf4 trimer can bind up to three client proteins concomitantly. These findings establish the architectural framework for further mechanistic studies of the elongation step of DNA replication in eukaryotic cells (Figure 3). replisome to prevent the accumulation of vulnerable single-stranded DNA segments and the onset of genomic instability. In this study, single-particle electron microscopy and crystallography were used to establish the architecture of the Ctf4 ‘helicase-polymerase bridging factor’ either alone or bound to components of the Cdc45-MCM-GINS (CMG) helicase and the DNA Polymerase α/primase assemblies. Ctf4 was found to form a disc-shaped trimer, suggesting that it has the ability to link multiple factors at replication forks. Indeed, the Ctf4 trimer contains three docking sites that can simultaneously bind to the GINS component of the CMG helicase and Pol α. The helicase and polymerase share a common mechanism of interaction with Ctf4, with the N-terminal tails of the catalytic subunit of Pol α and the Sld5 subunit of GINS containing a common element that docks onto the C-terminal extension of a Ctf4 protomer within the trimer. Therefore, Ctf4 acts as a platform Erlotinib + MEKi Final 4 weeks of treatment Before After MEKi Erlotinib Figure 4 Lung tumours in mice expressing activated EGFR. Tumours respond to combination therapy with EGFR inhibitor plus MEK inhibitor even after they have become resistant to EGFR inhibitor alone due to NF1 down-regulation. 56 Scientific report 2014 LONDON RESEARCH INSTITUTE Reduced NF1 expression confers resistance to EGFR inhibition in lung cancer. de Bruin EC, Cowell C, Warne PH, Jiang M, Saunders RE, Melnick MA, Gettinger S, Walther Z, Wurtz A, Heynen GJ, Heideman DA, Gómez-Román J, García-Castaño A, Gong Y, Ladanyi M, Varmus H, Bernards R, Smit EF, Politi K, Downward J. Cancer Discovery. 2014; 4(5):606-619 Activating mutations in the EGF receptor (EGFR) are found in about 15% of lung cancers and are associated with clinical responsiveness to EGFR tyrosine kinase inhibitory drugs (TKIs), such as erlotinib and gefitinib. However, resistance to these drugs eventually arises, often due to a second mutation in EGFR, that prevents the drugs from binding. Through a functional genomic screen in a human lung cancer cell line and analysis of gene expression of mutant EGFR-driven lung cancers in mice, we found that erlotinib resistance was associated with reduced expression of NF1, a tumour suppressor that negatively regulates RAS proteins, which are key drivers of malignant growth. Treatment of NF1-deficient lung cancers with a MEK inhibitory drug, which targets the RAS pathway, restored sensitivity to erlotinib. Low levels of NF1 expression were associated with resistance of lung cancers to EGFR TKIs in patients. These findings identify a subgroup of patients with EGFR-mutant lung cancer who might benefit from combination therapy with EGFR and MEK inhibitors (Figure 4). RAS interaction with PI3K p110α is required for tumor-induced angiogenesis. Murillo MM, Zelenay S, Nye E, Castellano E, Lassailly F, Stamp G, Downward J. Journal of Clinical Investigations. 2014; 124(8):3601-3611 Direct interaction of RAS proteins with the lipid kinase PI3K p110α mediates RAS-driven tumour development. However, it is not clear how p110α/ RAS-dependent signalling mediates interactions between tumours and host tissues. We show here that disruption of the interaction between RAS and p110α within host tissue reduced tumour growth and tumour-induced angiogenesis, even when this interaction was intact in the tumour. Functional interaction of RAS with p110α in host tissue was required for efficient establishment and growth of metastatic tumours and also for efficient angiogenesis. Additionally, disruption of the RAS and p110α interaction altered the nature of tumour-associated immune cells such as macrophages, inducing expression of markers typical for macrophage populations with reduced tumour-promoting capacity. These results indicate that a functional RAS interaction with PI3K p110α in host tissue is required for the establishment of a growth-permissive environment for the tumour, particularly for tumour-induced angiogenesis. Targeting the interaction of RAS with PI3K has the potential to impair tumour formation by altering the tumour-host relationship, in addition to previously described tumour cell-autonomous effects. The role of differential VE-cadherin dynamics in cell rearrangement during angiogenesis. Bentley K, Franco CA, Philippides A, Blanco R, Dierkes M, Gebala V, Stanchi F, Jones M, Aspalter IM, Cagna G, Weström S, Claesson-Welsh L, Vestweber D, Gerhardt H. Nature Cell Biology. 2014;16(4):309-321 During blood vessel formation, certain molecules and cues tell endothelial cells to take on different characteristics, with some becoming ‘polarised tip cells’ and other becoming ‘stalk cells’. Together, these two cell types form new capillaries. Tip and stalk cells were previously thought to have established roles, with tip cells leading the way and stalk cells following along to create a vessel tube, but recent research has suggested that endothelial cells are much more spontaneous and actually undergo dynamic changes and frequently switch positions, with stalk cells overtaking tip cells at the leading edge of a new blood vessel. This work proposes that when endothelial cells are stimulated by vascular endothelial growth factor (VEGF) – and not inhibited by Notch signalling – they are ‘active’ and can either form a new branch as tip cells or ‘shuffle up’ through the existing sprout by cell rearrangement mechanisms and it validates simulation predictions in vivo, demonstrating that Notch regulates shuffling movement via VE-cadherin adhesion. The team also found that during cancer development and progression, there is a switch: the regulation of the adhesion between cells becomes more uniform so that there are clustered regions of cells in an all-active or all-inhibited shuffling state. When simulant cells are let loose in a simple simulated tumor environment, their collective behavior changed dramatically. The cells go through cyclic phases of adhesion and junctional movements now as a group, in which they all clamber to move at once or all remain still. In either case they are getting nowhere as overtaking requires differential movement of one cell compared to its neighbors. The disrupted rearrangement provides a new explanation for the enlarged blood vessels we see in pathologies, such as in mouse models of tumors or retinopathies. Oncogenic RET kinase domain mutations perturb the autophosphorylation trajectory by enhancing substrate presentation in trans. Plaza-Menacho I, Barnouin K, Goodman K, Martínez-Torres RJ, Borg A, Murray-Rust J, Mouilleron S, Knowles P, McDonald NQ. Molecular Cell. 2014; 53(5):738-51 Receptor tyrosine kinases (RTK) are a class of trans-membrane proteins that are frequently targeted for oncogenic activation, whether by site-specific mutation, overexpression or through fusion/translocations. Ligand-dependent RTK activation arises through trans phosphorylation (autophosphorylation) through allosteric or oligomerisation mechanisms. Oncogenic activation often arises from a dramatically enhanced tyrosine kinase activity and subversion of intrinsic RTK control mechanisms. In many cases the increased tyrosine kinase activity is a consequence of autophosphorylation of the RTK ‘activation-loop’. However, some RTKs such as the EGFR and RET are not stimulated by activationloop phosphorylation suggesting mechanisms distinct from most RTKs may operate. By monitoring the appearance and timing of autophosphorylation sites upon RET activation we found that rapid phosphotyrosine sites are all outside of the tyrosine kinase domain, whereas sites within the kinase domain appear at much later time points, consistent with a role in RET signalling rather than activation. We then explored the basis for oncogenic activation of RET by point mutations found in patients with multiple endocrine neoplasia type 2. Using biochemical, mass spectroscopy and crystallography, we show how such mutations alter the RET autophosphorylation trajectory and produce a greatly enhanced autophosphorylation substrate, despite reducing protein stability. Our study reveals an underappreciated role for oncogenic RTK mutations in promoting intermolecular autophosphorylation through an enhanced substrate presentation. RESEARCH HIGHLIGHTS 57 Replication origin selection regulates the distribution of meiotic recombination. Wu PY, Nurse P. Molecular Cell. 2014; 53(4):655-662 The pattern of origin firing during DNA replication is altered during development and also in diseases such as cancer, suggesting that it may play a role in regulating biological processes. To investigate this, we have used fission yeast meiosis as a model for a developmental pathway and examined how the meiotic process is altered under different patterns of origin firing during pre-meiotic DNA replication. We found that changing the nutritional conditions lead to changes in the pattern of origin firing. These changes had no overall effect on progression of meiosis, suggesting that a particular pattern of origin firing was not required for meiotic progression. However, changes in the origin firing pattern was correlated with local changes in the binding of the recombination factor Rad51 and meiotic recombination frequencies, with increases in origin efficiency being associated with increased binding of Rad51 and increased recombination levels. Changes in the efficiency of origin firing and Rad51 binding could be induced by directly modulating the level of the replication factor Cdc45, independently of the nutritional status. Here we show that external factors such nutritional conditions can influence the pattern of origin firing in a cell and that this may lead directly to changes in the pattern of Rad51 binding and meiotic recombination. Mitotic catenation is monitored and resolved by a PKCε- regulated pathway. Brownlow N, Pike T, Zicha D, Collinson L, Parker PJ. Nature Communications. 2014; 5:5685 The metaphase to anaphase transition is a pivotal point in cell division, where for the first time, the newly replicated, paired sister chromatids become physically separated. Mistakes resulting in Figure 5 A DLD-1 cell with knockdown of PKCε displaying a PICH positive ultrafine bridge (green) stretched out between separating sister chromatids (blue) in anaphase. These PICH positive structures are more prevalent with the increased metaphase catenation triggered by the loss or inhibition of PKCε. 58 Scientific report 2014 LONDON RESEARCH INSTITUTE chromosome non-disjunction are broadly implicated in cancer and widely associated with a poor prognosis. Using model systems, we were able to show that a metaphase to anaphase transition delay is triggered by the presence of sister chromatid catenation in metaphase. This catenation-induced delay is dependent on the protein kinase PKCε. Under conditions of excessive catenation, loss or acute inhibition of PKCε leads to precocious mitotic exit and extensive cytokinesis failure. To determine how this delay is effected, Nicola Brownlow monitored components of the spindle assembly checkpoint (SAC) and found that associated with this delay there is a PKCεdependent retention of Bub1 and BubR1 at kinetochores but no retention of Mad2, indicative of a partially silenced SAC. The engagement of this metaphase pathway was found only in certain transformed cells where the normal catenation induced G2 checkpoint was defective. This G2 checkpoint is independent of PKCε and is lost in various human tumours and tumour cell models, leading to a tumour-specific dependence on the mitotic pathway. Dissection of this mitotic catenation monitoring system therefore presents a potential opportunity for selective intervention in cancer, and also has implications for our understanding of chromosome instability in cancer (Figure 5). Antiviral immunity via RIG-I-mediated recognition of RNA bearing 5’-diphosphates. Goubau D, Schlee M, Deddouche S, Pruijssers AJ, Zillinger T, Goldeck M, Schuberth C, Van der Veen AG, Fujimura T, Rehwinkel J, Iskarpatyoti JA, Barchet W, Ludwig J, Dermody TS, Hartmann G, Reis e Sousa C. Nature. 2014; 514(7522):372-375 RIG-I is a very important innate immune sensor of RNA viruses and can be activated by RNAs bearing 5’ tri-phosphate moieties such as found in the genomes of influenza virus, measles virus, mumps virus and many other human viral pathogens. However, a few publications suggested that RNAs lacking 5’ tri-phosphates can, in some instances, act as RIG-I agonists and the crystal structures of RIG-I show that the terminal gamma phosphate in 5’ ppp RNA does not make appreciable contact with the protein. In this study, we found that RIG-I can, in fact, respond equally well to 5’ diphosphate bearing RNA such as found in the genome of reovirus and other viruses like the human rotaviruses. Furthermore, the same provides a conceptual understanding of a major protein interaction network at microtubule ends, which is important in order to understand the regulation of microtubule dynamics and cargo transport initiation in health and disease (Figure 6). Figure 6 Fluorescence microscopy images (top right) and a kymograph (bottom right) showing recruitment of the dynactin component p150 (green) to the growing end of a dynamic microtubule (red) by EB1. p150 is a key component in the +TIP network that targets dynein to microtubule ends (schematic, right). RECQL5 controls transcript elongation and suppresses genome instability associated with transcription stress. Saponaro M, Kantidakis T, Mitter R, Kelly GP, Heron M, Williams H, Söding J, Stewart A, Svejstrup JQ. Cell. 2014; 157(5):10371049 di-phosphate moieties are present in poly I:C, a synthetic stimulus that has been used for many decades to induce interferons. These data indicate that the minimal determinant of RIG-I recognition is in fact a 5’ di-phosphate-bearing blunt-ended base-paired RNA. Thus, innate self/non-self recognition extends to the detection of 5’ diphosphate-containing RNAs and RIG-I must henceforth be seen as a sensor of both 5’ di- and tri- phosphate RNAs. Reconstitution of a hierarchical +TIP interaction network controlling microtubule end tracking of the human dynein complex. Duellberg C, Trokter M, Jha R, Sen I, Steinmetz MO, Surrey T. Nature Cell Biology. 2014; 16(8):804-811 Microtubules are filaments of the cytoskeleton that constantly switch between phases of growth and shrinkage, called dynamic instability. This property is essential for many cellular processes such as mitosis, migration and intracellular transport. Microtubule ends are of particular interest because regulation of dynamic instability by proteins takes place directly at these ends. A second important feature is that cargo transport towards the cell centre is initiated directly at these ends. Many proteins that are enriched at microtubule end regions are known, but the mechanism of how these so-called +TIPs get recruited to these ends is not well understood. Many diseases are associated with mislocalised +TIPs. Using a bottom up approach, we have rebuilt the recruitment of an important motor protein, called dynein, which facilitates almost all cell centre-directed cargo transport. We could reveal how a hierarchically structured protein interaction network recruits dynein in the presence of competition for microtubule end binding. Our study This study provides evidence that transcription stress (RNAPII stopping and stalling in the transcribed region of genes) can cause dramatic genome instability, and that the tumoursuppressor RECQL5 is important to suppress it. Interestingly, transcription stress-induced genome instability is particularly frequent in regions that are devoid of ‘back-up’ DNA replication origins, causing chromosomal breakage and unscheduled/ detrimental DNA recombination in so-called common fragile sites. Hence, when replication stress meets transcription stress, chromosomes break and become rearranged. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. de Bruin E, McGranahan N, Mitter R, Salm M, Wedge DC, Yates L, Jamal-Hanjani M, Shafi S, Murugaesu N, Rowan AJ, Gronroos E, Muhammad MA, Horswell S, Gerlinger M, Varela I, Jones D, Marshall J, Voet T, Van Loo P, Rassl DM, Rintoul RC, Janes SM, Lee S, Forster M, Ahmed T, Lawrence D, Falzon M, Capitanio A, Harkins TT, Lee CC, Tom W, Teefe E, Chen SC, Begum S, Rabinowitz A, Phillimore B, Spencer-Dene B, Stamp G, Szallasi Z, Matthews N, Stewart A, Campbell P, Swanton C. Science. 2014; 346(6206):251-256 Lung cancer affects over 1.4 million people worldwide per year and is associated with an average survival of 18 months in advanced disease. To gain more insights into this disease, we performed detailed genome sequencing analyses of seven lung tumours. By comparing the DNA from different regions within each tumour, we found that about one third of the DNA errors could be found in only a subset of regions and not throughout the whole tumour. Investigating the mutations in DNA in more detail, we found the typical smoking associated ‘C-to-A’ mutations in tumours of patients that had smoked, particularly for mutations that had occurred early in tumour RESEARCH HIGHLIGHTS 59 Figure 7 Multiregion DNA sequencing of a tumour allows for the analysis of genetic intra-tumour heterogeneity and provides insight into the tumour’s life history. Mutations that are present in only one or a subset of regions, heterogeneous mutations, occurred at later stages of tumour development compared to mutations that are present in all regions. In lung tumours, ‘early’ mutations are mainly caused by smoking, and ‘late’ mutations by APOBEC enzymes. development. We noticed different types of mutations occurred after tumour initiation, linked to APOBEC DNA-editing enzymes encoded by the human genome. Normally, these enzymes are induced by cells in response to viral infection in order to damage viral DNA and limit their infectious capacity. This work demonstrates a role for this enzyme family in the initiation of mutations later in tumour evolution that drive the expansion of distinct cell populations within tumours, resulting in a diversity of cancer genomes within one patient that might have an impact on clinical outcome and drug resistance (Figure 7). Tolerance of whole-genome doubling propagates chromosomal instability and accelerates cancer genome evolution. Dewhurst SM, McGranahan N, Burrell RA, Rowan AJ, Grönroos E, Endesfelder D, Joshi T, Mouradov D, Gibbs P, Ward RL, Hawkins NJ, Szallasi Z, Sieber OM, Swanton C. Cancer Discovery. 2014; 4(2):175-185 In this study we examined the impact of genome doubling (a complete doubling of the genetic content of a cell) in cancer evolution, a phenomenon that occurs in up to 60% of cancers. Using cancer cells cultured in the laboratory and tumour data from over 500 cancer patients, we found that genome doubling can be an important event in tumour progression, likely driving disordered chromosome distribution between daughter cells (chromosomal instability), which can contribute to cancer drug resistance and poor patient outcome. We developed a novel system of diploid and genome doubled (tetraploid) colorectal cancer cell lines that are genetically identical apart from the amount of DNA within each cell. Growing these cells in parallel for over two years in the laboratory 60 Scientific report 2014 LONDON RESEARCH INSTITUTE allowed us to investigate how they evolved over time. We found that genome doubled cells could tolerate chromosomal changes better than diploid cells, and therefore evolved much more rapidly over time. We reasoned that since genome doubling occurred before genomic instability in our cell lines, genome doubling might be a useful clinical measure to identify high-risk patient groups at an early disease stage. We used two independent data sets comprising 539 patients with colorectal cancer, and found that genome doubling was predictive of poor relapse free survival. Genome doubling has relevance as a new means to predict outcome in colorectal cancer, potentially allowing the escalation of therapy in higher risk groups (Figure 8). WIPI2 links LC3-conjungation with PI3P, autophagosome formation, and pathogen clearance by recruiting Atg12-5-16L1. Dooley HC, Razi M, Polson HEJ, Girardin SE, Wilson MI, Tooze SA. Molecular Cell. 2014; 55(2):238-252 Autophagy (self-eating) is a process that cells use to remain healthy and survive in stressful conditions, in particular nutrient deprivation. However, autophagy can also be exploited by cancer cells to survive, therefore, we aim to understand the details of how autophagy can be controlled in human cells. Autophagy is performed by specialised membranes, called autophagosomes, which can be made on-demand in response to a stress signal. A set of proteins, so called Atg proteins, and an important lipid, phosphatidylinositol-3-phosphate (PI3P), are required for the formation of the autophagosome. The PI3P involved in autophagy is a unique pool present on the ER. LC3, a protein modified by another lipid, phosphatidylethanolamine, during autophagy is required for autophagosome membrane growth and closure. However, the specific activation of either MRTFs or TCFs can reset the circadian clock. Many MRTF targets are implicated in cancer invasiveness, metastasis, and mechanosensing, suggesting that MRTF-SRF signalling constitutes a nuclear arm of the cellular response to matrix adhesion. Figure 8 Clonal FISH (fluorescence in-situ hybridisation) of a tetraploid colorectal cancer clone derived from HCT-116. Chromosome 2 (red) and chromosome 8 (green) show copy number variation between cells, indicating chromosomal instability. Nuclei are stained with DAPI (blue). Biochemical reconstitution of topological DNA binding by the cohesin ring. Murayama Y, Uhlmann F. Nature. 2014; 505(7483):367-371 connection between Atg proteins, LC3 and PI3P, was not known. Our publication describes the discovery of a direct interaction between the Atg protein WIPI2b, which binds PI3P and Atg16L1, a component of the LC3-lipidation complex. This discovery provides the missing link between ER-localised production of PI3P, which is triggered by autophagy, and recruitment of the LC3conjugation complex crucial for autophagosome membrane formation. Furthermore, this enables us to manipulate the process at the molecular level to further our understanding of how autophagosomes form and how autophagy can be regulated. Rho-actin signaling to the MRTF coactivators dominates the immediate transcriptional response to serum in fibroblasts. Esnault C, Stewart A, Gualdrini F, East P, Horswell S, Matthews N, Treisman R. Genes and Development. 2014; 28(9):943-958 Gene transcription is a prerequisite for quiescent cells to enter the cell cycle. Classical experiments identified the SRF (‘Serum Response Factor’) transcription factor as master regulator of ‘immediate-early’ genes that are induced within minutes by mitogenic stimuli. SRF is regulated by two families of signal-regulated cofactors. The TCFs link SRF activity to the classical mitogenic Ras-ERK signal pathway to control genes such as c-fos, while the MRTFs couple SRF activity to Rho signalling, responding to signal-induced changes in cellular concentration of G-actin. This paper evaluates the extent to which the SRF network controls the immediate-early transcriptional response, using genomic methods and pathwayspecific inhibitors. Several hundred direct targets for MRTF-SRF and TCF-SRF signalling were identified. MRTF targets encode regulators of the cytoskeleton, transcription, and cell growth, and Cohesion between replicated sister chromatids is mediated by the chromosomal cohesin complex and is a prerequisite for faithful chromosome segregation in mitosis. Cohesin plays vital roles also in DNA repair and transcriptional regulation. The ring-shaped cohesin complex is thought to encircle sister DNA strands, but its molecular mechanism of action is poorly understood and the biochemical reconstitution of cohesin activity in vitro had remained an unattained goal. We were now successful in reconstituting topological cohesin loading onto DNA using purified fission yeast cohesin and its loader complex, Scc2Mis4/Scc4Ssl3. Surprisingly, we found that incubation of cohesin with DNA leads to spontaneous topological loading, but that remains inefficient. Interaction site mapping revealed that the loader makes contact with cohesin at multiple sites around the ring circumference, including the hitherto enigmatic Scc3Psc3 subunit. The loader furthermore stimulates cohesin’s ATPase. This leads us to a first molecular model of the cohesin loading reaction, in which the cohesin loader acts as a template for a conformational change in the cohesin complex that triggers ATP hydrolysis-dependent loading. This provides mechanistic insight into the initial steps of establishing sister chromatid cohesion and other chromosomal processes mediated by cohesin. The Scc2-Scc4 complex acts in sister chromatid cohesion and transcriptional regulation by maintaining nucleosome-free regions. Lopez-Serra L, Kelly G, Patel H, Stewart A, Uhlmann F. Nature Genetics. 2014; 46(10):11471151 The cohesin complex lies at the heart of many chromosomal activities, including sister chromatid cohesion and transcriptional regulation. Loading of cohesin onto chromosomes depends on the Scc2/ Scc4 cohesin loader complex, but the chromatin features that attract Scc2/Scc4 and thereby form cohesin loading sites remained poorly understood. In this work, we showed that the budding yeast Scc2/Scc4 complex is recruited to broad RESEARCH HIGHLIGHTS 61 nucleosome-free regions in the promoters of highly expressed genes by the RSC chromatin remodelling complex. Unexpectedly, Scc2/Scc4 itself is required to maintain the nucleosome-free status of its binding sites. Consequently, inactivation of the cohesin loader or RSC complex has similar effects on nucleosome positioning, gene expression and sister chromatid cohesion. These results revealed an intimate link between local chromatin structure under control of the RSC chromatin remodelling complex and higher order chromosome architecture rendered by cohesin. These findings also pertain to the similarities between two severe human developmental disorders, Cornelia de Lange syndrome, most often caused by mutations in the human Scc2NIPBL cohesin loader subunit, and Coffin-Siris syndrome, resulting from mutations in human RSC complex components. We suggest that both could arise from gene misregulation due to overlapping changes in the nucleosome landscape (Figure 9). Ena/VASP proteins cooperate with the WAVE complex to regulate the actin cytoskeleton. Chen XJ, Squarr JA, Stephan R, Chen B, Higgins TE, Barry DJ, Martin MC, Rosen MK, Bogdan S, Way M. Developmental Cell. 2014; 30(5):569-584 Directed cell motility in response to a wide variety of signalling events requires temporal and spatial control of actin polymerisation at the plasma membrane. Ena/VASP proteins and the WAVE regulatory complex (WRC) have emerged as important regulators of cell motility by virtue of their ability to independently promote actin polymerisation at the leading edge of migrating cells. We have now demonstrated that the N-terminal EVH1 domain of Ena/VASP proteins Figure 9 Colocalisation of the budding yeast cohesin loader subunit Scc2 with the RSC chromatin remodelling complex subunit Sth1. The Sth1 binding profiles, centred at each Scc2 binding site are collated to visualise the close correlation. Further work showed how the cohesin loader cooperates with RSC to maintain nucleosome free regions, regulate transcription from the bound promoters and load cohesin onto DNA. 62 Scientific report 2014 LONDON RESEARCH INSTITUTE interacts with an extended proline rich motif in the Abi subunit of the WRC. In vitro, VASP cooperatively enhances the ability of the WRC to stimulate Arp2/3 complex-induced actin assembly in the presence of Rac1. This increased activity depends on VASP tetramerisation and its interaction with Abi1. In Drosophila, Ena also interacts with the WRC via two LPPPP motifs in Abi. Loss of this interaction in Drosophila hemocytes results in defects in membrane protrusion and cell spreading, as well as redistribution of Ena to the tips of filopodia-like extensions. Rescue experiments in an abi mutant background also reveal a physiological requirement for the Abi:Ena interaction in photoreceptor axon targeting and oogenesis in vivo. Our data have demonstrated that the activity of Ena/VASP and the WRC are intimately linked to ensure optimal control of actin polymerisation. Dual control of Yen1 nuclease activity and cellular localization by Cdk and Cdc14 prevents genome instability. Blanco MG, Matos J, West SC. Molecular Cell. 2014; 54(1):94-106 The careful orchestration of cellular events such as DNA replication, repair and chromosome segregation is essential for the equal distribution of the duplicated genome to the two daughter cells. To ensure that persistent recombination intermediates are resolved prior to cell division, the enzymes that resolve junctions need to be activated at the appropriate time in the cell cycle. This paper defines the mechanism by which the Yen1 Holliday junction resolvase is regulated in yeast. We found that Yen1 undergoes a dual mode of regulation, by modulation of both its activity and subcellular localisation. Cdk phosphorylation Scc2- inhibits Yen1 at S phase by reducing its DNA binding affinity, whereas it is activated at anaphase by the Cdc14 phosphatase. We find that proper regulation is critical for chromosome segregation. Spatial control of the GEN1 Holliday junction resolvase ensures genome stability. Chan YW and West SC. Nature Communications. 2014; 5:4844 Holliday junction (HJ) resolvases are necessary for the processing of persistent recombination intermediates before cell division. Their actions, however, need to be restricted to the late stages of the cell cycle to avoid the inappropriate cleavage of replication intermediates. Control of the yeast HJ resolvase Yen1 involves phosphorylation changes that modulate its catalytic activity and nuclear import. Here, we show that GEN1, the human ortholog of Yen1, is regulated by a different mechanism that is independent of phosphorylation. GEN1 is controlled exclusively by nuclear exclusion, driven by a nuclear export signal (NES) that restricts GEN1 actions to mitosis when the nuclear membrane breaks down. Construction of a nuclear-localised version of GEN1 revealed that its premature actions partially suppress phenotypes associated with loss of BLM and MUS81 but cause elevated crossover formation. The spatial control of GEN1 therefore contributes to genome stability by avoiding competition with non-crossover promoting repair pathways. Roles of SLX1-SLX4, MUS81-EME1 and GEN1 in avoiding genome instability and mitotic catastrophe. Sarbanjna S, Davies D, West SC. Genes and Development. 2014; 28(10):1124-1136 The resolution of recombination intermediates containing Holliday junctions (HJs) is critical for genome maintenance and proper chromosome segregation. Three pathways for HJ processing exist in human cells and involve the following enzymes/complexes: BLM–TopoIIIa–RMI1–RMI2 (BTR complex), SLX1–SLX4–MUS81–EME1 (SLX– MUS complex), and GEN1. Cycling cells preferentially use the BTR complex for the removal of double HJs in S phase, with SLX–MUS and GEN1 acting at temporally distinct phases of the cell cycle. Cells lacking SLX–MUS and GEN1 exhibit chromosome missegregation, micronucleus formation, and elevated levels of 53BP1-positive G1 nuclear bodies, suggesting that defects in chromosome segregation lead to the transmission of extensive DNA damage to daughter cells. In addition, however, we found that the effects of SLX4, MUS81, and GEN1 depletion extend beyond mitosis, since genome instability is observed throughout all phases of the cell cycle. This is exemplified in the form of impaired replication fork movement and S phase progression, endogenous checkpoint activation, chromosome segmentation, and multinucleation. In contrast to SLX4, SLX1 (the nuclease subunit of the SLX1–SLX4 structure-selective nuclease) plays no role in the replication-related phenotypes associated with SLX4/MUS81 and GEN1 depletion. These observations demonstrate that the SLX1– SLX4 nuclease and the SLX4 scaffold play divergent roles in the maintenance of genome integrity in human cells. Structure and mechanism of action of the BRCA2 breast cancer tumour suppressor. Shaid T, Soroka J, Kong E, Malivert L, McIlwraith MJ, Pape T, *West SC and *Zhang X. Nature Structural Molecular Biology. 2014; 21:962-968 *Joint corresponding authors Mutations in BRCA2 increase susceptibility to breast, ovarian and prostate cancers. The product of the BRCA2 gene, BRCA2 protein, has a key role in the repair of DNA double-strand breaks and interstrand crosslinks by RAD51-mediated homologous recombination. In this study, we present a biochemical and structural characterisation of full-length (3,418 amino acid) BRCA2, alone and in complex with RAD51. We found that BRCA2 facilitates nucleation of RAD51 filaments at multiple sites on single-stranded DNA. Three-dimensional electron microscopic reconstructions revealed that BRCA2 exists as a dimer and that two oppositely oriented sets of RAD51 molecules bind the dimer. Single-stranded DNA binds along the long axis of BRCA2, such that only one set of RAD51 monomers can form a productive complex with DNA and establish filament formation. Our data define the molecular mechanism by which this tumour suppressor facilitates RAD51-mediated homologousrecombinational repair. RESEARCH HIGHLIGHTS 63 Drosophila wing imaginal discs expressing DIAP1-GFP (green) and ubi-RFP (red) and stained with DAPI (blue). Image: Maxine Holder, Apoptosis and Proliferation Control Group. 64 Scientific report 2014 LONDON RESEARCH INSTITUTE LINCOLN'S INN FIELDS The London Research Institute, Lincoln’s Inn Fields laboratories are located in the centre of London. The research laboratories work within the broad research themes of cellular regulatory mechanisms, biology of tumours and tissues, and genomic integrity and cell cycle. By carrying out basic research at the Institute we will continue to increase the understanding of cancer biology. The researchers are supported by an excellent range of Technology Core Facilities. LINCOLN'S INN FIELDS 65 LYMPHOCYTE INTERACTION www.london-research-institute.org.uk/research/facundo-batista Group Leader Facundo D Batista Postdoctoral Scientists Shweta Aggarwal Bruno Frederico Francesca Gasparrini Selina Keppler Nuria Martínez-Martín Graduate Students Marianne Burbage Mauro Gaya Carlson Tsui Scientific Officers Julia Coleman Cecilia Deantonio Christoph Feest David Mestre Beatriz Montaner Paul Newman Microscope Developer Andreas Bruckbauer B lymphocytes are important immune cells. They produce antigenspecific antibodies by which they can protect us against disease. In the Lymphocyte Interaction Laboratory, we investigate the cellular and molecular events leading to the activation of B cells and their differentiation into antibody producing cells. We use a variety of techniques based on genetics, biochemistry and state-of-the-art imaging technology, both in vitro and in vivo. As examples, by tracking single B cell receptor (BCR) particles we have previously shown that diffusion of the BCR is restricted by an ezrin-defined actin network (Treanor et al., 2010; Immunity. 32(2): 187-199), and that this restriction regulates receptor signalling; or by following B cells in vivo we can understand where and when B cell activation occurs (Carrasco and Batista, 2007; Immunity. 27(1): 160-171). We have since explored novel concepts of BCR signalling regulation and its involvement in B cell activation, as well as delving deeper into our studies of how and when B cells become activated in vivo. The behaviour of B cells on both molecular and cellular levels can aid our understanding of immune responses to pathogens and disease control We know that B cell activation is triggered by specific recognition of antigen through the BCR and that both positive and negative regulatory molecules modulate this process. We have been dissecting the detail of BCR regulation, and, in particular, the role of the organisation and dynamics of these regulators within the cell membrane. Additionally, we are looking at the behaviour and interactions of B cells with innate immune effector cells within secondary lymphoid organs. We have directed our focus in this respect to the behaviour of subcapsular sinus (SCS) macrophages during natural infections and the resulting implications on B cell responses to secondary pathogens. We have, therefore, been working towards a clearer understanding of B cell activation, antibody production and the dynamics of lymph node organisation following infections; knowledge that will aid us not only in the fight against cancer but also infectious diseases. Cdc42 organisation and dynamics B cells recognise foreign antigens by virtue of cell surface immunoglobulin receptors and are most effectively activated by membrane-bound ligands. Previously, work in the group has shown that the early stages of this process involve a two-phase 66 Scientific report 2014 LONDON RESEARCH INSTITUTE response involving spreading and contraction and that the extent of this signalling- and actindependent response determines the quantity of antigen accumulated and, hence, the degree of B cell activation (Fleire et al., 2006; Science. 312(5774): 738-741). Further investigation revealed that the cortical actin cytoskeleton was involved in the control of B cell dynamics and signalling and that the Ezrin-Radixin-Moesin (ERM) proteins are a crucial link in this control mechanism (Treanor et al., 2011; J Exp Med. 208(5): 1055-1068; Treanor et al., 2010). Delving deeper into the signalling pathway, we found a role for the adaptor protein Nck in BCR signalling, enabling the recruitment of BCAP in the essential P13K-Akt pathway (Castello et al., 2013; Nat Immunol. 14(9):966-975). Building on this work, we demonstrated that cytoskeleton disruption triggers B cell signalling not only through the BCR but involves the co-receptor CD19 and tetraspanin (CD81), suggesting that receptor compartmentalisation regulates antigen-induced activation (Mattila et al., 2013; Immunity. 38(3): 461-474). We have most recently focused on Cdc42, a small Rho GTPase that is known to be a regulator of actin remodelling, so we sought to investigate whether it was therefore also implicated in B cell activation. In mice with Cdc42 deleted in the B cell lineage only, we saw that antibody responses were eliminated and the mice were incapable of forming germinal centres or of generating plasma B cells. 3D view Top view 3D side view Control CpG Figure 1 3D multiphoton microscopy images showing that CpG-induced inflammation in mice leads to a decrease in the number and density of CD169+ macrophages (green) in the draining lymph nodes, along with a retraction and change in morphology of these cells when compared with PBS controls. The cyan shows a second harmonic signal generated by collagen fibrils. This was the case for either viral infection or for immunisation. There were profound and multiple B cell faults seen to be causing this severe immune deficiency. These included early blocks during B cell development; impaired antigen-driven BCR signalling and actin remodelling; defective antigen presentation and in vivo interaction with T cells; and a severe B cell intrinsic block in plasma cell differentiation. Our study adds a new perspective on Cdc42 function as a master regulator of B cell physiology (Burbage et al., 2014;doi:10.1084/ jem.20141143). SCS macrophage disruption following infection As well as studying lymphocyte interactions at the micro- and nano-scale, we actively pursue the understanding of how, where, and when B cells are activated in vivo. The lymph node is an important location for the presentation of antigen to B cells and we have observed that antigen accumulates in the macrophage-rich area at the SCS boundary, and identified this area as a site for the initiation of B cell responses (Carrasco and Batista, 2007). We have also shown that SCS macrophages are capable of presenting certain antigens to other immune cells, including iNKT cells (Barral et al., 2010; Nat Immunol. 11(4): 303-312). Recently, we have focused special attention on the importance of lymph node architecture with relation to SCS macrophages and the implications for antigen presentation to B cells and other immune effector cells. In particular, we studied B cell activation in a model of double infection. The layer of CD169+ SCS macrophages that captures pathogens as they enter the lymph node prevents the spread of pathogens and triggers immune responses. So far, the impact of infection on the organisation and function of SCS macrophages has been largely unexplored, so, using innovative imaging approaches, we investigated this scenario. We found that virus- and bacteria-induced inflammation led to a dramatic disruption of SCS macrophages caused in part by mature dendritic cells entering the lymph node in response to pathogen-associated signals. This change in the organisation of SCS macrophages reduces their ability to retain and present antigen in subsequent secondary infections, leading to reduced B cell responses. We believe that the SCS macrophage layer acts as a kind of safety valve during infection, which makes the lymph node temporarily unable to fully respond to further antigenic challenge. This observation provides an intriguing additional level of temporal compartmentalisation during immune responses and offers a potential mechanism for the known phenomenon of increased susceptibility to secondary infections. In addition, this has important implications for our understanding of infection and for the optimisation of vaccination protocols (Gaya et al., 2015; Science. 347(6222): 667-672). BliNK Therapeutics Ltd As ever, one of the long-term objectives of our institution is to make discoveries that can lead to new therapies. In a good example of how our research has translated to treatments, after the inception and development of a novel platform by the Lymphocyte Interactions Laboratory, BliNK therapeutics has now been successfully launched as an independent company operating in Stevenage, UK and is producing therapeutic antibodies. Publications listed on page 164 LINCOLN'S INN FIELDS 67 MAMMALIAN GENETICS www.london-research-institute.org.uk/research/axel-behrens Group Leader Axel Behrens Postdoctoral Scientists Atanu Chakraborty Markus Diefenbacher Ralph Gruber Omar Khan Cristina Pasi Fabio Pucci Eva Madi Riising Edgar-Josue Ruiz Medina Rocio Sancho Antonio Tedeschi Graduate Students Rute Ferreira Christopher Gribben Hanna Halavach Hendrik Messal Scientific Officers Catherine Cremona Clive Da Costa 68 Every organ harbours adult stem cells, which have the potential for long-term replication, together with the capacities of self-renewal and multi-lineage differentiation. These stem cells function in tissue homeostasis and contribute to regeneration in response to injury. In addition, many cancers are caused by transforming mutations occurring in tissue-specific progenitor cells. Our major focus is to elucidate the molecular mechanisms governing stem cell function and cancer. Loss of Fbw7 reprograms adult pancreatic ductal cells into α, δ, and β cells The adult pancreas is capable of limited regeneration after injury, but has no defined stem cell population. The cell types and molecular signals that govern the production of new pancreatic tissue are not well understood. Previous studies from our laboratory and others have shown that Fbw7, the substrate recognition component of an SCF-type E3 ubiquitin ligase, controls the stability of several key cell fate determinants and oncoproteins. Loss of Fbw7 alters the balance of stem and progenitor cells in tissues such as the nervous system and intestine. Surprisingly, we found that inactivation of Fbw7 in the adult pancreas induces a subset of pancreatic ductal cells to reprogram into β cells, and to a lesser extent α and δ cells. Loss of Fbw7 stabilised the transcription factor Ngn3, a key regulator of endocrine cell differentiation. Moreover, expression of a stable form of Ngn3 in pancreatic ductal cells induced a similar frequency of reprogramming to β cells. The induced β cells resemble islet β cells in morphology and histology and express a comprehensive panel of β cell markers. A frequent stumbling block in previous models inducing cell reprogramming has been the functionality of the newly formed β cells. In contrast, Fbw7-mutant induced β cells secrete comparable amounts of insulin after glucose challenge as bona fide β cells isolated from pancreatic islets, suggesting that they are functional. Thus, loss of Fbw7 appears to reawaken an endocrine developmental differentiation program in adult pancreatic ductal cells, identifying Fbw7 as a master regulator of cell fate decisions in the pancreas. Our study highlights the plasticity of seemingly differentiated adult cells and reveals adult pancreatic duct cells as a latent multipotent cell type (Sancho et al., 2014; Cell Stem Cell. 15(2):139-153) (Figure 1). Scientific report 2014 LONDON RESEARCH INSTITUTE The deubiquitinase USP28 controls intestinal homeostasis and promotes colorectal cancer Colorectal cancer is the third most common cancer worldwide. Although the transcription factor c-MYC is misregulated in the majority of colorectal tumours, it is difficult to inhibit it directly. Researchers are therefore looking for alternative ways to target c-MYC, for example by altering its stability. The deubiquitinase USP28 stabilises c-MYC as well as other oncogenic factors, but its role in tumourigenesis and in the intestine was unknown. Using murine genetic models, we determined that USP28 antagonises the ubiquitin-dependent degradation of c-MYC, as well as 2 additional oncogenic factors, c-JUN and NOTCH1, in the intestine. Mice lacking USP28 were healthy, but showed reduced intestinal proliferation and increased differentiation of secretory lineage cells. In a murine model of colorectal cancer, mice harbouring Usp28 deletion developed fewer intestinal tumours. More importantly, even in mice with established tumours, deleting Usp28 reduced tumour size and dramatically increased lifespan. USP28 deficiency promoted tumour cell differentiation accompanied by decreased proliferation, suggesting that USP28 acts similarly in intestinal homeostasis and colorectal cancer models. Moreover, we identified Usp28 as a c-MYC target gene highly expressed in murine and human intestinal cancers. USP28 and c-MYC form a positive feedback loop that maintains high c-MYC protein levels in tumours. As deubiquitinases similar to USP28 have been successfully inhibited by small molecules, inhibition of USP28’s enzymatic activity may be a promising strategy for cancer therapy (Diefenbacher et al., 2014; J Clin Invest. 124(8):3407-3418). UBR5-mediated ubiquitination of ATMIN is required for IR-induced ATM signalling and function The checkpoint kinase ATM directs the cellular response to ionising radiation (IR) by localising to DNA damage sites and actively phosphorylating proteins involved in repair and survival. ATM is recruited and activated at damage sites via an interaction with the Mre11/Rad50/NBS1 (MRN) complex. An alternative ATM binding partner, ATMIN, is not involved in the response to IR but mediates ATM kinase signalling in response to chromatin changes. The molecular mechanism favouring either MRN or ATMIN in response to specific stimuli is enigmatic. We have previously shown that ATMIN competitively inhibits ATM’s interaction with the NBS1 subunit of MRN, suggesting that there must be a mechanism preventing ATMIN from disrupting ATM signalling in IR conditions. We have now identified the E3 ubiquitin ligase UBR5 as a key component of ATM activation in response to IR. We discovered that UBR5 ubiquitinates ATMIN, which favours its dissociation from ATM, freeing ATM to interact with NBS1. This mechanism allows efficient ATM activation at damage sites and promotes cell survival after irradiation. UBR5 interacts with ATMIN and catalyses ubiquitination of ATMIN at lysine 238 in an IR-stimulated manner. We showed that UBR5 deficiency, or mutation of ATMIN lysine 238, prevents ATMIN dissociation from ATM and inhibits ATM and NBS1 foci formation after IR. This reduction in ATM signalling impairs checkpoint activation and increases radiosensitivity. Thus, UBR5-mediated ATMIN ubiquitination is a vital event for ATM pathway selection and activation in response to DNA damage (Zhang et al., 2014; Proc Natl Acad Sci U S A. 111(33):12091-12096). Publications listed on page 164 Figure 1 Fbw7 maintains pancreatic ductal cell fate. Tamoxifen-inducible inactivation of Fbw7 in adult pancreatic ductal cells (expressing CK19) results in an increase in the endocrine transcription factor Ngn3 and emergence of insulin-positive β cells within the ducts. Ngn3 is a direct target of ubiquitination by the Fbw7-containing SCFFbw7 E3 ubiquitin ligase complex, which in wild type ducts marks Ngn3 for degradation. In the Fbw7 knockout (KO) ducts, Ngn3 protein is stabilised, favouring transdifferentiation of selected ductal cells to an endocrine fate. LINCOLN'S INN FIELDS 69 HAEMATOPOIETIC STEM CELL www.london-research-institute.org.uk/research/dominique-bonnet Group Leader Dominique Bonnet Postdoctoral Scientists Ander Abarrategi Alessandro Di Tullio Katie Foster Ashley Hamilton Diana Passaro Kevin Rouault-Pierre Graduate Students Alessandra Audia Amy Bradburn Alexander Waclawiczek Scientific Officers Linda Ariza-McNaughton Erin Currie Fernando dos Anjos Afonso Jashu Patel Our group is interested in studying human normal haematopoietic stem cells (HSCs) and leukaemic stem cells (LSCs). We are at present investigating the relationship between normal HSCs, LSCs and their microenvironment. For that we have developed in vivo imaging techniques allowing us to visualise and define the HSC and LSC niche in vivo. We have also developed a 3D scaffold system where human stroma cells could be co-cultured in vivo with HSCs or LSCs to study the effect of the interaction, and the cross-talk between stroma and HSCs/LSCs. These projects should shed light into pathways or interactions that are more specifically used by LSCs and where therapeutic intervention might be developed. Maintenance of LSCs ex vivo In Acute Myloid Leukemia (AML), we know that LSCs cannot be maintained ex vivo without the addition of stromal support indicating that LSCs are dependent on their microenvironment for their survival/maintenance. By modelling key elements of the bone marrow niche using different stromal feeder layers and hypoxic culture conditions, we recently demonstrated that we can maintain LSCs over at least three weeks and support their self-renewal properties. This culture system can be used as an in vitro surrogate for xenotransplantation and has the potential to dramatically increase the throughput of the investigation about LSCs. This would further provide the means by which to identify and target the functionality of the different signalling pathways involved in the maintenance and resistance of LSCs to improve AML treatments (Griessinger et al., 2014; Stem Cell Trans Med. 3(4): 520-529). LSC niche(s) and cross-talk between LSCs and their microenvironment Understanding the crosstalk between LSCs and their microenvironment is crucial to better understand the dependency of LSCs to their microenvironment. We thus started a project trying to better define the factors involved in the crosstalk between AML and their stroma. AML samples were co-cultured ex vivo with mesenchymal stroma cells (MSCs) and after one week, microarray analysis was performed on sorted MCSs. Using pathway analysis and the Gene Go program, we built a network of the combined datasets. Further studies will be crucial to evaluate the effect of the different keys factors. If any of 70 Scientific report 2014 LONDON RESEARCH INSTITUTE these factors affect the growth, differentiation or apoptosis of AML, we will further investigate one or more pathway(s) in more detail in vitro but also in vivo using knock-down approaches. HSC and LSC niche(s) In order to better characterise the bone marrow microenvironment, stem cells and their niches, we have developed different technologies for in vivo contrasting procedures as well as for tracking normal and leukaemic cells in vivo, combining whole body near infrared fluorescence, bioluminescence imaging, intravital microscopy of intact live bone marrow as well as histology and flow cytometry. We believe that the combined use of advanced multimodal and multiscale analysis of the bone marrow will very likely contribute to shed new light on our understanding of haematopoietic stem cells and their niches in health and disease (Lassailly et al., 2010; Blood. 115(26): 5347-5354; Lassailly et al., 2013; Blood. 122(10): 1730-1740). Using this technique on live animals adding time-lapse imaging, we visualised the arrival of human HSCs in the bone marrow and their behaviour over time. This should allow us to better define the normal HSC niche (Foster et al., under revision). We are now also starting to look into how leukaemia develops in vivo and visualise its invasion (Figure 1). Efficacy of immunotherapy against LSCs using CAR-T cells As significant numbers of AML patients are still refractory to conventional therapies or experience relapses, immunotherapy using T cells expressing chimeric antigen receptors (CARs) might represent A Figure 1 Seeing leukaemia development: Visualisation of the expansion and invasion of leukaemia over time (A: day 8; B: day 15 and C: day 21) using intravital non-invasive imaging of the bone marrow cavities of the calvaria of this mouse. HL-60 AML cells transduced with GFP (green) were injected into immunodeficient mice. Red: blood vessels contrasted with a blood pool agent. B a valid treatment option. AML cells frequently overexpress the myeloid antigens CD33 and CD123, for which specific CARs can be generated. However, CD33 is also expressed on normal haematopoietic stem/progenitor cells (HSPCs), and its targeting could potentially impair normal haematopoiesis. In contrast, CD123 is widely expressed by AML, while low expression is detected in HSPCs, making it a much more attractive target. In a recent study we demonstrated the in vivo efficacy and safety of using cytokine-induced killer (CIK) T cells genetically modified to express anti-CD33 or C anti-CD123 CAR to target AML. We showed that both these modified T cells are very efficient in reducing leukaemia burden in vivo, but only the anti-CD123 CAR has limited killing in normal HSPCs, thus making it a very attractive immunotherapeutic tool for AML treatment (Pizzitola et al., 2014; Leukemia. 28(8): 1596-1605). Publications listed on page 164 LINCOLN'S INN FIELDS 71 Immunity and Cancer www.london-research-institute.org.uk/research/dinis-calado Group Leader Dinis Calado Postdoctoral Scientists Rita Barbosa Giulia Morlino Nikolay Popov Andrea Taddei Graduate Students Djamil Damry Magdalena Gabrysiak Amparo Toboso-Navasa Scientific Officer Nora-Ann McFadden Our immune system is remarkable. It is composed of a network of cells including T and B cells, which work together to protect our body from pathogens such as bacteria, viruses and parasites. During an immune response to infection, B cells undergo a T cell-dependent maturation stage called germinal centre (GC) within secondary lymphoid organs in order to become activated and produce high affinity antibodies against the invading pathogen. During this maturation stage, B cells undergo processes of DNA mutation and recombination while rapidly dividing. Infidelities in these processes may lead to oncogenic DNA lesions. Indeed, the most common types of haematological malignancies in adults such as Hodgkin and nonHodgkin lymphomas as well as multiple myeloma originate from GC or post-GC B cells emphasising the importance of the maturation events within the GC microenvironment for lymphomagenesis. Interplay between immunity and cancer development A major interest of the Immunity and Cancer group is to understand how healthy cells become cancerous. To address this question we study B cells as they provide an ideal model system: firstly, their normal development is well defined; secondly, most common types of human haematological malignancies are derived from the transformation of healthy B cells into cancer cells. Although cancer cells may arise from healthy B cells at several stages of development, lymphomas derived from GC or post-GC B cells account for the most common haematological malignancies in adults, including the vast majority of Hodgkin and non-Hodgkin lymphomas (e.g. Diffuse Large B Cell Lymphoma (DLBCL) and Burkitt Lymphoma) and multiple myeloma. Therefore, using an integrative approach to immunology and cancer biology our group aims to understand the mechanisms by which healthy B cells within the GC become cancerous. For that, we primarily perform studies on the following complementary research themes: Identification and characterisation of specific subpopulations within healthy GC or post-GC B cells with high potential of becoming cancer cells, forming the source of the so-called cancer’s ‘cells of origin’ An example of such work is the identification of c-Myc+ B cell subpopulations in immature and 72 Scientific report 2014 LONDON RESEARCH INSTITUTE mature GCs, playing indispensable roles in the formation and maintenance of GCs (Calado et al., 2012; Nat Immunol. 13(11): 1092-1100). The identification of these functionally critical cellular subsets has important implications for human B cell lymphomagenesis, given that it frequently involves MYC chromosomal translocations. As these translocations are generally dependent on transcription of the recombining partner loci, the c-Myc+ GC subpopulations may be at a particularly high risk for malignant transformation. Investigation of causative mutations responsible for the transformation of normal B cells into cancer cells High-throughput sequencing of human cancer genomes, including those of haematological malignancies, has produced large datasets. However, in human cancers the driver mutations are sometimes masked by their inclusion on large amplifications or deletions or by other so-called passenger/bystander mutations that do not contribute to the phenotype or progression of the cancer. Strategies aiming to discriminate these mutations are required to prioritise further functional validation. If truly important causative genes and their associated mechanisms are evolutionary conserved, mouse models of human cancers can be employed. Mouse versus human inter-species oncogenomic comparisons may serve as a very powerful tool to identify causal mutations and to interrogate their role in a defined genetic context. A 100 80 Percent survival 60 40 Cγ1-cre e YFP stopFL (n=27, Med. surv: und) 20 Cγ1-cre IKK2castopFL (n=16, Med. surv: und) ** Cγ1-cre Blimp1FF eYFPstopFL (n=50, Med. surv: und) Cγ1-cre Blimp1FF IKK2castopFL (n=24, Med. surv: 466 days) 0 0 B C 100 300 Time (days) 400 *** 500 Cγ1-cre Blimp1FF Cγ1-cre Blimp1FF eYFP stopFL IKK2castopFL Control H&E 200 ns Control IRF4/Mum1 Control Cγ1-cre Blimp1FFeYFP stopFL Cγ1-cre Blimp1FFeYFP stopFL Cγ1-cre Blimp1FFIKK2castopFL Cγ1-cre Blimp1FFIKK2castopFL Expressed in ABC-DLBCL Study the processes of cancer cell evasion from the immune system through analysis of the interplay between a developing B cell lymphoma and immune cells Host immunity plays a fundamental role in tumour formation and progression and cancer immune escape is an emerging ‘Hallmark of Cancer’. For that reason, the study of cancer cannot be confined to the intrinsic cellular mechanisms of oncogenic transformation, but needs to be expanded to involve interactions of (pre-) cancer cells with their microenvironment. However, most of the current mouse models do not fulfil the requirements to study these interactions as the introduction of oncogenic mutations is neither tissue-specific nor can it be temporally or spatially regulated, targeting only a small subset of cells. We are currently developing novel genetic approaches in vivo, which allow us to introduce oncogenic mutations in a small fraction of GC B cells, mimicking the sporadic nature of the cancer initiating cells. Furthermore, we employ an inducible gene modification strategy, which allows us to trace mutant cells during cancer development and progression. Non NFκB targets NFκB targets Expressed in GC DLBCL D As an example of such work, it was known that constitutive NF-κB activity and abrogation of terminal B cell differentiation through Blimp1 disruption are two recurrent genetic events in human DLBCL. In order to investigate if these two events were causal for DLBCL, an in vivo mouse model targeting specifically GC B cells was generated. Not only did the combination of these events lead to lymphomagenesis; these lymphomas also recapitulated many of the features of the most aggressive DLBCL subtype, the so-called Activated B Cell DLBCL (ABC-DLBCL), which has a poorer clinical prognosis (Calado et al., 2010; Cancer Cell. 18(6): 580-589). Thus, this work suggests that both NF-κB signalling as an oncogenic event and Blimp1 as a tumour suppressor, play causal roles in the pathogenesis of human ABCDLBCL, and illustrates well the relevance of mouse models in the understanding of cancer pathogenesis (Figure 1). Figure 1 Constitutive canonical NF-κB activation cooperates with disruption of Blimp1 in the pathogenesis of activated B cell-like diffuse large cell lymphoma. A. Kaplan-Meier survival plots of mice carrying the Cγ1-cre transgene (for GC B cell-specific Cre expression) together with a conditional Blimp1 allele (Blimp1FF) and/or a conditional R26 allele expressing a constitutively active IKK2 kinase (IKK2castopFL) leading to NF-κB activation. Mice carrying the Cγ1-cre transgene together with a conditional R26 allele expressing eYFP (eYFPstopFL) served as controls. Med. surv, median survival; und, undefined. B. Representative pictures of spleens from mutant and control mice. C. Left panel, representative H&E staining of spleens from mutant and control mice. Right panel, representative immunohistochemical staining for MUM1/IRF4 (characteristic of the ABC-DLBCL subtype but not of the GC-DLBCL subtype) of spleens from mutant and control mice. Scale bar, 1000 μm; inset, 100 μm. D. Heat map showing the relative transcript levels of candidate genes in lymphoma samples compared to normal GC B cells. Heat map represents Hprtnormalised value of each candidate gene normalised to equal a mean of 0 and variance of 1. Adapted from Calado et al., 2010. GC : Germinal Center B-cells L1/L2/L3: Cγ1-cre Blimp1FFeYFPstopFL lymphomas L4/L5/L6: Cγ1-cre Blimp1FFIKK2castopFL lymphomas LINCOLN'S INN FIELDS 73 SIGNAL TRANSDUCTION www.london-research-institute.org.uk/research/julian-downward The Signal Transduction Laboratory is interested in the mechanisms by which cancer cells become addicted to growth and survival signals generated by activated oncogenes and loss of tumour suppressor genes. We particularly focus on identifying unique dependencies of oncogene addicted cancer cells that might be targetable in the therapy of human cancer. Group leader Julian Downward Associate Scientist David Hancock Postdoctoral Scientists Ralph Fritsch Madhu Kumar Miriam Molina Arcas Clare Sheridan Davide Zecchin Alice Zhou Graduate Students Matthew Coelho Daniël Lionarons Heike Miess Clinical Research Fellow Alexandra Pender Scientific Officers Lai-Kay Cheung Christopher Moore Patricia Warne Masters Student Nadia Lima 74 Investigation of mechanisms of oncogene driven transformation and drug resistance Much of our work has focused on the RAS family of oncogenes and the signalling pathways that they control. RAS genes are activated by point mutation in some 20% of all human tumours and are known to play a key role in the establishment of the transformed phenotype. While the signalling pathways activated by RAS are well characterised, it remains a major challenge to identify what proteins are selectively important in the establishment and maintenance of the RAS transformed phenotype and may therefore act as potential therapeutic targets for cancer treatment. When cells become progressively transformed during the evolution of cancer, they suffer stresses that are not seen by normal cells and become increasingly dependent on stress management pathways. This means that the tumour cells show a unique set of dependencies, both on the oncogenic drivers and also on stress handling pathways, termed oncogene addiction and non-oncogene addiction, respectively. We have investigated these dependencies by functional genomic screening using RNA interference, comparing a cancer cell line containing an activated KRAS allele with a normal (‘isogenic’) derivative in which this has been removed (Wang et al., 2010; Oncogene. 29(33): 4658-4670), and also using a panel of thirty or so cancer cell lines, half of which were mutant and half wild type for KRAS (Steckel et al., 2012; Cell Res. 22(8): 1227-1245). This approach uncovered proteins whose therapeutic targeting might be expected to provide differential toxicity towards KRAS mutant tumour cells. One example of a determinant of non-oncogene addiction found in this way, the transcription factor GATA2, has been investigated in detail using genetic mouse models. The development and continued maintenance of KRAS-induced lung cancer is highly dependent on the expression of GATA2 (Kumar et al., 2012; Cell. 149(3): 642-655). Scientific report 2014 LONDON RESEARCH INSTITUTE We have also used similar genome-wide screening approaches to investigate how tumours develop resistance to targeted therapies, for example the EGFR receptor tyrosine kinase inhibitor erlotinib in the case of EGFR mutant lung cancer. This has revealed a number of genes whose loss of function can cause resistance to erlotinib in vitro. Comparison of these hits with expression profiles of erlotinib resistant and sensitive tumours from a mouse model of EGFR-driven lung cancer reveals a number of genes that are under-expressed in the resistant tumours. One of these is NF1, a negative regulator of RAS, and we find evidence in erlotinib resistant tumours that this can lead to endogenous RAS becoming activated independent of EGFR, providing an alternative mechanism for erlotinib resistance in addition to the well-documented T790M gatekeeper mutation in EGFR itself (de Bruin et al., 2014; Cancer Discov. 4(5): 606-619). The role of phosphatidylinositol 3-kinase in RAS-driven oncogenesis RAS proteins signal through direct interaction with a number of effector enzymes, including type I phosphatidylinositol 3-kinases (PI3Ks). Mice with mutations in the RAS binding domain (RBD) of the Pik3ca gene encoding the PI3K catalytic p110α isoform are highly resistant to endogenous KRAS oncogene induced lung tumourigenesis and HRAS oncogene induced skin carcinogenesis (Gupta et al., 2007; Cell. 129(5): 957-968). The interaction of RAS with p110α is thus required in vivo for RAS-driven tumour formation. We have also generated mice with inducible expression of the inactivating mutation in the RBD of p110α, so that the requirement of this interaction for maintenance of established tumours can be assessed. Blocking the RAS/p110α interaction causes partial regression and stasis of tumours, although more complete regression requires coordinate inhibition of the MEK pathway (Castellano et al., 2013 Cancer Cell. 24(5): 617-630). However, not all of these effects are tumour cell erlotinib erlotinib + MEKi final 4-weeks treatment before after MEKi Figure 1 Effectiveness of combination treatment of an erlotinib resistant EGFR driven lung cancer mouse model with a combination of EGFR and MEK inhibitors. Erlotinib resistant lung cancers in mice expressing L858R mutant EGFR in the lung were treated with erlotinib and/or the MEK inhibitor trametinib over four weeks. Tumour size before and after treatment was determined by micro CT scanning. For details, see de Bruin et al., 2014. autonomous: using a murine tumour cell transplantation model, we have demonstrated that disruption of the interaction between RAS and p110α within host tissue reduced tumour growth and tumour-induced angiogenesis, leading to improved survival of tumour-bearing mice, even when this interaction was intact within the transferred tumour. Furthermore, functional interaction of RAS with p110α in host tissue was required for efficient establishment and growth of metastatic tumours with normal PI3K, indicating that a functional RAS interaction with PI3K p110α is required in host tissue, as well as in the tumour, for the establishment of a growth-permissive environment (Murillo et al., 2014; J Clin Invest. 124(8): 3601-3611) While combined inhibition of the RAS effector pathways MEK and PI3K can cause impressive tumour regression, this combination has high toxicity that may be problematic in the clinic. We have sought to find less toxic ways of inhibiting PI3K in KRAS-mutant lung cancer cells using a drug library screen and have found that inhibition of IGF1 receptor allows this. It appears that the activation of PI3K by mutant KRAS in lung cancer cells is dependent on basal signalling by IGF1 receptor. The combination of MEK and IGF1 receptor inhibition shows potential in preclinical models of KRAS-mutant lung cancer (Molina-Arcas et al., 2013; Cancer Discov. 3(5): 548-563). We have also created a mouse with inactivating mutations in the RAS binding domain of p110β, the other ubiquitously expressed PI3K catalytic subunit isoform. Unexpectedly, we have found that the p110β isoform is not controlled by direct interaction with RAS, unlike p110α, γ and δ, but rather that the RBD of p110β interacts directly with a number of other small GTPases with distinct biological function – the RAC and CDC42 proteins. This has led us to a significantly revised model of how extracellular stimuli, especially those signalling through G protein coupled receptors, activate the PI3K activity of p110β, and the importance of this mechanism in cancer metastasis and also fibrosis (Fritsch et al., 2013 Cell. 153(5): 1050-1063). Publications listed on page 165 LINCOLN'S INN FIELDS 75 VASCULAR BIOLOGY www.london-research-institute.org.uk/research/holger-gerhardt Group Leader Holger Gerhardt Postdoctoral Scientists Raquel Blanco Claudio Franco Martin Jones Andrea Taddei Anne-Clemence Vion Graduate Students Irene Aspalter Véronique Gebala Filipa Neto Benedetta Ubezio Scientific Officers Russell Collins Anan Ragab The Vascular Biology Laboratory aims to unravel the fundamental cellular principles and the molecular control of blood vessel patterning in development and disease. Blood vessels are critical for tissue growth and healthy organ function. Effective blood vessel function requires that the endothelial cells lining the vessel assemble a patterned network of interconnected tubes with adequate diameter and branching frequency to support blood flow. As different organs serve distinct functions, and possess different metabolic requirements, blood vessel patterning bears organ specific characteristics. We use a cell biology approach in various model systems in vivo and in vitro, in combination with computational modelling, to investigate how endothelial cells respond to signals from the tissue and communicate with each other in order to orchestrate cell behaviour leading to functional network formation. Our work over the past decade established some of the fundamental cellular and molecular principles of vascular patterning through sprouting angiogenesis. Endothelial cells activated by hypoxia-regulated VEGF-A are specified into tip cells spearheading new vessel growth, and stalk cells, which proliferate and form vascular lumen. VEGF-A activity drives both tip cell migration and stalk cell proliferation, but Dll4/Notch signalling establishes the differential specification of tip and stalk cells, and thereby also the differential functional response of the endothelial cells. Our work identified that endothelial cells dynamically compete for the tip cell position by means of VEGF receptor (VEGFR) regulation. Dll4/Notch lateralinhibition appears to function as amplifier of stochastic variations in VEGF receptor levels, ultimately leading to two populations of cells: some which possess more of the signalling receptors VEGFR2 and VEGFR3 and less of the decoy/sink receptor VEGFR1 – these are tip cells; and some which possess more of the sink receptor and less of the signalling receptors – these will be stalk cells. Although it has become clear that this feedback loop operates to generate the differential pattern and thus regulates vascular branching frequency, we currently know little about the true downstream effectors of Notch. In a study focussing on a VEGF co-receptor, Neuropilin 1 (Nrp1), we tested its role in tip cell formation using mosaic loss-of-function in a 3D 76 Scientific report 2014 LONDON RESEARCH INSTITUTE mouse embryonic stem cell sprouting assay as well as in vivo (collaboration with Ian Rosewell, LRI Transgenic Services). To our surprise we found that heterozygous cells lacking just one allele of Nrp1 are incapable of forming tip cells when competing with wild type cells (Figure 1), and this holds true even when Notch is inactive. This is in marked contrast to all previous examples as Notch inhibition would normally lead to dramatic hypersprouting by driving endothelial cells to their default tip-cell response to VEGF-A stimulation. For Nrp1 loss or inhibition, this is uniquely different. We also found that Notch differentially regulates Nrp1 levels in the endothelium, and these relative differences are most fundamental in establishing the ability of cells to form tip cells. In short, these findings identified Nrp1 as the first and critical downstream effector of Notch signalling in establishing tip and stalk cells in angiogenesis. How would Nrp1, a cell surface molecule with diverse functions, achieve this? In search for the mechanism, we considered a possible cross-talk with a pathway recently shown to also impact on the ability of cells to form tip cells, that is the BMP/Smad signalling pathway. BMP9 and BMP10 have been shown to supress sprouting and tip cell formation through activation of Alk1, and downstream effectors Smad1/5. Also, TGF-β has been shown in vitro to limit sprouting by activating Alk5. Using cell culture models and Nrp1 depletion or overexpression, we found that the levels of Nrp1 affect activation of Smad2/3 when Figure 1 Nrp1-heterozygous cells do not form tip cells when competing with wild type cells in vivo. Chimeric retinal vessels derived from blastocyst injection of a wild type host with Nrp1LacZ/+ embryonic stem cells, assayed at post natal day P5. The segmented image shows the overall vasculature stained with Pecam (blue) and wild type cells, expressing a DsRED marker (red). The nuclei of the two respective genotypes were segmented using Erg staining and displayed in pseudo-colours; wild type nuclei in white and nuclei from Nrp1LacZ/+ cells in green. cells are stimulated with TGF-β. Surprisingly, although Nrp1 has been proposed to bind TGF-β and increase its activity on lymphocytes, Nrp1 expression reduced Smad2/3 activation in endothelial cells. In a series of genetic compound experiments in collaboration with the team of Anne Eichmann (Yale, USA) and drug treatments we discovered that reducing Alk1 or Alk5 expression or activity in Nrp1-deficient cells rescued their ability to form tip cells. Also, unlike Notch inhibition, inhibiting TGF-β signalling restored sprouting in Nrp1 deficient vessels, illustrating that overactivation of the TGF-β/Smad2/3 pathway is responsible for the sprouting defects in Nrp1 mutants. More work is required to unravel precisely how BMP and TGF-β signalling interact and jointly affect vascular sprouting and what the signalling effectors of Smad2/3 in the endothelium are that mediate this effect. However, given that the TGF-β pathway is genetically involved in a number of vasculopathies including hereditary hemorrhagic telangiectasia (HHT) and arterio-venous malformations (AVM), the finding that Nrp1 levels quantitatively regulate this signalling axis may turn out to be therapeutically meaningful. From the basic angiogenesis angle, our work challenged previous concepts that posit that endothelial tip cells are a default response of the endothelium to VEGF activation, whereas the stalk cell phenotype is acquired through Notch. Instead, it appears that Nrp1 expression, which is highest in tip cells, functions to actively suppress the stalk cell phenotype by limiting Smad2/3 signalling. These findings open new avenues and directions to investigate mechanisms of vascular patterning in various organs in physiology, as well as in pathological conditions. Acknowledgement: We would like to thank Ian Rosewell and his team for enormous support generating chimeric experimental mouse cohorts. We are grateful to Caroline Hill and team members, who shared tools and experience/insight into the intricacies of TGF-β signalling. Publications listed on page 165 LINCOLN'S INN FIELDS 77 Developmental Systems www.london-research-institute.org.uk/research/nathan-goehring Group Leader Nathan Goehring Postdoctoral Scientists Florent Peglion Nelio Rodrigues Graduate Students Lars Hubatsch Jacob Reich Scientific Officer Nisha Hirani Pattern-forming systems provide essential spatial cues to guide the complex multi-dimensional puzzle that is organismal development. For many of these systems, we have identified the key molecules involved; yet we are only beginning to understand how the collective activities of these molecules give rise to spatiotemporal patterns at the cell and tissue scale. How are pattern boundaries established? What sets the scale of patterns? What are the properties of networks that permit pattern formation? How do pattern-forming networks adapt during development? Our group takes an integrative approach to these questions, combining genetics, pharmacology, quantitative imaging and mathematical modelling to identify the core design principles of these systems. We are currently exploring these questions in the context of symmetry-breaking in polarised cells, a process that is essential for animal development and is commonly affected in cancer. The PAR polarity network The PAR cell polarity network is an intracellular pattern-generating system that regulates asymmetry in polarised animal cells. PAR polarity is intricately tied to the establishment of cell form, fate, and function. It plays a key role in processes as diverse as cell migration, the orientation of cells with respect to their environment, cell fate specification during asymmetric stem cell-like cell divisions, and the generation of complex cell morphologies. Not surprisingly, failure of this system is associated with developmental defects and cancer. The embryo of the nematode worm, Caenorhabditis elegans, is a near unmatched system for quantitative, imaging-based analysis of PAR polarity network behaviour in a living system: symmetry-breaking by the PAR proteins is highly stereotyped and rapid; there is a robust genetic toolkit to identify, perturb and manipulate the molecules involved; and embryos are transparent and highly amenable to quantitative microscopy. To date, our primary focus has been to develop quantitative models for polarity establishment in the one-cell embryo. Kinetic measurements of the behaviour of core network components as well as cytoskeletal disruption experiments have led us to propose that PAR proteins comprise a 78 Scientific report 2014 LONDON RESEARCH INSTITUTE self-organising pattern-forming system. Mathematical modelling confirms that a minimal reaction-diffusion scheme based on measured behaviours reproduces observed patterns of PAR polarity seen in the embryo. Notably, this model can account for symmetry-breaking in the PAR network in response to cytoplasmic fluid flows, demonstrating, in principle, how a purely mechanical cue can trigger pattern formation by this biochemical network. Going forward, our focus will be several aspects of this pattern forming system, including defining the core network features of the PAR network that enable pattern formation, identifying the detailed mechanisms that govern PAR protein mobility on cellular membranes, and examining how PAR network behaviour adapts to and is affected by changing developmental contexts. Defining core features of the PAR network At the core of the PAR polarity network is a mutually antagonistic feedback loop between two groups of membrane-associated PAR proteins. Data suggests that these two groups, anterior PARs (aPARs) and posterior PARs (pPARs), reciprocally exclude one another from the plasma membrane (Figure 1). Although we have some insight into the molecular activities responsible for mutual exclusion, the lack of mechanistic details or information regarding reaction kinetics makes formation of explicit mathematical models problematic. Indeed, existing models require assumptions about the nature of PAR feedback that remain to be tested. We are developing tools to enable acute, quantitative perturbation of network connections within the embryo, which we hope will allow us to better define core design features of this essential polarity network. Figure 1 A simplified, two-component reaction-diffusion model reproduces symmetry-breaking of the PAR polarity network in the one-cell C. elegans embryo. Here, two mutually antagonistic components exchange between membrane and cytoplasmic states. When membraneassociated, each component can displace the opposing component from the membrane such that co-existence of the two components at any given space is strongly disfavoured. This model yields a multi-stable system, capable of supporting either a stable unpolarised or a stable polarised state, reflecting the state of the embryo before and after symmetry-breaking. In the embryo, switching between these two states is triggered by cortical flows, which induce an asymmetry in membrane-associated anterior PARs (red), thereby allowing loading of posterior PARs (blue) onto the posterior membrane. Figure 2 Polarisation of the polarity protein PAR-2 during early C. elegans development. PAR-2 is segregated asymmetrically in a series of 4 polarised cell divisions before being partitioned equally into two germ line stem cells. Regulation of PAR membrane association PAR polarity is ultimately a question of membrane dynamics. Although PAR proteins actively exchange between membrane and cytoplasmic pools, and PAR polarity is ultimately responsible for setting up cytoplasmic gradients of downstream effectors, spatial segregation of PAR proteins occurs only on the membrane. Rapid diffusion and mixing of soluble PAR protein in the cytoplasm ensures that no significant spatial asymmetries arise. A key aim of our current work is to define the molecular determinants that govern association with and mobility of PAR protein on the plasma membrane, as well as how association and mobility is regulated in time and space, potentially through interactions with other members of the PAR network. Ultimately, it is these kinetic behaviours, such as membrane binding rates and diffusion coefficients that define both the ability of reaction-diffusion systems to form patterns and the characteristics of the patterns that result. PAR network behaviour in the establishment of a developmental lineage PAR proteins are responsible for the polarity of diverse cell types that vary distinctly in size, shape, and context. We are particularly interested in exploring whether physical limits impact mechanisms of symmetry-breaking in different cell types, and whether strategies have evolved to cope with such physical limits, e.g. by the addition of alternative molecular players. The early cell divisions of the C. elegans embryo provide a simplified model system to begin to explore these issues. Beginning with the division of the one-cell embryo, PAR proteins regulate a series of four consecutive asymmetric divisions during C. elegans development that are critical for restricting germ cell determinants to a pair of germ-line stem cells (Figure 2). In each of these divisions, the PAR proteins are polarised and asymmetrically distributed between the two daughter cells. However, both the size of these cells and their arrangement with respect to neighbours differ significantly, thus, providing an ideal system for examining how properties of the PAR system adapt over time in a developmental lineage. We are using our quantitative models as a starting point for testing predictions for how a minimal PAR system could adapt to this changing environment. Publications listed on page 166 LINCOLN'S INN FIELDS 79 Immuno Surveillance www.london-research-institute.org.uk/research/adrian-hayday Group Leader Adrian Hayday Postdoctoral Scientists Lucie Abeler-Dorner Sara Cipolat Livija Deban Deena Gibbons Yasmin Haque Rosie Hart Marialuisa Iannitto Fernanda Kyle Adam Laing Olga Sobolev Pierre Vantourout Martin Woodward Graduate Students Deborah Enting Rafael di Marco Barros Bodhi Hunt Sean O’Farrell Rick Woolf Scientific Officer Anett Jandke Figure 1 The murine small intestinal epithelium is normally enriched in γδ T cells, relative to αβ T cells (green vs red; left-hand panel) viewed by confocal microscopy of the gut. Conversely, mice mutant in a single intestinal-specific member of the B7 superfamily (right-hand panel) show a severe reduction in γδ T cells because of the selective loss of cells expressing Vγ7, the signatory T cell receptor of the small intestine. Moreover, most residual γδ T cells express lower levels of the TCR, and hence show less bright green staining (image courtesy of Rosie Hart). 80 Infections occur largely at body surfaces, and it is therefore not surprising that many organs, including skin, intestine and reproductive tract harbour major immune compartments. Such tissue-resident immune cells may play key roles in the initiation and progression of solid tumours, many of which develop at body surfaces. Ironically, tissue-resident immune cells have received little attention relative to their circulating counterparts. In seeking to redress this balance, we hypothesised that epithelial cells in discrete anatomical sites express molecules that determine the unique composition of their associated T cell compartments. Such molecules may thereby play key roles in cancer immunosurveillance. Supporting our hypothesis, we identified Skint1 as a novel gene regulating the signatory T cell compartment of the murine epidermis. We have now obtained data suggesting the generality of this principle, and showing that molecules such as Skint1 constitutively communicate to local T cells whether a tissue is normal or dysregulated. Epithelial orchestration of immune compartments An iconic, 20-year-old picture in immunology depicts the association of discrete sets of T cells with different organs. Thus, γδ T cells expressing Vγ5+ T cell receptors (TCRs) compose the signatory epidermal γδ T cell compartment; Vγ6+ T cells populate the reproductive tissues; while Vγ7+ T cells populate the small intestine. These associations are significant, since mice in which we replaced skin Vγ5+ cells with other γδ T cells became hypersensitive to skin irritants and carcinogens (Strid et al., 2008; Nat Immunol. 9(2): 146-154). However neither the biological rationale nor the molecular basis for organ-specific T cell associations is known. We considered that epithelial cells in different organs might express unique ‘address-ligands’ that selectively engage T cells specifically associated with the respective organs. Supporting this, we and our collaborators identified Skint1, a novel immunoglobulin superfamily gene expressed exclusively by thymic epithelial cells and keratinocytes and upon which Scientific report 2014 LONDON RESEARCH INSTITUTE the normal development of skin Vγ5+ T cells depends (Turchinovich and Hayday, 2011; Immunity. 35(1): 59-68). Whereas initial studies of Skint1 related to its developmental regulation (‘selection’) of thymic Vγ5+ T cell progenitors, our studies throughout 2012-2014 established that Skint1 regulates steady-state contacts of mature Vγ5+ T cells with suprabasal keratinocytes, thereby maintaining an appropriate state of peripheral immune regulation. Our ongoing studies focus on the nature of those Skint1-dependent contacts: do they include direct Skint1-TCR interactions, or do they feature novel, organ-specific checkpoint regulators akin to the activity of PD1 on T cells that infiltrate tissues from the systemic compartment? Novel epithelial T cell regulators Skint1 provides a prototype for how the epithelium determines the composition and status of an organ-specific T cell compartment. Skint1 is distantly related to B7 genes that include CD80, B7H4, and PD-L1 that are profoundly effective, clinically significant T cell regulators. Thus, other B7-like, Skint1-related genes might encode epithelial regulators of other T cell compartments. Indeed, we have identified mice mutant in a B7-like gene, expressed specifically in small intestinal epithelial cells, and in which intestinal T cells expressing the signatory Vγ7+ TCR are almost completely absent (Figure 1). However, unlike yet unknown, tissue-specific B7-like factors that we now seek (Figure 2). We are likewise investigating the pathophysiologic roles played by human innate-like γδ T cells, and developing clinically applicable methods to regulate tissue-associated γδ T cell activities within the context of human tumour immunotherapy. Figure 2 Schematic representation of members of the extended B7-supergene family that includes epithelial regulators of discrete γδ T cell compartments. (IgV; IgC refer to domains similar to Immunoglobulin Variable and Constant regions, respectively; B30.2 denotes a domain unique to BTN/Btnl and TRIM proteins; diagonal line denotes membranespanning region; upper case type denotes a human protein; lower case denotes murine proteins) thymic, Skint1-dependent regulation of epidermal T cells, epithelial ‘selection’ of Vγ7+ T cells occurs exclusively in the gut, perhaps consistent with the required turnover of intestinal γδ T cells throughout life, even after thymic involution. The biology and underpinning mechanism of this first example of intestinal T cell repertoire selection is a prime focus of ongoing studies. Figure 3 T lymphocytes in newborn babies display a novel functional potential to produce IL-8 (CXCL8), which correlates with increases in CRP, commonly diagnostic of neonatal sepsis. No such correlation is shown by conventional T cell cytokines, e.g. interferon-gamma (IFNγ) or interleukin-17 (IL-17). Thus, CD4 + T cell production of CXCL8 may prove a useful new neonatal biomarker, and offer functional insight into human T cell mediated immunosurveillance. Human tissue T cells orchestrate immune responses We first showed that epidermal T cells can respond in vivo to innate ‘alarmins’, rather than require specific TCR-dependent antigen stimulation. These findings, since validated by many other laboratories, fuelled the notion of ‘innate-like’ T cells that orchestrate downstream immune responses following their rapid activation by tissue dysregulation. Such tissue-resident, innate-like T cells might form a first-line of defence to infections and/or early stages of tumourigenesis. To assess whether humans harbour such innate-like T cells, our clinical research team at Guy’s Hospital has refined methods for T cell isolation, permitting us to confirm that human skin and gut each harbour vast lymphocyte compartments comprising many different T cell types. Among those, we have identified the first examples of human tissueresident, innate-like γδ T cells specifically associated with either skin or gut. Moreover, some similarities of those cells with their mouse counterparts are so striking as to fuel the hypothesis that they, too, may be regulated by as In the very young Although the young are most vulnerable to infection, there has been little study of T cells in human neonates. Moreover, such data as exist show an inability of neonates’ T cells to produce cytokines such as interferon-gamma, underpinning the view that neonates are immunodeficient. Based on our elaboration of innate-like T cell responses, we asked whether T cells in neonates – rather than lacking function – perform functions more commonly associated with innate cells, e.g. monocytes. Indeed, we provided the first evidence that the major effector potential of neonates’ CD4+ T cells is production of interleukin-8 (IL-8), a major activator of neutrophils with the potential to protect babies against relentless exposure to bacteria (Gibbons et al., 2014; Nat Med. 20(10): 12060-1210) (Figure 3). We are now asking whether IL-8-producing T cells are limited to childhood, or are implicated in immunoprotection and/or immunopathologies in adults. Big Science We further built up ‘3i’, a mutli-centre consortium aiming to comprehensively immunophenotype >800 gene knockout mouse strains over five years. After only 18 months of unprecedentedly highthroughput immunological analysis, 3i is already providing substantial new insight into genes regulating immune cell development and function, including within tissues, thereby germinating many new projects. In parallel, our comprehensive immunemonitoring of human responses to vaccination has been expanded via collaboration with Momenta Pharmaceuticals (Cambridge, MA) with whom we are developing novel ways to analyse and disseminate huge human immunology data-sets. A key goal is to improve immunological assessment of cancer patients, thereby guiding clinical decision-making in relation to immunotherapies. Publications listed on page 166 LINCOLN'S INN FIELDS 81 DEVELOPMENTAL SIGNALLING www.london-research-institute.org.uk/research/caroline-hill Group Leader Caroline Hill Postdoctoral Scientists John Chesebro Claire Heliot Annasuya Ramachandran Antonius van Boxtel Graduate Students Davide Coda Tessa Gaarenstroom Daniel Miller Scientific Officers Debipriya Das Ilaria Gori Work in the Developmental Signalling group focuses on the signalling mechanism and function of Transforming Growth Factor β (TGF-β) superfamily ligands, which include the TGF-βs, Activins, Nodals, BMPs and GDFs. Many of these ligands play fundamental roles in early vertebrate development, acting as morphogens in the specification and patterning of the germ layers, and aberrant TGF-β, BMP and Nodal signalling is implicated in many different types of cancer. A major goal of the lab is to understand at the molecular level how these ligands signal from the plasma membrane to the nucleus and how they regulate transcription of target genes. We want to determine how they function and are regulated in embryonic development, using zebrafish as a model system, and how these signalling pathways are hijacked by tumour cells to promote growth of primary tumours and metastasis to distant sites. The highlights this year have been our genome-wide analyses addressing how activated Smads induce transcriptional activation via chromatin, our mechanistic work focused on signalling dynamics, and our in vivo studies on Nodal signalling in zebrafish. Regulation of transcription by activated Smad complexes The main function of the TGF-β superfamily/Smad pathways is to induce new programmes of gene expression, but apart from previous work from my group showing that Smads activate transcription by remodelling the chromatin template, little is understood about how exactly they achieve this. Our work in this area is focused on Nodal signalling, which is not only critical for embryonic development, and for maintaining pluripotency of human embryonic stem (ES) cells, but is exploited by cancer cells to promote tumour progression and metastasis. We are using the embryonic carcinoma cell line, P19, which responds to Nodal and Activin both acutely and chronically as a model system. As well as being a model for cancer, these cells express a combination of pluripotency factors and mesendoderm markers in response to Activin/ Nodal. To characterise the transcriptional responses to Activin we performed RNA-seq on P19 cells in the non-signalling state, in cells treated with Activin for short (1 h) or extended (8 h) times, and in the untreated state (autocrine signalling). This has allowed us to define four main classes of response genes (transiently induced; induced sustained; delayed induced; repressed), which is 82 Scientific report 2014 LONDON RESEARCH INSTITUTE enabling us to define different classes of enhancers that are co-regulated. In the same conditions we have also performed ChIP-seq analysis for Smad2, for total histone H3, for two different histone modifications characteristic of active chromatin (H3K27ac and H3K9ac), and for two different forms of RNA Polymerase II (initiating state phosphorylated at Ser 5, and elongating state phosphorylated at Ser 2). Ongoing work integrating the RNA-seq data with all of these ChIP-seq datasets is revealing exciting and novel insights into the mechanism whereby activated Smad2containing complexes find their targets in chromatin and activate transcription. Motif enrichment analysis is being used to define what other cofactors bind with Smad2 at enhancers of Activin-responsive genes, distinguishing between those that bind with Smad2 1 h after Activin stimulation and those that are synthesised in response to Activin and bind with Smad2 at later time points. This is yielding a number of interesting candidates, which are being experimentally verified. The dynamics of TGF-β signalling Understanding how cells respond to ligands in complex physiological and pathological contexts in vivo requires knowledge of the dynamics of signalling in addition to an understanding of the molecular wiring of the pathway. This year we have been building on our analysis of the dynamics of TGF-β signalling, which indicated that an acute TGF-β stimulation resulted in refractory behaviour as a result of rapid internalisation of activated receptors from the cell surface, and their very slow recovery. We have now performed several whole genome siRNA screens to identify new regulators of TGF-β signalling dynamics and are building receptor biosensors to track the trafficking route of activated receptors. We are also extending this analysis to other TGF-β superfamily signalling pathways and have shown that neither BMP, nor Activin or Nodal stimulation triggers refractory behaviour. In these cases, cell surface receptors appear to continuously monitor ligand in the extracellular milieu. We are investigating mechanistically how this occurs. Spatial regulation of Nodal signalling To understand how TGF-β superfamily ligands function in vivo, we are using early zebrafish embryos as a model system. We want to determine how ligand activity is regulated, how the ligands function in a dose-dependent manner and how they contribute to tissue specification. This year we have been focusing on spatial and temporal control of Nodal signalling. To visualise Nodal signalling in vivo, we generated a transgenic zebrafish line in which an EGFP reporter is controlled by 3 copies of the Activin-responsive enhancer (ARE), which binds a complex of activated Smad2–Smad4 with the transcription factor FoxH1. Using EGFP mRNA as a readout we can track active Nodal signalling in a developing embryo, and have made some surprising discoveries. Shortly, after Nodal signalling is initiated, it is visible in the embryo margin in a shallow dorsal-ventral gradient, but contrary to long standing assumptions in the field, it extends only four or five cell diameters towards the animal pole, coincident with the expression domains of cyclops and squint (the fish Nodal ligands), and lefty1/2 (the antagonists), and is not graded in this direction. The predominant model to explain the generation of domains of Nodal signalling is the reaction-diffusion model, which is based on the assumption that a highly diffusible inhibitor (in this case Lefty1/2) and a less diffusible activator (Nodal ligands) can create a network as a result of short-range activation and long-range inhibition. Our findings do not support this model. Instead, our results are leading us to an alternative model, whereby a temporal delay in the translation of the ligand antagonists Lefty1/2 allows Nodal signalling to become established in four to five cell tiers at the margin, at which point Lefty protein levels reach a sufficiently high threshold to prevent further spread of Nodal signalling. We are determining the mechanism underlying this behaviour. Our data is suggesting that rather than a spatial gradient of Nodal signalling in the early embryo, the gradient may be temporal. Publications listed on page 166 Figure 1 Regulation of transcription by activated Smad complexes. Schematic indicating the molecular events that occur to enable activated Smad2–Smad4 complexes to regulate transcription. The Smad complexes bind to enhancer DNA through interactions with other transcription factors, such as FoxH1, as well as other cofactors. Smad binding induces chromatin remodelling through the SWI/SNF complex and via histone modification, which may include demethylation of H3K27 and H3K9, acetylation via p300 of multiple lysines on H3 including H3K27 and H3K9 (depicted as AcH3) and trimethylation of H3K4. Chromatin looping results in recruitment of mediator and RNA polymerase II (Pol II) at the transcription start site. Pol II becomes phosphorylated initially at serine 5 and subsequently at serine 2, which is its elongating form. LINCOLN'S INN FIELDS 83 COMPUTATIONAL BIOLOGY www.london-research-institute.org.uk/research/nicholas-luscombe Group Leader Nicholas Luscombe Postdoctoral Scientists Federico Agostini Aylin Cakiroglu Elodie Darbo Borbala Gerle Raphaelle Luisier Anna Poetsch Andrew Steele Alessandra Vigilante Katharina Zarnack Graduate Students Filipe Cadete George Cresswell Arsham Ghahramani Robert Sugar Cellular life must recognise and respond appropriately to diverse internal and external stimuli. By ensuring the correct expression of specific genes at the appropriate times, the transcriptional regulatory system plays a central role in controlling many biological processes: these range from cell cycle progression and maintenance of intracellular metabolic and physiological balance, to cellular differentiation and developmental time-courses. Numerous diseases result from a breakdown in the regulatory system and a third of human developmental disorders have been attributed to dysfunctional transcription factors (TFs). Furthermore, alterations in the activity and regulatory specificity of TFs are now established as major sources for species diversity and evolutionary adaptation. Indeed, increased sophistication of the regulatory system appears to have been a principal requirement for the emergence of metazoan life. Much of our basic knowledge of transcriptional regulation has derived from molecular biological and genetic investigation. In the past decade, the availability of genome sequences and development of new laboratory techniques have generated (and continue to generate) information describing the function and organisation of regulatory systems on an unprecedented scale. Genomic studies now allow us to examine regulatory systems from a whole-organism perspective; on the other hand however, many observations made with these data are unexpected and appear to complicate our view of gene expression control. The continued flood of biological data means that many interesting questions require the application of computational methods to answer them. The combination of computational biology and genomics enables us to uncover general principles that apply to many different biological systems; any unique features of individual systems can then be understood within this broader context. The Computational Biology Group applies computational and genomic methods to answer three main questions: (i) How is gene expression regulated? (ii) How do these mechanisms control interesting biological behaviours? (iii) How does gene regulation interact with evolutionary processes? Much of our work until now has been purely computational, either analysing publicly available 84 Scientific report 2014 LONDON RESEARCH INSTITUTE data or in collaboration with experimental laboratories performing functional genomic investigations. Research highlights • Prevention of aberrant exonisation of Alu elements In collaboration with Jernej Ule’s group (UCL , Institute of Neurology) we developed nucleotide-resolution, genome-wide techniques to identify protein-RNA interactions (Konig et al., 2010; Nat Struct Mol Biol. 17(7):909-15). We demonstrated how hnRNP C binds to enhanced and repressed splice sites. Recently, we discovered how competitive binding between U2AF65 and hnRNP C protects the transcriptome from the detrimental exonisation of thousands of Alu elements (Zarnack et al., 2013; Cell. 152(3):453-66). • Statistical models of gene expression in fly development Using compiled in situ hybridisation images from the Virtual Embryo dataset, we developed statistical models that for the first time accurately reproduce even skipped expression. Importantly, the models precisely forecast behaviours beyond the training data, making them truly predictive (e.g. effects of regulatory perturbations). The study generated experimentally testable hypotheses and provided new insights into the underlying mechanisms of transcriptional regulation (Ilsley et al., 2013; Elife. 2:e00522) Figure 1 Schematic explaining how hnRNP C binding to pre-mRNAs aids accurate splicing, whereas its loss leads to aberrant exonisation. Future work Nuclear organisation of chromosomes It is increasingly appreciated that the spatial organisation of chromosomes profoundly influences gene expression; however the details of how this is achieved are poorly understood. We will build on our successful collaborations with the Akhtar group (Max Planck Institute of Immunobiology and Epigenetics, Freiburg, DE) to study the effects of X-chromosomal positioning on dosage compensation. Excitingly, we recently initiated collaborations with Peter Fraser (Babraham Institute, Cambridge), a world-expert on ChIA-PET and Hi-C, to investigate nuclear organisation in mammalian cells. Gene regulation in disease states We will apply our basic knowledge of gene regulation to disease systems. There are indications that bacterial infections cause changes to the host’s regulatory system, so affecting expression patterns. We have initiated Figure 2 A. Predicted expression of even-skipped 2 (eve 2) wild-type Drosophila embryos. B. Predicted eve 2 expression in giant (gt) mutant Drosophila embryos. C. in situ hybridisation of gt mutant from Small et al., 1992: EMBO J. 11(11):4047-57. A B collaborations with Richard Hayward (University College London) to apply genomic techniques to investigate the prevalence of these effects, and the influence they have on the progression of bacterial infections. Gene regulation and DNA-damage repair A major implication of our mutation rate study is that highly expressed genes are preferentially protected from DNA damage; however mechanisms such as transcription-coupled repair do not explain our observations. There are early indications that similar mechanisms operate in cancer. DNA damage repair is traditionally studied from a molecular perspective: we have initiated collaborations with the Mammalian Genetics Group to examine this phenomenon from a genomic viewpoint, too. This will dramatically improve understanding of how DNA damage repair operates on a genome-wide scale. Publications listed on page 166 C LINCOLN'S INN FIELDS 85 TUMOUR HOST INTERACTION www.london-research-institute.org.uk/research/ilaria-malanchi Group Leader Ilaria Malanchi Postdoctoral Scientists Laurie Gay Luigi Ombrato Graduate Students Yaiza del Pozo Martin Sathya Muralidhar Misa Ogura Stefanie Wculek Scientific Officer Robert Moore Figure 1 A. Schematic representation and immune-fluorescent images of cancer cells (green) within the lung during metastatic colonisation and B. during micro-metastatic establishment. Modifications within the metastatic tissue are detected by Smooth Muscle Actin staining (red). 86 A cancer cell surrounded directly by the ‘normal’ host environment of a tissue would be unable to develop into a tumour. Only by modifying their environment, cancer-initiating cells survive, proliferate and build a supportive stromal structure consisting of tumour associated host cells locally or systemically recruited. The aim of our group is to understand the crosstalk that tumour cells establish with the host organism to allow both tumour onset and metastatic progression. Tumours are composed by a growing mass of cancer cells embedded into a heterogeneous assembly of tissue-derived cells. These cells are collectively referred to as tumour-associated stroma. Cancer cell growth is always accompanied by a concomitant modification in the surrounding host tissue and these two components co-evolve during tumour progression. Within the cancer cell compartment, cancer cells are very heterogeneous in their intrinsic tumourigenic potential and only a small fraction of cells, termed Cancer Stem Cells (CSCs), retain the potential of sustaining long-term growth. CSCs are the precursor of all cancer cells and they can replace part or the entire tumour if required. The relevance of CSCs’ intrinsic potential is maintained during metastatic progression. We have previously provided direct evidence that a sub-pool of cells (metastatic CSCs) identified within A Metastatic Colony B Micro-metastasis Scientific report 2014 LONDON RESEARCH INSTITUTE the primary tumour, drive metastatic colonisation (Malanchi et al., 2011; Nature. 481(7379): 85-89). Importantly, metastatic CSCs also rely on signals derived from their local microenvironment, referred to as ‘niche’, for controlling their behaviour and maintenance. We aim to clarify how the metastatic cells create their niche, as well as the crosstalk they establish with it to allow metastatic outgrowth. Tumour-host interactions extend well beyond the local tissue microenvironment and tumours not only respond to, but also actively perturb host organs at distant anatomic sites. For instance, some inflammatory cells, once assumed to collectively act only to attenuate tumour development, clearly play a role in tumour promotion and malignancy. Indeed, inflammatory Figure 2 Metastasis from ‘niche-labelling cancer cells’. A. Immunofluorescence of metastasis from niche-labelling cells expressing both GFP and CHERRY proteins. Niche cells are the tissue cells retaining only CHERRY protein. White lines outline the borders between cancer and niche cells, and which are removed in the inset. B. FACS analysis of lungs injected with niche-labelling cancer cells. Unstained cells represent lung tissue cells. GFP-CHERRY double positive cells represent the cancer cells growing in the lung. CHERRY single positive cells are the subset of lung tissue cells labelled by the cancer cells and which represent the metastatic niche. A B Metastatic niche conditions are able to increase the speed of tumour progression and chronic inflammation is the best-known critical carcinogenic risk factor. By focussing on a particular immune cell compartment, the neutrophils, which play a fundamental role in inflammatory responses, we investigate the interaction of tumour and inflammatory cells in the context of primary tumour formation and metastatic onset. Cancer stem cells and their metastatic potential What defines a cancer cell’s metastatic potential? To answer this question, we consider two facts: firstly, cells capable of metastasising belong to a sub-pool of cancer cells that retain the intrinsic tumour initiation potential (stemness) of reconstituting a cancer cells mass. Secondly, during early metastatic colonisation, cancer cells change their microenviroment to create their niche (Figure 1). Therefore, the ability of cancer cells to initiate an extrinsic niche will also impact metastatic competence. We have previously identified a sub-pool of metastatic CSCs (mCSCs) within primary tumours of the MMTVpyMT mouse model for their exclusive metastatic initiation potential. Therefore, studying the characteristics of those mCSCs in comparison to the rest of the cancer cells is guiding us to identify specific intrinsic and extrinsic metastatic potential. Despite its crucial function, metastatic niche composition and its evolution during the different phases of metastatic progression, is yet to be understood. To help characterise the niche, we have generated a unique tool capable of labelling in vivo tissue cells coming in close proximity with metastatic cancer cells. This has not been achieved to date. A specially designed expression tool integrated in the genome of cancer cell lines allows for expression of two fluorescent proteins: a green fluorescent protein (GFP), which is retained within the cell and a red fluorescent protein (CHERRY), which is secreted by the cell and can be taken up by neighbouring cells (Figure 2). This unique tool allows us to identify niche cells surrounding the cancer cells during the different phases of metastatic establishment: from colonisation to first micro- and then macro-metastases. Finally, by combining the expression profile changes occurring in both cancer and niche cells during early and late stages of metastasis, we aim to clarify the signals of this crucial crosstalk with the final aim to develop blocking strategies for clinical applications. Pro-tumourigenic activity of neutrophils The pro-tumourigenic and pro-inflammatory activity of some innate immune cells has been characterised, however, few studies have concentrated on neutrophils. Neutrophils are the first cells migrating towards the inflammatory site and are crucial to amplify and control the inflammatory response. We found these cells consistently present during initiation of both primary tumours as well as metastases. Remarkably, lack of neutrophils reduces the efficiency of both tumourigenesis and metastatic progression. Using genetic and chemical strategies to block neutrophils or a certain neutrophil activity, we aim to clarify their tumour promoting functions during initiation of both primary tumour and metastases. These approaches will be clinically relevant to antagonise tumour onset and metastasis formation. Publications listed on page 166 LINCOLN'S INN FIELDS 87 STRUCTURAL BIOLOGY www.london-research-institute.org.uk/research/neil-mcdonald Group Leader Neil Q McDonald Postdoctoral Scientists Rakhee Chauhan Julio Martinez-Torres Agata Nawrotek Ivan Plaza-Menacho Peter Saiu Graduate Students Emily Burns Robert Constable Marina Ivanova Scientific Officers Maureen Bowles Phillip Knowles John Lally Andrew Purkiss Growth factor signalling deregulation is a hallmark of cancer and is frequently associated with altered catalytic functions of pathway components. These components form molecular complexes that transduce signals across or within the cell membrane and can combine allosteric, catalytic or recruitment mechanisms to propagate growth factor-initiated signals. Our primary research aim is to understand the assembly, activation and deregulation of growth factor activated complexes that influence spatial signalling. Our group uses structural methods (crystallography, electron microscopy and SAXS), biophysical methods (calorimetry and fluorescence polarisation), enzymatic and cell-based assays, to elucidate signalling mechanisms. Cell surface receptor activation and oncogenic deregulation Receptor tyrosine kinases (RTKs) respond to extracellular ligands received at the cell membrane and undergo ligand-dependent activation triggering intracellular signalling pathway activation. We study the RET receptor as a model RTK to understand how ligands are recognised and how this interaction drives RET tyrosine kinase activation by allosteric or/and clustering mechanisms. This is important as RET signalling is crucial for both embryonic and adult development, whilst RET missense mutations underlie at least three human diseases (Hirschsprung’s disease, kidney agenesis and cancer). The RET receptor is activated by a group of GDNF family ligands (GFLs) that belong to the cystine knot growth factor superfamily. GFLs can only engage RET if presented by a GFRα co-receptor. The co-receptor is linked to the cell surface by a GPI-linkage but a soluble form can also bind GFLs and stimulate RET activation in trans. To characterise the interaction between RET extracellular domain (ECD) and its bipartite GDNF-GFRα ligand, we reconstituted two vertebrate ternary complexes (TC) containing RET ECD and a GDNF-GFRα ligand. We used a hybrid structural approach to define the RET ECD organisation by low-angle X-ray scattering and determined negative-stain electron microscopy reconstructions for both vertebrate RET ternary complexes (Goodman et al., 2014; Cell Rep. 8(6): 1894-1904). The RET ternary complex structures indicate that RET contacts are primarily driven by co-receptor interaction. Dimeric GDNF primarily serves as a co-receptor cross-linking function. It 88 Scientific report 2014 LONDON RESEARCH INSTITUTE establishes that RET has an unusual composite binding site closer to cytokine receptor assemblies, with discrete contacts and large gaps between the bipartite ligand and the RET ECD. Another important feature of the RET–ligand complex is a homotypic RET cysteine-rich region (CRD) C-tail interaction. Deletion of this region enhances ligand affinity for RET ECD suggesting that ligandbinding forces an unfavourable membrane proximal self-association of the CRD C-tail. This is highly relevant for multiple endocrine neoplasia type 2A (MEN2A) cancer patients with missense mutations within the CRD C-tail generating a disulphide-crosslinked and constitutively activated form of RET. One downstream consequence of RET-ECD interaction with GDNF-GFRα ligand is the generation of highly specific phospho-tyrosine sites within the RET intracellular region. These sites are generated in trans by activation of the RET tyrosine kinase to phosphorylate a neighbouring RET molecule. We found that regions flanking the tyrosine kinase domain are targeted much earlier than the canonical RTK ‘activation’ loop sites. Comparing tyrosine phosphorylation site kinetics by label-free mass spectrometry revealed that aggressive RET tyrosine kinase domain mutations found in MEN2B patients present a better autophosphorylation substrate than wild type RET (Plaza-Menacho et al., 2014; Mol Cell. 53(5):738751). We are continuing to characterise how regions flanking the core RET kinase domain contribute to ligand-dependent activation as well as how they are perturbed in an oncogenic context. Figure 1 Recognition of a bipartite GDNF-GFRa1 ligand by the RET extracellular domain (RET-ECD). A. Reconstitution of recombinant RET-ECD complex containing a GDNF-GFRa1 ligand. B. Electron microscopy reconstruction and fitted structural model for the RET-ECD-GDNF-GFRa1 complex. C. Location of a crucial receptorligand interaction ‘hotspot’ centred on site III. D. Quantification of RET-ECD binding to immobilised GDNF-GFRa1 mutant ligands. Site III mutant contained a triple mutation within GFRa1. A B Our earlier structural work on full length RET identified a folding bottleneck that could be eliminated by substitution of just two amino acids. Removing these residues in RET Hirschsprung’s (HSCR) disease mutants restored cell surface localisation overcoming a maturation block that otherwise left these RET missense mutants trapped in the ER as immature forms. To uncover pathways unique to RET maturation, we recently carried out a biased siRNA knockdown screen to identify components that restored RET HSCR mutant cell surface location and ligand-dependent signalling. Several putative hits identified were amenable to selective chemical inhibition allowing us to rapidly validate our initial screen. Current efforts are characterising these hits to demonstrate RET signalling can also be restored. Understanding pathways and uncovering new components that control RET maturation and prevent RET HSCR mutant export may present new drug targets to modulate RET signalling and novel approaches to treat HSCR disease. Polarity signalling assemblies at the plasma membrane A second major project of our group investigates the structure, activation and substrate recruitment mechanisms of the atypical protein kinase C (aPKC) isoforms iota and zeta (PKCι and PKCζ). These serine/threonine kinases play important roles in many types of cell polarity and asymmetric cell division. They have less well understood functions in cell proliferation as well as in Ras-dependent transformation. The aPKC kinase is a component of a larger assembly called the Par complex that contains the binding partner Par6, the small GTPase Cdc42 and a transiently associated Par3 PDZ-containing protein. We have reconstituted the C D Par complex to investigate its biochemical properties, to define the functional role of each component and to initiate structural studies. Recent results for Par3 interaction with aPKC suggest two discrete binding modes. We have collaborated with the Protein Phosphorylation Laboratory and Epithelial Biology Laboratory to validate our biochemical and structural findings in drosophila and mammalian cell lines. The Par complex is localised to the apical membranes of epithelial cells where it targets a specialised set of membrane-associated substrates. Once phosphorylated, Par complex substrates alter their subcellular localisation, frequently being displaced from the apical membrane to other membrane domains. We are interested in substrate recruitment mechanisms used by the Par complex. Validated polarity substrates such as Par1, Par3, LLGL2 and Kibra all have basic residues flanking a phospho-acceptor site. However, short peptides (4-14 residues) derived from these proteins are often poor surrogates for intact protein substrate counterparts suggesting recognition elements may exist remote to the phospho-acceptor site. In collaboration with the Protein Phosphorylation Laboratory, we recently uncovered a motif used by aPKC to engage a subset of PKCι substrates including the tumour suppressor LLGL2 (Linch et al., 2013; Sci Signal. 6(293): ra82). Surprisingly, PKCι missense mutants associated with human cancer map within this motif highlighting the importance of PKCι substrate recruitment. We continue to investigate the structural basis for the recruitment of these PKCι substrates to the Par complex. Publications listed on page 167 LINCOLN'S INN FIELDS 89 CELL CYCLE www.london-research-institute.org.uk/research/paul-nurse We use the simple eukaryote Schizosaccharomyces pombe (S. pombe) as a model for understanding processes that are important for eukaryotic cell biology. S. pombe is a single celled rod-shaped eukaryote that grows by tip elongation and divides by medial fission. It has a typical eukaryotic cell cycle and genome organisation, with around 70% of its genes conserved in humans. It thus provides a simple system to study complex problems of eukaryotic cell biology. Our current research consists of four main areas: (i) The mitotic and meiotic cell cycles, (ii) Entry into the meiotic cell cycle, (iii) Size control and (iv) Chemical biology. Group Leaders Paul Nurse Jacqueline Hayles Research Co-ordinator Ryoko Mandeville Postdoctoral Scientists Pilar Gutierrez-Escribano Francisco Rivero Navarro Graduate Students Martina Begnis Helena Cantwell James Patterson Matthew Swaffer Elizabeth Wood Scientific Officers Jessica Greenwood Linda Jeffery Andrew Jones Richard Lewis Juan-Juan Li 90 The mitotic cell cycle Matthew Swaffer is using the bipartite CDK-CyclinB complex, which brings about an ordered progression of the eukaryotic cell division cycle to investigate how activity of a single CDK can differentially initiate S phase and mitosis. Using phospho-proteomics (with Andrew Jones in the Protein Analysis and Proteomics Facility), and chemical manipulation of CDK activity, Matthew has defined CDK-dependent phosphorylation events in S phase and mitosis, of both known and novel CDK substrates. Around 10% of the phosphoproteome in mitosis appears to be dependent on CDK activity with around 10-fold more CDKdependent phosphorylation events in mitosis than S phase. CDK-dependent phosphorylation events that first appear in S phase are sustained across the cell cycle. In contrast, mitotic phosphorylation events increase dramatically upon mitotic entry. Both S phase and mitotic substrates are dephosphorylated with similar kinetics at mitotic exit. These experiments are consistent with low CDK activity phosphorylating S phase substrates and higher kinase activity being required for mitotic substrate phosphorylation. Eukaryotes duplicate their genomes using multiple replication origins. Atanas Kaykov has investigated the organisation of origin firing along chromosomes, using a genome-wide analysis on single DNA molecules around the length of a fission yeast chromosome (Figure 1). He has shown that at S phase onset origins fire randomly and sparsely throughout the genome, whereas later in S phase clusters of fired origins appear in these sparser regions and form nuclear replication foci. The rate of origin firing during S phase gradually increases, peaking just before mid S phase. Towards the end of S phase nearly all unfired available origins fire within the unreplicated regions, contributing to the timely completion of genome replication. The Scientific report 2014 LONDON RESEARCH INSTITUTE data show that origin firing is mostly random and that the majority of origins do not fire as a part of a deterministic programme. To examine origin firing during the meiotic cell cycle Jenny Wu showed that local changes in origin efficiencies, led to changes in meiotic recombination frequencies due to an increase in the binding of Rad51 recombination factor. This suggests that origin selection determines recombination frequencies, and shows that modifications in the DNA replication programme can modulate cellular physiology (Wu and Nurse, 2014; Mol Cell. 53(4): 655-662). Wild type cells progressing through the meiotic cell cycle require both mitotic cyclins and meiosis specific cyclins. Pilar Gutierrez-Escribano has analysed the requirements for these different cyclins and shown that the bipartite CDK-CyclinB complex, when moderately overexpressed, allows cells to undergo a near normal meiotic cell cycle in the absence of other cyclins. This shows that qualitatively different CDK complexes are not required for cell cycle progression either during the meiotic or mitotic cycles. To identify gene products important for the timing of progression through the cell cycle, Linda Jeffery identified 17 genes that are haploinsufficient for cell cycle progress. These include cdc2, cdc25 and wee1, previously shown to be haploinsufficient for mitotic entry. Other genes that regulate CDK1 activity at the G2/M transition, as well as genes involved in nuclear cytoplasmic transport, and protein translation, were also identified as haploinsufficient. Recently, Jessica Greenwood and James Patterson have initiated new projects. Jessica is using Figure 1 A combed single 5.6Mb DNA molecule approximately the length of chromosome I. Yellow, newly synthesised DNA; red, unreplicated DNA. genome-wide ChIP-seq to investigate chromatin localisation of Cdc2 and Cdc13 throughout the cell cycle, and James is investigating how the variability in the number of molecules governs cellular processes and influences cell cycle progression. Entry into the meiotic cell cycle Deletion of the zfs1 gene affects timing of the mitotic cell cycle G2/M transition independently of CDK Tyr15 phosphorylation. Francisco Navarro identified a further role for Zfs1 at the transition between the mitotic and meiotic cell cycles. Zfs1 is a CCCH-type tandem zinc finger RNA binding protein and sequencing of Zfs1-associated RNAs identified transcripts, both protein coding and non-coding, as potential targets of Zfs1 regulation. The mRNA of the G1 cyclin Puc1 is a target of Zfs1, which has a destabilising effect on the puc1 transcript. Reduction in levels of Puc1 is necessary for entry into the meiotic programme, suggesting that the effect of Zfs1 on Puc1 levels is decisive for this transition. Size control Fission yeast shows a tight control over cell size at entry into mitosis, with wild type cells dividing at around 14µm. Lizzie Wood identified mutants that have increased variability in cell size at division in order to understand the underlying mechanism(s) affecting size control. She has identified three genes encoding, a regulator of lipid metabolism, a polyA polymerase and a sequence orphan, and is analysing how they affect cell size variability. We are also investigating how cells regulate the size of membrane bound organelles. There is a near constant nuclear-cytoplasmic ratio (N/C ratio) in fission yeast, which is not directly dependent on DNA content. Helena Cantwell is investigating whether an active mechanism corrects perturbations in N/C ratios. A pom1 deletion strain that divides asymmetrically generates cells with varied N/C ratios. Helena found that cells with low or high N/C ratios could recover to the wild type value within one cell cycle, whilst those with high N/C ratios recovered at a slower rate. Chemical biology Richard Lewis and Juan-Juan Li have identified several bacterial strains, which produce potentially new biologically important molecules affecting cell cycle progression, causing cells to become highly elongated or to misplace the nucleus and division septum. An alternative approach by Jun Funabiki, uses small molecule libraries to identify inhibitors of AAA ATPases, in collaboration with Tarun Kapoor’s Group (The Rockefeller University). Publications listed on page 167 LINCOLN'S INN FIELDS 91 PROTEIN PHOSPHORYLATION www.london-research-institute.org.uk/research/peter-parker Group Leader Peter Parker Postdoctoral Scientists Nicola Brownlow Katharina Deiss Tanya Pike Philippe Riou Frances Willenbrock Graduate Students Jeroen Claus Khalil Davis Joanna Kelly Yixiao Zhang Clinical Research Fellow Julien de Naurois Scientific Officers Veronique Calleja Jacqueline Marshall Narendra Suryavanshi Masters Student Alexander Wallroth Figure 1 PKCε loss of function is associated with PICH-positive ultrafine bridges in anaphase. HeLa cells (control or knocked down for PKCε as indicated) were grown on cover slips, fixed and stained for DNA (DAPI; blue) and the helicase PICH (green). Cells in anaphase were selected for imaging. The control cell image illustrates an apparently normal anaphase with no evidence of chromosome bridges or PICH-positive ultrafine bridges (only 25% of control cells show any aberrations of this type). In PKCε knockdown cells, there is consistent evidence of nondisjunction illustrated here by a PICH positive ultrafine bridge running between the two presumptive daughters (>65% of cells show such aberrations). Scale bar = 5μM. 92 It has long been understood that aberrations in normal cellular control hierarchies underpin the pathophysiology of cancer. The nature of individual dysfunctions and the way in which these lead to pathological properties and emergent tumour autonomous and tumour host dependencies remain areas of enormous interest and activity. Dissection of these processes in any given tumour setting provides both markers relevant to prognosis and targets for intervention. As pleiotropic cellular regulators, it is not surprising that protein kinases feature prominently as targets in the expanding portfolio of approved cancer treatments. The PKC subfamily of the AGC kinase arm of the human kinome remain central to our work, which addresses the potential of these as targets in cancer. An emergent PKCε dependency of transformed cells We have continued to dissect the underlying mechanisms associated with the cell cycle actions of PKCε that are particularly manifested in certain transformed cell models. This has led to the conclusion that PKCε is involved in a conditional pathway that triggers both a delay in the metaphase-anaphase transition and resolution of catenated DNA. The trigger for PKCε engagement is the sensing of catenation specifically in metaphase. Following DNA replication, the catenation of sister chromatids requires resolution, a process that is normally completed through S phase and G2. When cells are transiting G2, incomplete decatenation triggers a checkpoint arrest enabling elimination of catenation before completion of G2. Some transformed cells by-pass this G2 checkpoint and consequently enter metaphase faster but with DMSO Scientific report 2014 LONDON RESEARCH INSTITUTE siPKCε retained catenation. This is not compatible with effective metaphase exit/anaphase entry where there needs to be complete disjunction of sister chromatids in order to permit chromosomal segregation into daughter cells. It is this catenation-dependent delay to metaphase exit that we have shown is under the control of PKCε. Notably the earlier catenation checkpoint that operates in G2 is not influenced at all by PKCε and hence normal cells that retain their G2 checkpoint do not require the metaphase PKCεcontrolled pathway to effect segregation. However, the subset of tumour cells that have lost their G2 response to catenation are particularly dependent on the PKCε-mediated response in mitosis to prevent non-disjunction in anaphase (Figure 1). This distinct behaviour of tumour cells is a vulnerability providing an opportunity for intervention with an expectation of a high therapeutic index. When and how is PKCε engaged in cell cycle control? In the context of the cell cycle, defining when PKCε intervention might usefully be deployed, requires specific biomarkers informing on the loss of the G2 catenation checkpoint and/or defining the circumstances surrounding the engagement of the PKCε pathway. To provide insight into the key players in the G2 catenation checkpoint and so enable scrutiny of loss-of-function/expression patterns in tumour settings, we have completed a whole genome screen for gene products that when lost cause a by-pass of the checkpoint (collaboration with Dr Mike Howell, LRI). A series of ~80 genes have been identified through multiple rounds of screening and library deconvolution; we are currently pursuing these to (in)validate them as G2 checkpoint players and candidate biomarkers. In parallel, we are investigating the mechanisms involved in the triggering of PKCε action in response to metaphase catenation. This has also produced promising insights that inform on mechanism and may serve the purpose of marking mitotic catenation, and hence predict PKCε inhibitor sensitivity. Aberrant proliferation, polarity and targeting aPKCι aPKCι is an established regulator of cellular polarity and proliferation. We have defined aPKCι as a selective driver in certain oncogenic backgrounds and established models, which are highly dependent on aPKCι for both proliferation and polarity. Our studies on this multifunctional kinase have been divided between: (i) a drug development programme executed through CRT (Drs Jon Roffey and Christian Dillon) and a collaborating commercial activity, and (ii) mechanistic studies directed at understanding how mutant Ras influences aPKCι pathways and how aPKCι downstream targets define polarising and/or proliferative actions. The drug development programme has made excellent progress and now sits at a very late pre-clinical stage. Our particular input into this programme has centred upon the identification of substrates whose phosphorylation may serve as pharmacodynamic markers and/or as biomarkers for patient stratification. This has entailed the development of a substrate screening platform – a method we have termed KIPS for Kinase Proximal Substrate screening. Addressing more directly the pathways involved in promoting aberrant proliferation and polarity (including migration; collaboration with Dr Erik Sahai, LRI), we have focused on a Ras-pathway link to aPKCι and on specific partner proteins that are recruited through the ‘RIPR’ motif, a substrate recruitment surface that has been found mutated rarely but repeatedly in tumours (collaboration with Dr Neil McDonald, LRI). The change of function associated with mutations of this RIPR motif, appears to selectively disable polarising capabilities of aPKCι. How specific partner proteins contribute to aPKC-dependent polarity is currently the subject of ongoing work. Directly related to the proliferative programme in which aPKCι can also sit, we have identified aPKCι-dependent proximal functions that are sensitive to oncogenic Ras. Understanding the underlying mechanisms will prove important in relating aPKCι action with intervention in Ras-driven tumours. Emerging properties; PKNs and pseudokinases The PKN members of the PKC family remain perhaps the least well characterised. We have continued to elaborate functions for this family from a genetic perspective and have evidence that PKN is involved in regulation of various aspects of cellular migration. In parallel, we have pursued their roles in in vivo models, particularly in prostate cancer (collaboration with Dr Angus Cameron, QMUL). From a mechanistic, substrate perspective, we have also initiated studies on downstream screening employing the KIPS approach successfully deployed for aPKCι. Extrapolating our work on nucleotide-driven allostery defined in PKC family members, we have investigated how the nucleotide binding pocket of the pseudokinase HER3 is involved in signalling through heterodimerisation with HER2 and also how this influences behaviour of HER3 in the context of models of drug resistance. This has in particular addressed the underlying organisational changes associated with the receptor and its heterodimer partner as a function of inhibitor treatment (collaboration with Prof. Tony Ng, KCL and Dr Marisa Martin-Fernandez, STFC). Importantly, the evidence indicates that nucleotide binding pocket occupation has a profound effect on signalling through this inactive kinase and highlights HER3 as both an opportunity and a liability for targeted interventions. Publications listed on page 167 LINCOLN'S INN FIELDS 93 IMMUNOBIOLOGY www.london-research-institute.org.uk/research/caetano-reis-e-sousa Group Leader Caetano Reis e Sousa Postdoctoral Scientists Sophie Acton Jan Boettcher Safia Deddouche Julie Helft Jatta Houtari Pierre Maillard Barbara Schraml Naren Srinivasan Annemarthe van der Veen Paul Whitney Santiago Zelenay Graduate Students Susan Ahrens Oliver Gordon Pavel Hanč Janneke van Blijswijk Scientific Officers Aaron Farrugia Delphine Goubau Sonia Lee Neil Rogers Oliver Schulz Kathryn Snelgrove Dendritic Cells (DCs) are thought to function as antigen-presenting cells for priming T cell responses. Over the last year, we have found that DCs in fact perform a broader array of functions. We uncovered a key role for DCs in orchestrating innate resistance to systemic fungal infection and we have found that signals from DCs allow for stretching of lymph node stromal cells and permit acute lymph node expansion at the outset of an immune response. In parallel, we have continued our work on receptors used by DC and other cell types to detect viral invaders and thereby ensure host protection from infection and from viral oncogenesis. Our work adds to our understanding of the role of DCs and other cells in detecting infection and cancer and induction of responses aimed at restoring homeostasis. Cell-intrinsic detection of viruses Antiviral defence in vertebrates is orchestrated by type I interferons, cytokines that can be synthesised by all cells in response to triggering of viral sensors present in the cytosol (Goubau et al., 2013; Immunity. 38(5): 855–869). The latter include the RIG-I-like receptors (RLRs), RIG-I, LGP2, and MDA5, which detect atypical RNAs associated with viral presence. To identify RNAs that trigger MDA5 activation in infected cells, we purified and characterised LGP2/RNA complexes from cells infected with encephalomyocarditis virus (EMCV), a picornavirus detected by MDA5 and LGP2 but not RIG-I (Deddouche et al., 2014; Elife. 3:e01535). We found that those complexes were highly enriched for MDA5-stimulatory activity and for a specific sequence corresponding to the L region of the EMCV antisense RNA (Deddouche et al., 2014). Genomic deletion of the L region in EMCV generated viruses that were less potent at stimulating MDA5-dependent IFN production (Deddouche et al., 2014). Thus, the L region antisense RNA of EMCV is a key determinant of innate immunity to the virus and represents the first natural RNA isolated from virally-infected cells that activates MDA5. RIG-I and MDA5 are activated by distinct viral RNA structures. Earlier evidence from our group and others indicated that RIG-I selectively responds to RNAs bearing a triphosphate (ppp) moiety at the 5’-end (Pichlmair et al., 2006; Science. 314(5801): 997-1001, Hornung et al., 2006; Science. 314(5801): 994-997). This year we showed that RIG-I also mediates antiviral responses to RNAs bearing 94 Scientific report 2014 LONDON RESEARCH INSTITUTE 5’-diphosphates (5’pp) (Goubau et al., 2014; Nature. 514(7522): 372–375). Genomes from mammalian reoviruses with 5’pp termini, 5’pp-RNA isolated from yeast L-A virus, and base-paired 5’pp-RNAs made by in vitro transcription or chemical synthesis, the latter in collaboration with Gunther Hartmann’s group in Bonn, all bound to RIG-I and served as RIG-I agonists (Goubau et al., 2014). Furthermore, a RIG-I-dependent response to 5’pp-RNA was essential for controlling reovirus infection in cultured cells and in mice (Goubau et al., 2014). Thus, the minimal determinant for RIG-I recognition is an RNA with 5’pp. Such RNAs are found in some viruses but not in uninfected cells, indicating that recognition of 5’pp-RNA, like that of 5’ppp-RNA, acts as a means of self/non-self discrimination by the innate immune system. Syk function in DCs and innate resistance to fungal infection Earlier work from our group and others indicated that protection from fungal infection ensues from the activity of Syk-coupled C-type lectin receptors and MyD88-coupled toll-like receptors in myeloid cells, including neutrophils, macrophages and DCs (Osorio et al., 2011; Immunity. 34(5): 651-664). Given the multitude of cell types and receptors involved, elimination of a single pathway for fungal recognition in a single cell type such as DCs, primarily known for their ability to prime T cell responses, would be expected to have little effect on innate resistance to fungal infection (Osorio et al., 2011). Earlier this year we reported that this is surprisingly not the case and that selective loss of Syk but not MyD88 in DCs abrogates innate Figure 1 Paraffin embedded section of inguinal lymph node from PDGFRα-H2B-GFP mouse showing nuclei of fibroblastic reticular cells in brown and podoplanin staining in pink. Surrounding leukocytes are shown in blue. resistance to acute systemic Candida albicans infection in mice (Whitney et al., 2014; PLoS Pathog. 10(7): e1004276). In collaboration with Salomé LeibundGut-Landmann (a former lab member, now independent in Zürich), we showed that Syk expression by DCs is necessary for IL-23p19 production in response to C. albicans, which is essential to transiently induce GM-CSF secretion by NK cells that are recruited to the site of fungal replication (Whitney et al., 2014). NK cell-derivedGM-CSF in turn sustains the anti-microbial activity of neutrophils, the main candidicidal effectors (Bär et al., 2014; Immunity. 40(1): 117–127). Thus, the activity of a single kinase in a single myeloid cell type orchestrates a complex series of molecular and cellular events that underlies innate resistance to fungal sepsis. DC control of lymph node expansion Lymph nodes (LNs) are essential meeting places for T lymphocytes and DCs. T cell and DC interactions within LNs are supported by fibroblastic reticular cells (FRCs), a complex interconnected stromal cell network that produces and ensheathes dense bundles of collagen and other extracellular matrix components (Katakai et al., 2004; Int Immunol. 16(8): 1133–1142). FRC networks additionally provide key physical routes for leukocyte traffic, as well as act as a source of chemoattractants for T cells and DCs. Notably, contact with FRCs promotes chemokinesis in immigrant DCs to facilitate their migration within lymph nodes (Acton et al., 2012; Immunity. 37(2): 276-89). This is in part due to cytoskeletal changes that take place in DCs in response to signalling by the Syk-coupled C-type lectin CLEC-2 upon engagement by its ligand, the glycoprotein podoplanin (PDPN), expressed by FRCs (Acton et al., 2012). Interestingly, we had previously noticed that CLEC-2 expression by DCs is upregulated in inflammatory conditions (MourãoSá et al., 2011; Eur J Immunol. 41(10): 3040–3053). We therefore wondered whether, in addition to signalling to help promote DC movement along FRCs, CLEC-2 might also work in reverse to modulate PDPN function and alter the properties of the FRC network. In collaboration with the group of Erik Sahai (Tumour Cell Biology Group, LRI), we found that PDPN positively regulates actomyosin contractility in FRCs via activation of RhoA/C and the downstream effector Rho-kinase (Acton et al., 2014; Nature. 514(7523): 498–502). PDPN engagement by CLEC-2 causes clustering of PDPN and rapidly and robustly uncouples PDPN from RhoA/C activation, which relaxes the actomyosin cytoskeleton, permitting FRC stretching (Acton et al., 2014). Notably, we found that LN expansion upon immunisation was significantly constrained in mice lacking CLEC-2 expression in DCs but this could be reversed by administering recombinant protein (Acton et al., 2014). Thus, the same DCs that initiate immunity by presenting antigens to lymphocytes also initiate remodelling of LNs by delivering CLEC-2 to FRCs and permitting FRC network stretching. CLEC-2 modulation of PDPN signalling allows for the rapid LN expansion driven by lymphocyte influx and proliferation that is the critical hallmark of adaptive immunity. Publications listed on page 168 LINCOLN'S INN FIELDS 95 TUMOUR CELL BIOLOGY www.london-research-institute.org.uk/research/erik-sahai Group Leader Erik Sahai Postdoctoral Scientists Esther Arwert Eishu Hirata Marco Montagner Danielle Park Graduate Students Stefanie Derzsi Anna Dowbaj Nil Ege Clinical Research Fellow Alexander Kuznetsov Scientific Officers Steven Hooper Robert Jenkins 96 The Tumour Cell Biology group aims to understand how cancer cells move through the body and their interactions with other components of the tumour microenvironment. In particular, we are interested in the cross-talk between cancer cells and a type of noncancerous cells that is found in most solid tumours: cancer-associated fibroblasts (CAFs). These non-cancerous cells can promote the invasion of cancer cells both by producing soluble factors and altering the composition and structure of the extracellular matrix. By understanding the interaction of cancer cells with their environment we hope to gain information that can be used to reduce the spread of cancer and improve responses to chemotherapies. We use a range of different approaches to learn about how cancer cells might interact with the tumour microenvironment. At one end of the spectrum, Rob Jenkins has been implementing computational modelling approaches to explore how varying the properties of cancer cells and CAFs leads to different patterns of cancer invasion. Ideas from the modelling are being tested experimentally by Takuya Kato, a year-long visitor as part of an Anglo-Japanese collaborative grant. Steve Hooper, Alex Kuznetsov and Danielle Park are exploring cancer cell invasion in 3D culture systems that recreate many features of physiological tissue. Nil Ege is trying to understand how both cancer cells and CAFs might sense the physical properties of the extracellular matrix. Anna Dowbaj has recently joined our group and is attempting to use light to control cell signalling in both cancer cells and CAFs. We believe that this will be a powerful tool to understand how changes in the behaviour of either a single cell, or a small group of cells, affect neighbouring cells. Marco Montagner, Esther Arwert, and Eishu Hirata are all using a combination of in vitro and in vivo approaches to learn how communication between cancer cells and non-cancerous enables the most lifethreatening aspects of cancer. Marco is studying how cancer cells arriving at metastatic sites manage to alter the behaviour of cells at their new location and thereby facilitate the outgrowth of metastases. Esther is trying to understand how the wound healing microenvironment after tumours excision affects residual disease, either locally or in distant organs. Finally, Eishu has been using intravital imaging to investigate why targeted therapies against oncogenic kinases might fail. He has been exploring the hypothesis that CAFs may Scientific report 2014 LONDON RESEARCH INSTITUTE provide signals that are able to compensate for inhibition of the oncogenic kinases. In particular, he has implemented methods for live imaging of the activity of ERK/MAP kinase signalling in tumours before and after the administration of targeted therapies. 2014 has also seen the award of a PhD to Stefanie Derzsi for her work into heterotypic cell-cell contacts between cancer cells and CAFs, and the completion of two projects initiated by former lab members, Chris Madsen and Cerys Manning (these are described in detail below). Cancer cell migration is necessary for tumours to spread through the body. As cancer cells transit from their site of origin to other locations in the body they encounter a diverse range of environments with varying matrix composition, stiffness, and geometry. To understand how cells adapt to enable migration through changing environments we have continued our collaboration with Paul Bates (Biomolecular Modelling, LRI). This analysis has revealed that localised coordination of the actomyosin cytoskeleton and coupling of the actin network to the plasma membrane is critical for cells to adapt to changes in matrix geometry. However, the molecular mechanisms that might achieve this coordination are not well understood. We hypothesised that the migration of cells during development might provide clues to this mechanism. In collaboration with Barry Thompson’s group (Epithelial Biology Laboratory, LRI), we screened for genes that play a role in both developmental cell migration in Drosophila and cancer cells. This led us to focus on the STRIPAK complex that includes both PP2A subunits and MST3&4 kinases. The MST3&4 kinases are able to found in tumours prevent negative regulation of MST3&4. This work has led us to propose that the STRIPAK complex is a key enabler of cancer cells being able to adapt their migratory behaviour depending on their environment and thereby metastasise. Figure 1 Image shows two breast cancer cells transfected with the phosphatase regulatory subunit PPP1R14C (blue) and stained for F-actin (red) 80x80microns. (Adapted from Madsen et al., 2015; Nat Cell Biol. 17(1): 68-80) coordinate the regulation of the actomyosin network with its coupling to the plasma membrane. They achieve this by directly phosphorylating the Ezrin/Radixin/Moesin family of proteins that link the actin network to the plasma membrane and by phosphorylating the PPP1R14A-D family of PP1 regulators. The phosphorylation of PPP1R14 proteins lead to reduced dephosphorylation of MLC, and consequently increased actomyosin contractility. If this mechanism is perturbed by depleting MST3&4 then cancer cells are unable to squeeze through small gaps, although they remain competent at migration of flat surfaces. This lack of adaptability ultimately leads to reduced metastatic efficiency. Excitingly, we have also been able to show that truncations of the STRIPAK component FAM40B In addition to our interest in the mechanistic aspects of how cancer cells move, we are also interested in what makes cancer cells invasive. Previous intravital imaging studies from our group and others have shown that only a small minority of cells are motile in tumours. To learn more about these cells we introduced fluorescent reporter constructs into melanoma cells that enable us to monitor the activity of SRF and Notch signalling. Intravital imaging of tumours generated from these melanoma cells revealed that both of these signalling pathways are activated in the motile sub-population of cells. Further analysis of transcription changes associated with the activation of these pathways lead us to identify an overlapping set of genes associated with motile melanoma cells. We identified the histone trimethylase EZH2 as a critical regulator of this set of genes. Not only is EZH2 required for the expression of this genes, it is also required for the invasive behaviour of melanoma cells and their ability to form lung metastases. Publications listed on page 169 LINCOLN'S INN FIELDS 97 CANCER EPIGENETICS www.london-research-institute.org.uk/research/paola-scaffidi The Cancer Epigenetics group is interested in uncovering fundamental principles of cancer development, with particular emphasis on the role of epigenetics in determining malignant cellular features. By focussing on cancer stem cells (CSCs), the cells that fuel the long-term growth of tumours, we aim to identify novel therapeutic targets and promote the design of more effective strategies to treat the disease. Group Leader Paola Scaffidi Graduate Students Josep Monserrat Sanchez Thomas Mortimer Scientific Officer Christina Morales Torres Masters Student Tristan Henser-Brownhill Epigenetic mechanisms regulating tumour maintenance Cancer is a clonal disease originating from a single cell. Yet, most human cancers are characterised by astounding intra-tumour heterogeneity and comprise various subpopulations of cells with distinct phenotypes and biological properties. Even neighbouring cells within a tumour may have different morphologies, express differential transcriptional programs and display specific repertoires of surface molecules (Figure 1). Most importantly, not all cancer cells possess the same proliferative potential and in most cancers only a subset of cells is truly immortal. These cells act as CSCs and are responsible for maintaining the long-term growth of the tumour. We are interested in understanding how epigenetic mechanisms involving chromatin and DNA methylation contribute to intra-tumour heterogeneity and how they affect CSC function. Findings over the past ten years have strongly implicated deregulation of epigenetic instructions in cancer. Epigenetic alterations in chromatin and DNA methylation are universal features of neoplasia and recurrent mutations in proteins involved in epigenetic control are increasingly identified in various cancers. Although comprehensive epigenetic profiles of various cancer types can now be generated, discriminating Figure 1 Phenotypic intratumour heterogeneity. Immunofluoresce microscopy of a breast cancer section showing highly heterogeneous patterns of methylated histone H3 (green) and histone H1.0 (red). 98 Scientific report 2014 LONDON RESEARCH INSTITUTE between ‘driver’ epigenetic alterations, which play critical roles in cancer development, and ‘passenger’ epigenetic changes, which simply occur as consequence of altered cell function, remains often challenging. This is partially due to the fact that traditional bulk approaches analyse tumours in their entirety and disregard intratumour heterogeneity. CSCs form in vivo heterogeneous and hierarchically-organised tumours in which only a small subset of cells has unlimited proliferative potential and maintains tumour growth. The molecular basis underlying functionally distinct subpopulations of cells in a tumour are at present unclear. Since CSCs generate their nontumourigenic progeny through a differentiation process, epigenetic mechanisms are likely to play a critical role in defining the malignant phenotype of a cancer cell within a growing tumour (Figure 2). We are employing genome-wide mapping approaches (ChIP-Seq and DHS-seq) to characterise the chromatin landscape of CSCs and identify epigenetic features, which distinguish them from the rest of the tumour. By combining these studies with in vivo gain- and loss-of function experiments, we aim to identify epigenetic features that are critical for CSC function and can be modulated for therapeutic purposes. Using these approaches we have recently identified the differentiation-related histone variant H1.0 as an important regulator of CSC function. We have found that H1.0 is downregulated in a variety of cancers, including glioblastoma, breast cancer, kidney cancer and melanoma, and that its downregulation is required for CSC self-renewal. H1.0 loss in CSCs leads to genome-wide activation of oncogenic transcriptional programs by altering the higher-order structure of chromatin and inducing coordinated changes in gene expression in large chromosomal domains. We are currently trying to understand how we can modulate H1.0 expression, in order to inhibit CSC self-renewal and drive their terminal differentiation. Figure 2 Epigenetic and functional heterogeneity of cancer cells. Tumours comprise functionally distinct subpopulations of cells. Histone modifications, DNA methylation, and higher-order chromatin structure define the epigenetic status of CSCs and contribute to the maintenance of CSC properties. The bulk of the tumour arises through differentiation of CSCs into heterogeneous cell types, characterised by various epigenetic states that activate or silence distinct sets of genes. Changes in the epigenetic landscape occur during differentiation and are associated with loss of self-renewal and tumourigenicity. Extracellular signalling and cancer cell plasticity What drives epigenetic heterogeneity within tumours? While many epigenetic abnormalities in cancer are the consequence of mutations, some epigenetic changes are reversible and, as such, cannot be the result of genetic changes. Interactions of cancer cells with the tumour microenvironment strongly affect cancer development. These interactions are based on extracellular signalling and they are likely to result in epigenetic changes that affect the differentiation status of cancer cells and, as a consequence, their proliferative potential. We are developing novel microscopy-based tools, which will allow assessment of the epigenetic status of CSCs and their plasticity in response to extracellular signalling in intact tumours. These tools will also allow visualisation of CSCs in living animals and the analysis of CSC dynamics. It is at present unclear whether tumour-maintaining cells are a stable subpopulation of cells within tumours or whether cells can fluctuate between a CSC and a non-CSC state as a result of reversible epigenetic changes. This has major implications for the design of targeted therapeutic strategies. We aim to understand how stable the CSC phenotype is and whether it can be modulated by interfering with chromatin-based mechanisms. Oncogenic reprogramming and CSC formation In many cancers, CSCs arise through reprogramming of committed cells that lose their cellular identity and acquire self-renewal ability. Thus, alterations in proteins involved in maintenance of epigenetic memory are likely players in the genesis of CSCs. Indeed, we have recently identified a tumour-suppressive mechanism that utilises the chromatin-binding protein BRD4 to inhibit oncogenic reprogramming of committed cells into CSCs. We have discovered that BRD4, a histone acetylation ‘reader’, maintains a gene expression programme, which protects normal cells from de-differentiation in response to oncogenic stimuli. We found evidence for this tumour protective mechanism in lung and breast tissues. Interestingly, the same protein is critically important for disease maintenance in various haematological cancers, suggesting that BRD4 can exert a tumour-protective or a tumourpromoting function, depending on the cellular context (Fernandez et al., 2014; Cell Rep. 9(1): 248-260). Extending these studies, we are currently performing unbiased, CRISPR/Cas9based, loss-of-function screens targeting proteins involved in the establishment and recognition of DNA methylation patterns, writer, readers and erasers of chromatin marks, chromatin remodellers and proteins controlling the highorder structure of chromatin, in order to identify other epigenetic regulators which either prevent (tumour suppressors) or promote (therapeutic targets) the appearance of CSCs. Publications listed on page 169 LINCOLN'S INN FIELDS 99 MACROMOLECULAR STRUCTURE AND FUNCTION www.london-research-institute.org.uk/research/martin-singleton Group Leader Martin R Singleton Postdoctoral Scientists William Chao Stephan Lefevre Silva Zakian Graduate Students Vera Leber Thibaud Perriches Ben Wade Scientific Officer Xiao Hu Our group studies the molecular mechanisms of eukaryotic chromosome segregation, with a particular focus on the macromolecular complexes involved in attaching replicated sister chromatids to each other and to spindle microtubules. We are interested in determining the atomic structures of these complexes, and understanding how their overall architecture and function aids the cell in bringing about the rapid and accurate dissemination of the genome. We employ a combination of biophysical and biochemical techniques to address these questions, primarily those of X-ray crystallography for high-resolution studies and electron microscopy to analyse larger complexes. Regulation of kinetochore formation Kinetochores are structures that attach replicated sister chromatids to spindle poles via microtubules. There is considerable complexity involved in the formation of an attachment that is both strong enough to carry spindle forces, yet at the same time remain bound to the dynamically unstable microtubule. In addition, the kinetochore must allow for rapid dissolution of erroneous attachments that might result in chromosome missegregation. These requirements have lead to the evolution of a large, modular structure, with distinct functionality associated with each collection of sub-complexes. Two key elements are the so-called CCAN (constitutive centromere associated network) and the KMN (Knl1, Mis12, Ndc80) network (Figure 1a), which interact with centromeric chromatin and spindle microtubules respectively. The kinetochore does not remain attached to the underlying chromatin throughout the cell cycle, rather it appears to be assembled in a step-wise fashion after DNA replication, with the exact timing of events varying between organisms. The mechanisms controlling this assembly process have yet to be fully understood, but perhaps unsurprisingly, reversible phosphorylation of multiple kinetochore proteins are thought to be involved. In collaboration with David Glover’s group in the Department of Genetics, Cambridge, we have been characterising a novel phosphatase-kinetochore interaction. A proteomic analysis of centromereassociated factors in Drosophila revealed that the targeting subunit of the protein phosphatase 4 (PP4) complex, known as Falafel (Flfl) co-purifies with the CCAN constituent CENP-C. Unlike the 100 Scientific report 2014 LONDON RESEARCH INSTITUTE better-studied protein phosphatase 2A (PP2A), little is known about PP4 targets and activity. Like PP2A, it is a multi-subunit enzyme, comprising a catalytic subunit (which is extremely similar to the PP2A equivalent) and two other variable subunits, one of which, R3 is presumed to be the regulatory subunit, responsible for substrate recognition. Falafel is the Drosophila homolog of the human R3 protein, and the interaction with CENP-C provides a suitable model to better understand PP4 activity. The Glover group has demonstrated that a direct interaction between CENP-C and Falafel exists, and that the catalytic activity of PP4 is required for correct mitotic progression. Depletion or inactivation of the enzyme results in mislocalisation of CENP-C as well as outer kinetochore components at metaphase. Studies using truncation mutants demonstrated that the N-terminal of Falafel is required for the interaction with CENP-C. Using peptide array analysis (provided by the LRI Peptide Chemistry Facility) we were able to map the exact binding site on CENP-C, and define a minimal construct of Falafel required for the interaction. This corresponds to the N-terminal domain that had been previously predicted to adopt a PH-like fold. We solved the structure of this domain bound to the cognate peptide from CENP-C at 1.5Å resolution (Figure 1b). Structural homology analysis showed that the fold is closely related to the EVH1 domain, as typified by the Ena/ Vasp family of proteins. These usually bind a proline-rich sequence in a left-handed helical (PPII) configuration. However, in our structure, the recognition motif is FKKP, with the phenylalanine and proline making the key conserved contacts Figure 1 A. Schematic diagram of the kinetochore-centromere interface showing the two main conserved complexes, the CCAN and KMN network. B. Structure of the Falafel N-terminal domain bound with the target peptide from CENP-C. The critical interacting phenylalanine and proline residues are depicted in green. A B with Falafel. Despite this sequence variance, the peptide still adopts a classic PPII helical form, and provides the first insights into the molecular basis of substrate recognition by the PP4 family of enzymes. We believe that recruitment of PP4 family phosphatases to the inner centromere is likely to a conserved phenomenon, and it will be interesting to delineate how the process occurs in other eukaryotes. Sister chromatid cohesion and DNA replication The establishment of persistent sister chromatid cohesion requires the action of replication fork associated proteins, presumably to ensure that inter-sister rather than intra-chromatid linkages are maintained. A key reaction is the acetylation of the Smc3 subunit of cohesin by the acetyltransferase Eco1, which is thought to antagonise cohesin destabilisation by Wapl. In addition to this pathway, a number of other proteins are thought to contribute to cohesion establishment in both Eco1-depedent and independent manners. We have recently started to carry out structural and biochemical studies on some of these proteins, in order to understand their recruitment and function at the replication fork, and how these activities impinge on the process of sister chromatid separation. In addition, Eco1 has been proposed to play a role in double strand break repair via a replication-independent pathway, and it will be of great interest to understand how differential targeting of the enzyme is achieved. Publications listed on page 169 LINCOLN'S INN FIELDS 101 MICROTUBULE CYTOSKELETON www.london-research-institute.org.uk/research/thomas-surrey Group Leader Thomas Surrey Postdoctoral Scientists Jayant Asthana Todd Fallesen Franck Fourniol Johanna Roostalu Einat Schnur Graduate Students Hella Baumann Tanja Consolati Jonathon Hannabuss Rupam Jha Scientific Officers Nicholas Cade Christian Duelberg Iris Lueke Claire Thomas Masters Student Stefan Keller Microtubules are polymers with a variety of essential functions in all eukaryotic cells. The microtubule cytoskeleton forms a scaffold for the internal organisation of cells, it provides tracks for molecular motor transport and - during cell division - it forms the mitotic spindle that segregates the chromosomes. Our research aims at a better understanding of the molecular mechanisms that govern how key proteins regulate cytoskeleton organisation, dynamics and function, which is of crucial importance to maintain cells in a healthy state. To reach this goal, we study the proteins that control microtubule dynamics that give localised functionality, and that organise microtubules in space. We use biochemistry and cell biology approaches, in combination with advanced fluorescence microscopy. Local nanoscale conformational transitions at the ends of growing microtubules The dynamic properties of microtubules depend on complex structural rearrangements in their end regions. Members of the EB1 protein family interact autonomously with microtubule end regions and recruit several other proteins to localise their activities to specific sites in the cell. The autonomous end binding property of fluorescently labelled EB1 proteins can also be used to monitor nanoscale conformational transitions in microtubule end regions by fluorescence microscopy (Maurer et al., 2014; Curr Biol. 24(4): 372-84). Such analysis revealed two consecutive conformational transitions that freshly microtubule-incorporated tubulins undergo as microtubules grow. This shows how growing microtubule ends mature at the nanoscale. Further analysis revealed an important role of the detected conformational states for the control of the stability of microtubules. These results advance our understanding of the molecular mechanism that controls the fundamental characteristics of microtubule dynamics. A hierarchical protein interaction network controls microtubule end tracking of dynein Microtubule ends are hubs of protein activities, because a variety of proteins accumulate there in order to regulate microtubule dynamics and facilitate interactions of microtubules with cellular targets. Another function of end accumulation is motor protein loading and transport initiation. The major minus-end directed motor, dynein, is recruited to microtubule ends by several proteins; this is needed to initiate cargo transport along the 102 Scientific report 2014 LONDON RESEARCH INSTITUTE microtubule away from its end, typically towards the cell centre or - in mitosis - to the spindle poles. These dynein recruiting proteins form dynamic interaction networks, whose behaviour depends on a number of potentially competitive interaction modes. The rules that determine which of the various proteins are recruited to the limited number of available binding sites at microtubule ends remain elusive. In collaboration with the laboratory of Michel Steinmetz, PSI Villigen, Switzerland, we have examined how human dynein is targeted to growing microtubule ends in the presence of competing proteins (Duellberg et al., 2014; Nature Cell Biol. 16(8): 804-11). Using in vitro reconstitutions and fluorescence microscopy, we found that a hierarchical recruitment mode is needed to overcome competition. These results highlight how the connectivity and hierarchy within a localised protein interaction network is orchestrated. Motor-mediated microtubule organisation in lipid-monolayered micro-compartments The correct spatial organisation of microtubules is important for the establishment and maintenance of the internal architecture of eukaryotic cells. Microtubules are arranged in space by a multitude of biochemical activities and by spatial constraints imposed by the cell boundary. The principles governing the generation of distinct intracellular architectures are only poorly understood. We have studied the consequences of spatial confinement on the self-organisation of purified microtubules and motor proteins that were encapsulated in lipid-monolayered droplets in oil, using in vitro reconstitutions Figure 1 Microtubule end tracking of a dynactin and dynein. A fluorescence microscopy image sequence (left) and a kymograph (space-time plot, middle) of a dynamic microtubule (red) with the dynactin component p150 Glued (green) is shown. This is part of the hierarchical protein interaction network that recruits dynein to growing microtubule plus-ends (schematic, right). (Modified from Duellberg et al., 2014). (Baumann et al., 2014; J Biol Chem. 289(32): 22524-35). We varied the diameter of these micro-containers from five to a hundred micrometres, which covers the size range of typical cell bodies. We found that container size had a major organising influence. The presence of a microtubule-crosslinking motor protein decreased the number of accessible types of microtubule organisations. Depending on the degree of spatial confinement, the presence of the motor caused either the formation of a cortical array of bent microtubule bundles or the generation of single microtubule asters inside the droplets. These are two of the most prominent forms of microtubule arrangements in plant and metazoan cells. These results provide insight into how the combined organising influence of spatial constraints and crosslinking motor activities determines distinct microtubule architectures in a minimal biomimetic system. Publications listed on page 169 Figure 2 Self-organisation of motors and microtubules in microcompartments. Microtubule-crosslinking motors can organise microtubules either into astral (top) or cortical (bottom arrays) inside lipidmonolayered droplets. Scale bars: 20 μm. (Modified from Baumann et al., 2014). LINCOLN'S INN FIELDS 103 TRANSLATIONAL CANCER THERAPEUTICS www.london-research-institute.org.uk/research/charles-swanton The Translational Cancer Therapeutics group focuses on mechanisms generating cancer genetic diversity and its consequences on clinical outcome. Our group and others, through next generation sequencing studies have demonstrated that the principles of Darwinian evolution apply to the growth and adaptation of human tumours (Gerlinger et al., 2012; N Engl J Med. 366(10): 883-892, Gerlinger et al., 2014; Nat Genet. 46(3): 225-233, Nowell PC, 1976; Science. 194(4260): 23-28). Group Leader Charles Swanton Clinical Scientists Crispin Hile Mark Stares Samra Turajilic Postdoctoral Scientists Nicolai Birkbak Sebastijan Hobor Laurent L’Epicier-Sansregret Carlos Lopez-Garcia Carolina Navas Gareth Wilson Panos Zalmas Graduate Students Andrew Crockford Sally Dewhurst Nicholas McGranahan Tom Watkins Tom Webber Scientific Officers Eva Gronroos Andrew Rowan The causes and consequences of cancer diversity Our group has demonstrated that intratumour heterogeneity, through tumour sampling bias, impacts upon our ability to successfully qualify cancer biomarkers for clinical use (Gulati et al., 2014; Eur Urol. 66:936-48). We have also found evidence for extensive parallel evolution in human tumours, with multiple spatially separated subclones acquiring distinct mutations in the same gene, protein complex or signal transduction pathway, suggesting profound constraints to tumour evolution that might be exploited for therapeutic benefit (Gerlinger et al., 2012; Gerlinger et al., 2014) (Figure 1). Finally, we are building on recent findings from our group that DNA replication stress (Burrel et al., 2013; Nature. 494(7438): 492-496) and genome doubling events (Figure 2) (Dewhurst et al., 2014; Cancer Discov. 4(2): 175-185) appear to contribute to chromosomal instability and accelerate cancer evolution in order to develop deeper insight into how patterns of cancer diversity may be limited for patient benefit (Burrell et al., 2013; Nature. 501(7467): 338-345). Through the integration of our work with the UCL Cancer Trials Centre and UCL Cancer Institute, we are recruiting into the TRACERx study to decipher evolutionary processes in non-small cell lung cancer, and the DARWIN (Deciphering Anti Tumour response With INtratumour heterogeneity) trial program aimed at targeting clonally dominant driver events and deciphering how branched heterogeneous driver events contribute to drug resistance and treatment failure (Yap et al., 2012; Sci Transl Med. 4(127): 127ps10). TRAcking Cancer Evolution through Therapy/Rx (TRACERx) Clinical Study Our group, together with the UCL Cancer Trials Centre has initiated the TRACERx 842 patient clinical study that aims to decipher tumour evolutionary trajectories in early non-small cell 104 Scientific report 2014 LONDON RESEARCH INSTITUTE lung cancer. Through multi-region sequencing analysis of primary tumours and recurrent metastatic biopsies we will attempt to address the origins of the lethal tumour subclone, distinguish the changing patterns of tumour evolution over time, the associations of intratumour heterogeneity with disease outcome and the host immune response, and the impact of cancer upon the emergent subclonal genetic landscape at relapse. Through computational approaches we hope to identify epistatic relationships that dictate constraints to tumour evolution that might be exploitable for therapeutic benefit. Tumour genomic instability mechanisms are spatially and temporally heterogeneous Our early data from the TRACERx study has revealed that mutational processes shaping the lung cancer genome are dynamic over both time and space in early stage non-small cell lung cancer. Firstly, our work has shown that even in early stage lung cancer, multi-region sequencing reveals that genetic driver events may be missed through the analysis of a single biopsy. Secondly, we find evidence for an illusion of clonality, where genetic events within one biopsy may appear fully clonal, that are subsequently revealed to be branched heterogeneous events upon deeper genomic analysis of spatially separated tumour regions. Thirdly, we have found evidence that APOBEC mutational processes appear to be enriched at later time points following branched evolution of lung adenocarcinomas, even in current smokers. APOBEC mutational processes result in mutations in key driver genes that appear to generate the substrate of genetic diversity subject to further evolutionary selection and subclade expansion. Finally we have found early evidence suggesting that lung cancer evolutionary histories are prolonged with long latency periods between the development of mutations in key driver genes and clinical presentation, providing added support for lung cancer screening strategies. Figure 1 Multi-region exome sequencing of a clear cell renal cancer reveals evidence of parallel evolution with distinct somatic mutations affecting different members of the SWI/SNF complex in different regions of the primary tumour (Gerlinger et al., 2014). Parallel evolution of cancer subclones Our work in clear cell renal cell carcinoma of the kidney has found evidence for parallel evolution of tumour subclones (Figure 1). We find multiple spatially distinct mutations in SETD2, KDM5C, BAP1, PBRM1, PTEN and PIK3CA occurring within distinct spatially separated sites of the same tumour. We also find evidence that components of the same protein complex, SWI/SNF, may be subject to mutations in different regions of the same tumour (Figure 1). Similarly, we find evidence from an analysis of independent tumours within the same patient, developing from a germline VHL mutant background, converge upon activation of the mTOR signal transduction pathway, suggesting that despite the semblance of genomic chaos within these tumours, there are major constraints to the evolutionary histories of solid tumours (Fisher et al., 2014; Genome Biol. 15(8): 433). Our work continues to attempt to decipher such evolutionary constraints in solid tumours through longitudinal analysis of cancer genomes. Figure 2 Chromosomal Instability Index for diploid and genome doubled colorectal cancer cell line clones over time. Weeks in culture is indicated in the key. Over time diploid clones remain chromosomally stable, whereas genome doubled clones have higher chromosomal instability indices, which tend to increase over time. Genome doubled clones also have scores higher than 0.2 (dotted line) a threshold which in colorectal cancer patients separates highly chromosomal unstable anueploid tumours from diploid chromosomally stable tumours (Dewhurst et al., 2014). Identifying drivers of cancer diversity and cancer evolution We have recently found that cellular survival following a genome doubling event in colorectal cancer results in the tolerance and propagation of chromosomal instability in subsequent daughter cells. This results in accelerated cancer genomic evolution with tetraploid cells developing extensive chromosomal instability in contrast to their diploid progenitors which remain genomically stable throughout the 18 month time course of the experiment (Figure 2) (Dewhurst et al., 2014). Intriguingly, as well as becoming increasingly chromosomal unstable, genome doubled cells also drift towards triploid, similar to observations in colorectal cancers in vivo. Consistent with this observation, genome doubled/tetraploid early stage colorectal cancers have a significantly worse disease-free survival outcome than their diploid counterparts in multivariate analysis. Our current work is investigating the cellular mechanisms that lead to tolerance of an aneuploid or tetraploid genomic state. Our work has recently implicated loss of chromosome 18q in the initiation of chromosomal instability through loss of three CIN suppressor genes, MEX3C, ZNF516 and PIGN. Loss of each one of these genes generates both structural and numerical chromosomal instability and intercellular heterogeneity, a substrate for cancer evolution (Burrell et al., 2013). These data implicate replication stress in the generation of structural and numerical CIN and intratumour heterogeneity, and provide evidence that combining tumour bioinformatics approaches with intricate functional genomics analysis can reveal novel mechanisms contributing to intratumour heterogeneity. The TCT group is building on the experimental frameworks established through this approach to identify mechanisms generating genomic instability and addressing whether replication stress is a common contributor to chromosomal instability in other tumour types. Publications listed on page 170 LINCOLN'S INN FIELDS 105 APOPTOSIS AND PROLIFERATION CONTROL www.london-research-institute.org.uk/research/nicolas-tapon Group Leader Nicolas Tapon Postdoctoral Scientists Billel Benmimoun Teresa Bertra John Davis Ieva Gailite Graduate Students Anna Ainslie Jennifer Banerjee Nicola Brindle Yanxiang Zhou Scientific Officers Birgit Aerne Maxine Holder Masters Student Annabel Ebbing Our work is aimed at understanding how tissue size is specified during development, which remains one of the most challenging questions in biology. In order to achieve consistent organ and body size in individuals of the same species, cell growth and cell number must be tightly controlled, not only during development, but also to prevent tumour formation during adult homeostasis. Genetic screens in Drosophila have identified the Hippo (Hpo) pathway as a determinant of tissue size control. The Hpo pathway restricts tissue and organ size by both inhibiting cell proliferation and promoting apoptosis. Subsequent studies in mammals have shown that this growth control function is conserved and that Hpo signalling is dysregulated in many types of cancer. The core of the Hpo pathway is a kinase cascade comprising the Ste20-related kinase Hpo and the Dbf2-related kinase Warts (Wts). Upon Hpo activation, the downstream kinase Wts phosphorylates and inhibits the pro-growth transcriptional co-activator Yorkie (Yki). Hpo signalling has been proposed to sense various local cues relating to cell density (contact inhibition of growth and mechanical tissue properties), patterning (morphogen gradients) or nutrition, and translate these cues into a growth arrest signal once an individual tissue has reached its appropriate size. Several lines of evidence suggest that Hpo pathway activity is tightly coupled to epithelial architecture (Genevet et al., 2011; Biochem J. 436(2): 213-224). Firstly, Yki/YAP transcriptional activity has been shown to depend on the structure of the actin cytoskeleton, with F-actin promoting YAP nuclear translocation, though the precise mechanisms and the involvement of the core kinase cascade in this process remain unclear. Secondly, the basolateral polarity proteins Scribble and Lethal(2) giant larvae (Lgl) have been shown to promote Hpo pathway activity. Thirdly, the adherence junction protein α-catenin has been proposed to function as a membrane tether for YAP in keratinocytes. Finally, the apical protein Crumbs (Crb) antagonises Yki/ YAP activity, both in Drosophila and mammals. Crb is a transmembrane protein that contains multiple EGF repeats in its large extracellular domain. Crb is a key apical polarity determinant that recruits other polarity proteins through its short 37 amino acid intracellular domain. Beside its 106 Scientific report 2014 LONDON RESEARCH INSTITUTE well-documented role in polarity, Crb is also required for normal growth control, since loss of Crb function leads to tissue overgrowth. This has been ascribed to a role in both Notch and Hpo signalling. The function of Crb in Hpo signalling is thought to involve the recruitment of the FERM domain protein Expanded (Ex) to the apical membrane. Indeed, the FERM domain of Ex can bind the Crb FBM in vitro. Once apically localised, Ex forms a complex with the scaffold proteins Kibra and Merlin (Mer), which promotes inhibitory phosphorylation of Yki by Wts. In addition, Ex is thought to act as an apical tether for Yki by binding the Yki WW domains through its Pro-Pro-X-Tyr (PY) motifs. In agreement with a proposed role for Crb as a transmembrane receptor for the Hpo pathway, loss of crb promotes expression of Yki target genes. However, paradoxically, overexpression of the intracellular domain of Crb (Crbintra) leads to strong tissue overgrowth and Yki-target gene derepression. Although this could be due to a dominant-negative effect, it is important to note that Crbintra overexpression leads to loss of apical Ex in developing wings and eyes, while co-expression of Crbintra and Ex in cell culture leads to Ex phosphorylation and reduced expression. We recently reconciled these findings by showing that Crb is not only required for Ex tethering at the apical membrane but also to promote its degradation via the SCFSlimb/β-TrCP E3 ubiquitin ligase (Ribeiro et al., 2014; PNAS. 111(19): E1980-9). Indeed, immediately downstream of its FERM domain, Ex contains a sequence that conforms Figure 1 Depletion of slmb using two different dsRNAs leads to a stabilisation of Ex in the presence of Crb. Compare lane 1 with lanes 3 and 4. Expression of Ex with a Crb form lacking the FERM binding domain does not affect Ex stability. Western blot from S2 cell lysates expressing the indicated constructs. to the D/S/TSGφXS consensus sequence for canonical Slimb (Slmb) targets, which is conserved in Ex orthologues from arthropod species but absent from related FERM domain proteins such as Moe and Mer. In addition, Slmb depletion prevents Crbintra-induced Ex degradation in cell culture (Figure 1), while loss of Slmb increases Ex levels in vivo (Figure 2). Thus, in crb mutants, Ex no longer reaches the apical membrane, and is protected from degradation in the cytoplasm, where it accumulates but is presumably unable to repress Yki. When Crbintra is overexpressed, Ex turnover at the membrane (or in an endocytic compartment if Ex degradation occurs after Crb internalisation) is accelerated, leading to its depletion and consequent Yki activation. Therefore, in both cases, the outcome is Yki de-repression, albeit for different reasons. Our work indicates that Crb fulfils a dual function in Hpo signalling, both recruiting Ex apically to repress Yki activity and promoting its turnover through phosphorylation and Slmb-dependent degradation. This mechanism would ensure constant turnover of Ex at the apical membrane, allowing Yki activity to rapidly respond to changing environmental conditions. This dynamic equilibrium could be particularly important to promote fast tissue regeneration upon injury. Publications listed on page 171 Figure 2 Elevated Ex and Ci levels in hsFLP/ FRT generated slmb mutant clones in the Drosophila wing imaginal disc. Confocal micrographs of a wing imaginal disc stained with anti-Ex (A) and anti-Ci155 (B). The mutant tissue is negative for GFP (C). Merged image is shown in (D). Scale bar = 10 μm. LINCOLN'S INN FIELDS 107 EPITHELIAL BIOLOGY www.london-research-institute.org.uk/research/barry-thompson Group Leader Barry Thompson Postdoctoral Scientists Mario Aguilar Ahmed Elbediwy Graduate Students Graham Bell Mariana Campos Ichha Khanal Zoe Vincent Scientific Officer Georgina Fletcher Robert Ray We are interested in the biology of epithelial cells, the cell-type of origin for most human cancers. Epithelial cells form tissue layers and tubes by connecting with neighbours via adherens junctions. In tumours, epithelial tissue structure becomes disrupted, enabling groups of cells to become invasive. We are particularly interested in how the behaviour of epithelial cells is normally controlled, how they become polarised, and how they know to divide within the plane of the epithelium. Recent work from a PhD student, Graham Bell, has addressed how the mitotic spindle is oriented within the plane of the epithelium. Normally, spindles align with cell-cell junctions so that the two daughter cells will remain within the epithelium. Misorientation of the mitotic spindle is thought to be one possible mechanism by which epithelial cells can escape the epithelium to form invasive tumours. Graham has discovered a new role for the Lethal Giant Larvae (Lgl) protein in spindle orientation in the fruit fly Drosophila. Lgl is normally found at the lateral and basal sides of the cell, where it overlaps with a septate junction protein called Discs-Large (Dlg). Graham observed that in mitosis, Lgl Figure 1 Aurora kinases phosphorylate Lgl to induce mitotic spindle orientation in Drosophila. 108 Scientific report 2014 LONDON RESEARCH INSTITUTE relocalises to the cytoplasm. He showed that this change is caused by activation of the Aurora A and B kinases, which can directly phosphorylate the Lgl protein to disrupt its association with the plasma membrane. This loss of Lgl from the membrane then allows other proteins, such as the mitotic spindle orientation factor Pins (called LGN in mice and GPSM in humans), to bind to Dlg and thus align the mitotic spindle with septate junctions. When Aurora-mediated phosphorylation of Lgl was blocked by specific mutation of the phosphosites in Lgl, the entire process of mitotic spindle orientation was completely disrupted. As a result, spindles oriented randomly at mitosis instead of adopting their normal orientation within the plane Figure 2 Planar polarisation of the atypical myosin Dachs around a clone of cells expressing Fat. FbxL7 and Dachs localise to opposite sides of the cell. of the epithelium. Since both Lgl and Dlg are tumour suppressors in Drosophila, it could be that misorientation of the mitotic spindle is a possible tumour-initiating event. Graham is now exploring this possibility in more detail. Another PhD student in the lab, Mariana Campos, has examined how the mitotic spindle, once oriented in the plane of the epithelium, becomes further oriented in a planar-polarised fashion. Planar polarity is an asymmetry that appears at cell-cell junctions and is a form of polarity that is orthogonal to the normal apical-basal axis of epithelial cells. Mariana has identified a novel protein called FbxL7 that is essential to regulate this process. She has shown that FbxL7 acts as a ubiquitin ligase to control the planar polarisation of the Dachsous-Fat-Dachs system in Drosophila. The key effector of this system is the Dachs protein, which encodes an atypical myosin that localises to the distal side of cell-cell junctions. Mariana has found that FbxL7 localises to the proximal side of junctions, opposite to Dachs. In the absence of FbxL7, Dachs accumulates abnormally all around cell junctions, indicating that FbxL7 normally acts to remove Dachs from the proximal side of the cell. When FbxL7 is overexpressed, Dachs is degraded everywhere. Either loss or gain-of-function of FbxL7 is therefore associated with misorganisation of spindle orientation and tissue shape. In addition, tissue size is affected due to a role for Dachs in regulating the Hippo signalling pathway. Thus, FbxL7 is a novel regulator of planar polarity and tissue growth. Publications listed on page 172 LINCOLN'S INN FIELDS 109 CELL REGULATION www.london-research-institute.org.uk/research/takashi-toda Group Leader Takashi Toda Postdoctoral Scientists Takayuki Koyano Yuzy Matsuo Akiko Nishi Graduate Students Aldona Chmielewska Corinne Pinder Ngang Heok Tang Scientific Officer Hirohisa Masuda Masters Student Agathe Morand Visiting Scientist Masashi Yukawa Impeccable chromosome segregation during mitosis underlies genome stability and integrity. Any errors in this process would result in miscarriage, birth defects and/or aneuploidy, the hallmark of human cancers. Segregating each sister chromatid towards opposite poles is implemented by the mitotic spindle, a dynamic ensemble of microtubules, microtubule-associated proteins (MAPs) and motor proteins. Nucleation of microtubules in vivo does not occur spontaneously; instead specialised structures called microtubule organising centres (MTOCs) are required, in which the minus-end of microtubules is embedded. In animal cells, the centrosome comprises a major MTOC, whilst in fungi the spindle pole body (SPB) plays an analogous role. Our group has been uncovering the principles of microtubule structure, function and regulation using the genetically amenable model system, fission yeast (Schizosaccharomyces pombe). More recently we have been using zebrafish and human culture cells to scrutinise the evolutionary conservation of our findings obtained from work in fission yeast. The long-term goal of our research is simple: to understand the molecular mechanisms of how the mitotic spindle ensures faithful chromosome segregation. During 2014, we have made seminal progress in the following two areas: First, we have uncovered the conserved molecular mechanism by which the minus end of microtubules is tethered to the centrosome and further identified the physiological defects when this process is perturbed. Second, we have determined the molecular pathway leading to the recruitment of the conserved MAP complex, Alp7/ TACC-Alp14/TOG, to the SPB upon mitotic entry and shown that this process is critical for mitotic spindle assembly. The conserved Msd1 protein family plays a ubiquitous role in the anchoring of the microtubule to the centrosome The centrosome plays multi-layered roles in both yeasts and vertebrates. These include the canonical role in microtubule nucleation as the MTOC, cell cycle transition as a structural hub integrating cell cycle regulators and ciliogenesis as the basal body. In addition, there is at least one more critical role, i.e. anchoring of the microtubule minus-end. This mechanism ensures that interphase microtubule arrays emanate from the centrosome and that structure and orientation of 110 Scientific report 2014 LONDON RESEARCH INSTITUTE mitotic spindles are safeguarded. However, our knowledge of microtubule anchoring to this organelle remains surprisingly limited at the molecular level. We previously showed that in fission yeast, a mitosis-specific SPB component, mitotic spindle disanchored 1 (Msd1), is required for anchoring the minus-end of spindle microtubules to the SPB (Toya et al., 2007; Nat Cell Biol. 9(6): 646-653). Now, we have identified the human and zebrafish Msd1 orthologues (Figure 1A) and characterised their roles in microtubule anchoring. We have unveiled that the human Msd1 (hMsd1) protein is delivered to the centrosome in a centriolar satellite- and dynein-dependent manner, wherein it physically binds the microtubule-nucleating γ-tubulin complex (Figure 1B). siRNA-mediated hMsd1 knockdown results in disorganised interphase microtubules due to the inability of the microtubule to be tethered to the centrosome (Figure 1C). During mitosis, mitotic spindles become abnormally tilted and misoriented, which is attributed to the release of astral microtubules from the centrosome, leading to a faulty interaction between microtubules and the cell cortex (Figure 1C). Moreover, we have found that in both humans and zebrafish, the Msd1 orthologues are critical for ciliogenesis. Remarkably, the loss of Msd1 leads zebrafish embryos to left-right asymmetry defects (Hori et al., 2014; EMBO Rep. 15(2): 175-184). Collectively, the Msd1 proteins are the first molecules to be identified as the conserved microtubule-anchoring factors. Figure 1 The Msd1 family comprises the conserved microtubuleanchoring factors. A. Phylogenetic dendrogram showing the evolutionary relationship among Msd1 orthologues. Msd1 orthologues in human, mouse and Aspergillus nidulans are also called, SSX2IP, ADIP and TINA respectively. B. A model of hMsd1-mediated microtubule anchoring to the centrosome. hMsd1 (red) is transported to the centrosome accompanied by centriolar satellites (C.S., light blue) and the dynein motor (grey) along microtubules (thick pink lines). Upon delivery to the pericentriolar region, hMsd1 interacts with the γ-tubulin complex (not shown), thereby directly anchoring microtubule minus-ends to the pericentriolar material. Paired centrioles each containing procentrioles are shown in green. C. Disorganised microtubule morphologies upon hMsd1 depletion in U2OS cells. Microtubule structures during interphase (far-left panels) and mitosis (second panels) in control (top row) and hMsd1-depleted cells (bottom row) are shown. Schematic microtubule structures showing spindle (mis)orientation in relation to the substratum (coverslip) are depicted on the right-hand two panels (top and side views). Note that spindle microtubules are rotated randomly upon hMsd1 depletion, as astral microtubules become unstable, by which they cannot interact with the cell cortex to maintain horizontal spindle positioning. Scale bar = 5μm. A B C The conserved Alp7/TAC-Alp14/TOG microtubule associated protein complex is targeted to the SPB via the pericentrin-like molecule The Transforming Acidic Coiled Coil (TACC) family proteins were originally identified as a group of proteins implicated in human cancers, and the family is conserved throughout evolution. TACC orthologues in various organisms localise to the centrosome. These proteins form a stable complex with another conserved TOG/Dis1/XMAP215 MAPs in virtually all organisms examined, which is Figure 2 Recruitment of the Alp7-Alp14 complex to the SPB via Pcp1 and establishment of proper kinetochore-microtubule attachment. The Alp7-Alp14 complex is loaded on the SPB by directly binding to pericentrin-like Pcp1 (top). The SPB-localising complex is essential for mitotic spindle assembly. Upon recruitment to the SPB, the Alp7-Alp14 complex promotes spindle microtubule polymerisation towards the kinetochore, in which Alp7 binds to the outer kinetochore component Ndc80, thereby establishing proper microtubulekinetochore attachment. Dis1 is another TOG/XMAP215 protein that also binds to Ndc80. essential for spindle assembly and proper spindle microtubule-kinetochore attachment. In human cells, either downregulation or upregulation of the TACC proteins is intimately linked to tumourigenesis, and their expression profiles are used for cancer prognosis. In fact, centrosome abnormalities associated with defective spindle assembly are one of the hallmarks of cancer. The molecular understanding of the mechanisms underlying centrosomal recruitment of the TACC proteins, therefore, is of critical importance for the aetiology of cancer and other human diseases. However, at the moment, it remains elusive as of which molecule(s) localising to the centrosome/ SPB is/are responsible for the recruitment of the TACC-TOG complex, i.e. the receptor for TACC at the centrosome. In fission yeast, Alp7/TACC and Alp14/TOG also form a stable complex, which is critical for mitotic and meiotic spindle assembly and proper chromosome segregation (Sato et al., 2007; Nature. 447(7142): 334-337; Kakui et al., 2013; Nat Cell Biol. 15(7): 786-796). Now we have determined five amino acid residues clustered within the TACC domain of Alp7 required for SPB localisation. Critically, these sequences are essential for the functions of Alp7, including proper spindle formation and mitotic progression. Moreover, we have identified pericentrin-like Pcp1 as a receptor for Alp7 loading on the mitotic SPB (Figure 2). The pericentrin family consists of the conserved centrosomal/SPB component proteins and is required for centrosome biogenesis and maturation. However, it is not known whether human pericentrin is involved in the recruitment of TACC proteins. It would be of great interest to explore whether human pericentrin is responsible for TACCs recruitment to the centrosome, and if so, what is the physiological consequence when this interaction is disrupted. Publications listed on page 172 LINCOLN'S INN FIELDS 111 SECRETORY PATHWAYS www.london-research-institute.org.uk/research/sharon-tooze Group Leader Sharon A Tooze Postdoctoral Scientists Delphine Judith Christopher Lamb Maria New Tim van Acker Martina Wirth Graduate Students Hannah Dooley Andrea Gubas Justin Joachim Scientific Officers Harold Jefferies Minoo Razi Autophagy is a highly conserved, homeostatic membrane-mediated pathway that delivers cytoplasmic components to the lysosome. Once delivered to the lysosome the material is degraded and recycled for re-use. Autophagy maintains cell health by targeting damaged proteins and organelles for degradation, and restoring amino acid pools during starvation through recycling by the lysosome. Furthermore, autophagy plays an essential role in infectious diseases, and in pathological conditions, such as neurodegeneration and tumorigenesis. A further understanding of the molecular mechanisms underlying the process is essential to exploit the potential benefit of manipulating autophagy to treat disease. Autophagy requires intracellular membrane compartments, such as the endoplasmic reticulum, Golgi complex, endosomes and lysosomes, alongside dedicated protein machinery, the autophagy related (Atg) proteins. We study how Atg proteins, novel autophagy regulators and trafficking proteins function during acute amino-acid withdrawal to elucidate the molecular basis of autophagy. Macroautophagy (here referred to as autophagy: self-eating) is the non-selective engulfment and removal of cytoplasmic proteins and organelles. Autophagy occurs at basal levels in all eukaryotic cells but is upregulated during amino acid deprivation, which increases the flux through the autophagosome to the degradative autolysosome. From the degradative autolysosome the cytosolic pool of amino acids, lipids and macromolecules can be replenished. Formation of autophagosomes occurs within minutes of amino acid deprivation in a now well defined pathway initiated at the preautophagosomal structure (PAS) located at specialised sites on the ER known as omegasomes, from which the phagophore forms. The phagophore then closes to form an autophagosome. The formation of autophagosomes utilise 18 out of the 36 Atg proteins identified in yeast. The ULK complex, the most upstream complex in the pathway, is negatively regulated by the master growth and energy sensors mTORC1 and AMPK. Inhibition of mTORC1, and activation of AMPK triggers activation of the ULK complex, and the class III phosphatidylinositol 3-kinase (PI3K) complex and their translocation to the phagophore. Downstream effectors of the ULK and PI3K complex include WIPI proteins that bind PI3-phosphate (PI3P), and the ubiquitin-like conjugation systems, Atg12-Atg5- 112 Scientific report 2014 LONDON RESEARCH INSTITUTE Atg16 which produce LC3-PE (LC3-II). While the Atg protein machinery is now known, many questions remain unanswered about the regulation of membrane dynamics, the source of the rapidly expanding autophagosomal membrane, and the interaction between the autophagosome and the other subcellular compartments. WIPI2 links PI3P to Atg12-5-16 Upon induction of autophagy, the class III PI3K complex produces a specific pool of PI3P on the omegasome. The autophagy-specific PI3P effectors, the WIPI proteins, are then recruited to the nascent phagophore. WIPI proteins are seven-beta propeller domain proteins, which are essential for autophagy, and through a comparative analysis of WIPI1 and WIPI2 we discovered WIPI2 functions to recruit the ubiquitin-like conjugates Atg12-5-16 and subsequently LC3-II. Hannah Dooley, in collaboration with Michael Wilson at the Babraham Institute, Cambridge UK, showed WIPI2b directly binds Atg16L1 (Dooley et al., 2014; Mol Cell. 55(2): 238-252). By mapping the reciprocal binding site on each protein, WIPI2 and Atg16L1 (Figure 1), we identified a single pair of amino acids, which mediate binding. WIPI2b binding to Atg16L1 is necessary and sufficient for structures adjacent to endosomes and forming phagophores lined up along the ER (Figure 2). Figure 1 Structural model of the region of Atg16L1 207-246 and residues that interact with WIPI2b (E226 and E230) and FIP200 (E235-E239) modelled ab initio using I-TASSER. See Dooley et al., 2014 for details. starvation-induced autophagy, ectopic LC3-II lipidation and for Salmonella targeting to autophagy. Atg9, trafficking and Rab effectors Phagophore and autophagosome formation both depend on Atg9, a multi-spanning membrane protein. We have shown that Atg9 traffics in small, mobile vesicles between multiple organelles, including recycling endosomes, late endosomes, and the Golgi compartment. Atg9 is also found in a unique conserved vesicular-vacuolar compartment called the ‘Atg9 compartment’, which is adjacent to forming phagophores. The control of Atg9 trafficking is likely to involve a complex set of regulatory proteins and machinery, including ones responsible for trafficking under normal growth conditions as well as under nutrient-deprived conditions. The composition of this compartment is being studied by Delphine Judith who, in collaboration with Bram Snijders and the Protein Analysis and Proteomics facility at LRI, is analysing the vesicular pathway taken by Atg9 in mammalian cells at a proteomic level. Figure 2 A panel of images from the cryo-CLXM analysis of HEK293 cells expressing mRFP-Atg9 and GFP-LC3. Left, cryo-fluorscence, middle reconstructed tomoX stack, and right, tomogram of the Atg9-compartment (red arrows) adjacent to endosomes (yellow) and GFP-LC3-positive forming phagophores (green arrows), which are seen on ER membranes (blue). White/black arrows indicate gold particles used for orientation. See Duke et al., 2014 for details. One of the strengths of our work on autophagy is the morphological techniques we use to place our biochemical work in a cellular context. In collaboration with Lucy Collinson, head of the Electron Microscopy facility at LRI, and Elizabeth Duke (Diamond, Oxford), Minoo Razi has studied the morphology of the Atg9 compartment after using cryo-soft X-ray and correlative light microscopy (Duke et al., 2014; Ultramicroscopy. 143: 77-87). Under these near-native imaging conditions, Atg9 was seen in distinct vacuolar While recent data supports the role of the ER in the formation of the phagophore, it is equally clear that Golgi and other compartments may provide additional membranes. To understand the contributions of other membranes, we focused on Rab proteins, GTPases that mediate vesicular fusion, and RabGAPs, GTPase activating proteins that inactivate Rabs, called TBC proteins. TBC1D14, a Rab11 effector, robustly co-localises with ULK1 and is present on Golgi and recycling endosomes (Longatti et al., 2012; J. Cell Biol. 197(5): 659-675). Chris Lamb is exploring how TBC1D14 coordinates recycling endosome traffic to autophagosomes and the machinery, including the Rabs, which link TBC1D14 to autophagosome formation. Novel regulators of autophagy Our genome-wide siRNA screen under amino acid starvation identified several putative candidates that our group continues to investigate. SCOC, a short coiled-coil protein, is a Golgi-localised protein that interacts with ULK1 and UVRAG, dependent on FEZ1 (McKnight et al., 2012; EMBO J. 31(8): 1931-1946). Given that ULK1 and UVRAG (a subunit of the endosomal PI3K complex) act sequentially in autophagy, we are testing the hypothesis that SCOC may regulate the progression of the autophagosome membrane and maturation. Martina Wirth in collaboration with Stéphane Mouilleron (Structural Biology facility, LRI) are using a structure-function approach to understand the function of SCOC. Martina is complementing these studies by developing zebrafish models to study SCOC and other autophagy regulators. WAC, a WW domain-containing adaptor with coiled-coil protein, is another autophagy regulator identified in our genome-wide siRNA screen. WAC is found in both a nuclear and cytoplasmic pool, and Justin Joachim is dissecting the function of WAC in these two compartments, in particular the interaction of WAC with the Golgi complex. Publications listed on page 172 LINCOLN'S INN FIELDS 113 SIGNALLING AND TRANSCRIPTION www.london-research-institute.org.uk/research/richard-treisman Extracellular stimuli such as growth factors and mitogens act through signal transduction processes to induce alterations in cell proliferation, differentiation and mechanical activities including motility and adhesion. Our group focuses on analysis of the serum response factor (SRF) transcription factor network, which controls transcription of many genes involved in these processes. Group Leader Richard Treisman Associate Scientist Patrick Costello Postdoctoral Scientists Laura Collard Jessica Diring Cyril Esnault Charles Foster Anastasia Mylona Graduate Students Sofie Eriksson Francesco Gualdrini Magdalena Kratochvilova Richard Panayiotou Clinical Research Fellow Cynthia Yu-Wai-Man Scientific Officers Diane Maurice De Coulon Mathew Sargent SRF activity is regulated by two families of signalregulated cofactors. The myocardin-related transcription factors (MRTFs) sense changes in cellular G-actin concentration associated with altered Rho GTPase activity. We study both the MRTFs and other proteins subject to this novel form of regulation. In contrast, the ternary complex factors (TCFs) are classical ERK phosphorylation substrates downstream of Ras. Both Rho and Ras signalling are implicated in transformation, invasion and metastasis, but the role of the SRF network in these processes remains to be elucidated. Rho-actin signalling and the MRTF pathway A central focus of current work is the mechanism by which Rho GTPase signalling controls cell behaviour. The MRTFs are novel G-actin binding proteins, binding G-actin through an N-terminal regulatory domain containing three copies of the RPEL G-actin binding motif. Three other families of RPEL proteins have been identified: the Phactr proteins bind the PP1 catalytic subunit and represent a novel family of putative PP1 regulators; in addition, our recent studies have identified two protein families of cytoskeletal regulators. While G-actin-controlled nucleocytoplasmic shuttling represents a major mode of regulation for MRTF-A, MRTF-B, and Phactr1, our data shows that G-actin also regulates the MRTFs at other levels, and other regulatory mechanisms must clearly operate on non-shuttling RPEL proteins. Our previous work with the MRTFs identified a nuclear import signal with the RPEL domain whose ability to recruit importin a-b heterodimers is antagonised by G-actin binding. A similar mechanism operates in the case of the Phactr1 protein, which also shuttles to the nucleus in response to Rho GTPase activation. Structural studies of the N-terminal Phactr1 NLS-RPEL motif by Stéphane Mouilleron (Protein Structure, LRI) show that actin binding physically occludes sequences required for membrane association of Phactr3 and Phactr4, so Magdalena Kratochvilova 114 Scientific report 2014 LONDON RESEARCH INSTITUTE is studying whether G-actin also affects this interaction. Magdalena has demonstrated that the Phactr1-PP1 complex does not contain other cofactors, and with Stéphane, has shown that Phactr1 binding does not occlude the PP1 substrate binding site. Magdalena is now using proteomic and affinity-labelling approaches to identify potential Phactr1-PP1 substrates. The Phactr1-PP1 and MRTF-importin interaction studies demonstrate a common mechanism for G-actin regulation by direct competition for overlapping binding sites. Jessica Diring has now shown that a related mechanism operates in the case of the RPEL cytoskeletal regulatory proteins. These contain a catalytic domain adjacent to a single RPEL motif, and form a stable 1:1 complex with actin. Catalytic activity is inhibited in the complex, and Jessica finds that mutations that prevent actin binding relieve this inhibition. Structural studies conducted with Stéphane show that in the complex, G-actin makes additional specific contacts with the catalytic domain, and Jessica has shown that ablation of these contacts also relieves inhibition. Richard Panayiotou has continued his investigation of MRTF nuclear export, which is dependent on the Crm1 exportin and the integrity of the N-terminal RPEL motifs. Richard has mapped a Crm1 binding site in the MRTF N-terminal sequences, demonstrating that its activity is potentiated by a nearby phosphorylation event, and showing that it can confer regulated export on the Phactr1 protein, whose export is normally independent of Crm1. With data from others, these observations suggest that regulated MRTF localisation requires cooperation between multiple independent Crm1 binding sequences, together with G-actindependent occlusion of the importin binding site. Cyril Esnault and Francesco Gualdrini have continued studies on the regulation of MRTFs by G-actin. Francesco has continued the analysis of MRTF regulation by nuclear actin using genomic approaches to show that targeting of MRTFs to the Figure 1 Haematopoietic stem/progenitor cells (HSC/Ps) lacking SRF exhibit defective adhesive and polarisation responses to SDF-1. HSC/P cells (CFSE: green) were plated on monolayers of endothelial cells (SNARF: red) above a source of SDF-1 and imaged after 45 minutes. Cells lacking SRF failed to flatten and move into the endothelial monolayer toward the source of SDF-1. nucleus in the absence of G-actin depletion is insufficient to induce transcription. This appears to reflect the operation of two novel MRTF regulatory pathways controlled by G-actin through the N-terminal regulatory domain. One pathway acts to control MRTF-SRF interaction, and Francesco is using biochemical approaches to investigate the MRTF sequences involved, complementing Anastasia Mylona’s structural work on the MRTFSRF interaction. The other pathway appears to regulate RNA Polymerase II, and genomic data suggest that it prevents productive transcription by disruption of CTD phosphorylation. A central question is how these regulatory phenomena relate to the natural physiology of MRTF-SRF regulation. MRTFs are nuclear under resting conditions in certain cell types or environments. Cynthia Yu-Wai Man is investigating human Tenon’s fibroblasts, where MRTFs are nuclear under resting conditions, while Charlie Foster is looking at mouse carcinoma-associated fibroblasts, both of which appear to be ‘myofibroblast’-like. Given that Cyril Esnault’s genomic analysis of the SRF network identified a strong link between adhesion signalling and MRTF activation, Cynthia and Charlie are investigating the relation between MRTF nuclear accumulation and matrix stiffness, and in related work, Laura Collard is pursuing the relationship between adhesion, steady state G-actin levels and MRTF activation. Patrick Costello and Mathew Sargent have completed our analysis of the role of the SRF network in murine haematopoiesis. Patrick previously found that hematopoietic stem cells fail to colonise the foetal bone marrow, although other tissues remain normal. With Mathew, he extended this to show that this reflects uncoupling of SRF from MRTF activity, and that MRTF-A and MRTF-B function redundantly in this system. With Cyril Esnault, Patrick performed a transcriptome analysis, demonstrating that as in the fibroblast system, SRF-dependent transcripts are highly enriched in those encoding cytoskeletal structural genes and regulators. Curiously, however, many of these genes are specific to the stem cells, showing that cell context is an important determinant of specificity for the SRF-MRTF system. Regulatory properties of the TCF-SRF network Although SRF was first identified in studies of the fibroblast response to serum mitogens over two decades ago, its role in this response has remained unclear. Patrick Costello and Diane Maurice found that TCF-SRF signalling is important for both acute TCR-stimulated and homeostatic proliferation of CD4 T cells. They are using RNAseq to characterise the transcriptional responses involved. Diane has now also characterised the role of SAP-1 and SRF in T cell differentiation and the response to infectious challenge. Diane found that SRF is required for the initial proliferative expansion of short-lived effector cells and simultaneously suppresses early memory precursor differentiation. The TCFs are phosphorylated at multiple sites. Anastasia Mylona has shown that these exhibit variable phosphorylation kinetics, and with Charlie Foster showed these act both positively and negatively on mediator recruitment. Cyril Esnault and Francesco Gualdrini are investigating the relationship between ERK signalling, TCF phosphorylation, chromatin modifications and transcription. Using phorbol esters to activate ERK signalling at model TCF-dependent genes, they found that unphosphorylated Elk-1 is sufficient to induce certain chromatin modifications, while others require phosphorylation but not transcription, and others reflect an RNA polymerase II recruitment. They are now seeking to extend these studies genome-wide. Publications listed on page 172 LINCOLN'S INN FIELDS 115 CHROMOSOME SEGREGATION www.london-research-institute.org.uk/research/frank-uhlmann Group Leader Frank Uhlmann Postdoctoral Scientists Yasutaka Kakui Thomas Kuilman Lidia Lopez Serra Yasuto Murayama Catarina Samora Graduate Students Molly Godfrey Meghna Kataria Ainhoa Mariezcurrena Rahul Thadani Scientific Officers Celine Bouchoux Maria Ocampo-Hafalla Aneuploidy, i.e. missing or supernumerary chromosomes, is a hallmark of malignant tumour progression. A large number of genes that orchestrate faithful chromosome segregation during mitotic cell divisions are tumour suppressors or turn into potent oncogenes if misregulated. The aim of the Chromosome Segregation group is to investigate the function of these genes and the cellular mechanisms that safeguard accurate chromosome segregation. In particular, we are investigating the contribution of structural chromosomal proteins to sister chromatid cohesion and chromosome condensation, processes that ensure faithful segregation of centimetre-long chromosomal DNA molecules within micrometre-sized cells. We also investigate how the kinases and phosphatases of the cell division cycle machinery bring about ordered completion of chromosome segregation. Chromosome cohesion and tumourigenesis The cohesin complex is a central player in chromosome biology. Defects in cohesin and its regulators are responsible for chromosome missegregation in many human malignancies. They are also the cause for Cornelia de Lange syndrome, a severe developmental disorder. The genomic DNA that makes up the chromosomes is replicated during S phase of the eukaryotic cell cycle. After replication, the two newly synthesised sister chromatids remain connected with each other by the chromosomal cohesin complex. Our group has contributed over the years to understand how this ring-shaped multi-subunit protein complex works to build sister chromatid cohesion. Sister chromatid cohesion forms the basis for the pairwise alignment of DNA replication products on the spindle apparatus in mitosis, to allow their faithful segregation into daughter cells. Cells defective in sister chromatid cohesion make errors in chromosome segregation, giving rise to aneuploid cells that lack or contain extra copies of chromosomes. Aneuploidy is a hallmark of malignant tumour progression. Human heritable mutations that compromise the fidelity of chromosome segregation are inevitably linked to early onset tumourigenesis. This year, we have made progress towards understanding how cohesin works as a fascinating molecular machine that holds sister chromatids together. 116 Scientific report 2014 LONDON RESEARCH INSTITUTE Biochemical reconstitution of topological DNA binding by the cohesin ring The cohesin complex consists of at least four subunits that together form a large proteinaceous ring. It is thought that cohesin holds together sister chromatids by topologically embracing them. While the embrace model provides an important conceptual framework for sister chromatid cohesion, it leaves many fundamental question wide open. If cohesin topologically embraces DNA, how does the DNA get into the ring and where on chromosomes can this reaction happen? Equally, how does DNA come out of the ring again during cohesin’s dynamic DNA binding cycle? Finally, cohesin incorporates an ABC-type ATPase that is required for its function, so how does the ATPase fuel cohesin’s activities? Definitive answers to these questions require that we can study cohesin’s behaviour in vitro, however, the biochemical reconstitution of cohesin activity has remained an unattained goal. We were now successful in expressing and purifying the fission yeast cohesin complex, as well as its Mis4/Ssl3 cohesin loading factor, which is essential for cohesin function in vivo (Figure 1). Incubation of cohesin with DNA led to spontaneous topological loading of cohesin onto DNA, in an ATP hydrolysis-dependent fashion, but this reaction remained inefficient. Addition of the cohesin loader stimulated ATP hydrolysis and cohesin Figure 1 Biochemical reconstitution of topological cohesin loading onto DNA. A. Purification of fission yeast cohesin and the cohesin loader complex after overexpression of their respective subunits. The final gel filtration steps of the purification are shown. Fractions were analysed by SDS polyacrylamide electrophoresis followed by Coomassie blue staining. B. Schematic of the cohesin loading reaction. C. A circular DNA substrate is required for the loading reaction, hinting at the topological nature of cohesin binding. The input and bead-bound fractions following the loading reaction are shown and were quantified. The mean and standard deviation of three independent experiments are shown. B A C loading onto DNA. We found that the cohesin loader contacts cohesin at multiple sites around the ring circumference. One of these contacts lays on cohesin’s Psc3 subunit, an essential yet hitherto enigmatic part of the cohesin complex. Using mutational analysis and peptide competition experiments, we showed that at least three loader contacts along the cohesin ring coordinatedly stimulate the cohesin loading reaction. Considering the large dimensions of the cohesin ring, it is likely that a conformational rearrangement must take place to accommodate three simultaneous contacts of the loader with cohesin. The cohesin loader can thus be thought of as a template or mould onto which cohesin holds onto to facilitate the loading reaction. Our in vitro reconstitution of cohesin loading onto DNA provides mechanistic insight into the initial steps of the establishment of sister chromatid cohesion and other chromosomal processes mediated by cohesin. The results are important not only to understand cohesin, but also the ubiquitous family of chromosomal structural maintenance of chromosomes (SMC) complexes, of which cohesin is a member. SMC complexes share essential functions in various chromosomal activities in all organisms from bacteria to humans. Outlook Now that we have gained the ability to investigate cohesin behaviour in vitro, we would like to directly observe cohesin’s loading onto DNA. We will use a combination of biochemical, structural, single molecule and imaging approaches to do this. In particular, single molecule FRET-based assays and electron microscopy have the potential to shed unprecedented insight into both the cohesin loading reaction as well as the final product of the reaction, the cohesin ring on DNA. Once cohesin is loaded onto DNA, the probably most exciting time during its residence on chromosomes comes during DNA replication in S phase, when the sister chromatid is synthesised and cohesin will ensure to hold the two together. We will extend our biochemical assays to address how cohesin identifies the two replication products and establishes linkages between them. Publications listed on page 173 LINCOLN'S INN FIELDS 117 CANCER GENOMICS www.london-research-institute.org.uk/research/peter-van-loo Group Leader Peter Van Loo Graduate Student Stefan Dentro The advent and exponential cost decrease of massively parallel sequencing technologies over the past years has enabled sequencing entire cancer genomes. This resulted in unique opportunities for cancer research. Large-scale consortia (The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC)) have now produced whole genome sequences of thousands of cancer genomes, and are making their data available to the community. I argue that we have so far only skimmed the surface of what can be learned from this unprecedented wealth of data. Therefore, there is a clear need for in-depth large-scale pan-cancer analyses. Our group, which started in October 2014, focuses on integrative analyses of large-scale public ‘omics’ data, leveraging the wealth of cancer genomics data into large-scale pan-cancer analyses to understand carcinogenesis and cancer evolution. Characterising the landscape of tumour suppressor genes Many cancer genes are somatically altered in only a very low proportion of tumours, providing a clear rationale for large-scale pan-cancer analyses of driver mutations. We are applying approaches centred on copy number analysis to characterise the landscape of tumour suppressors. Tumour suppressors can be inactivated by a combination of a deleterious germline variant, combined with somatic loss-of-heterozygosity (LOH) of the other allele. We are performing a large-scale pan-cancer screen for this combination of events aiming to identify new tumour suppressors. Many tumour suppressors are targeted by homozygous deletions, removing both parental copies. Because any homozygous deletion that includes a gene that confers a survival advantage is eliminated by negative selection, homozygous deletions are rare and often focal. Admixture of normal cells in tumour samples has historically hindered the reliable identification of homozygous deletions. We previously developed ASCAT (Allele-Specific Copy number Analysis of Tumours, Van Loo et al., 2010; Proc Natl Acad Sci U S A. 107(39): 16910-16915), a method to derive copy number profiles of tumour cells accounting for normal cell admixture and tumour aneuploidy. Methods such as ASCAT can now effectively deconvolute copy number profiles of tumour cells from those of admixed normal cells and reliably identify homozygous deletions in tumour samples. 118 Scientific report 2014 LONDON RESEARCH INSTITUTE We are applying ASCAT to thousands of samples across cancer types, to screen for tumour suppressors through recurrent homozygous deletions. For a subset of these cases, point mutation data, gene expression data and/or DNA methylation data will also be available, which we will correlate with detected homozygous (and hemizygous) deletions, allowing us to more clearly delineate target genes within regions of homozygous deletions. Through this screen, we aim to characterise the landscape of tumour suppressors and, particularly, identify rare tumour suppressors. Molecular archaeology of cancer: inferring timelines of cancer development and evolution The cancer genome contains within it an archaeological record about its past, and we previously pioneered methods to disentangle a cancer’s life story from sequencing data (Nik-Zainal et al., 2012; Cell 149(5): 994-1007) (Figure 1). We anticipate that a large-scale pan-cancer approach to obtain detailed evolutionary histories of tumours would give profound insights into carcinogenesis and cancer evolution. We can construct life histories of thousands of tumours from their genome sequences, using both driver and passenger mutations. By obtaining detailed timelines of many cancers’ evolutionary histories that include driver mutations, copy number changes, rearrangements and mutational processes, we aim to identify the initiating events transcriptome is confounded by expression signals originating from admixed normal cells. Gene expression analysis by massively parallel sequencing (RNAseq) allows allele-specific expression measurements. It can be shown that the expression in tumour cells can be separated from that in normal cells, given the fraction of tumour cells, the allele-specific copy number profiles of tumour cells, and applying a few reasonable hypotheses (Figure 2). We aim to develop such bioinformatics approaches to deconvolute the tumour cell transcriptomes from transcriptomes of admixed normal cells. Figure 1 Molecular archaeology of cancer: an example. From the picture on the right, one can infer that the purple mutations happened first, then the blue chromosome duplicated, and then the yellow mutations occurred. In addition, from the relative numbers of yellow and purple mutations, one can infer when in the tumour’s lifetime the blue chromosome duplicated. of cancer development, and the events that are selected for later in a cancer’s lifetime, including those that drive late clonal expansions and that may play a role in tumour malignancy. In addition, these analyses will allow blueprints of the subclonal architecture across cancer types in unprecedented detail and on an unprecedented number of cases, allowing a glimpse into a tumour’s future. Complementary to this, we are performing smaller-scale collaborative studies of tumour bulk sequencing, in combination with single-cell and multi-sample sequencing of primary tumours, metastases and circulating and disseminated tumour cells, aiming to gain insight into tumour evolution and metastasis. Deconvoluting expression in tumour and normal cells We aim to understand how changes to the genome lead to transcriptomic changes to eventually cause cancer. Deep understanding of the cancer Figure 2 Principle of a method to deconvolute the tumour cell transcriptomes from transcriptomes of admixed normal cells, using copy number data and (allele-specific) expression from RNAseq. We will apply these methods to large pan-cancer RNAseq datasets, allowing a transcriptome-wide view of cancer across cancer types. We expect these tumour cell-specific expression profiles will result in a better taxonomy of cancer than mixed cell population expression profiles. Finally, expression profiles of admixed normal cells will allow insight into the cellular composition and transcriptional state of the tumour stroma. In the longer term, we aim to develop integrative genomics-transcriptomics approaches that study the influence of point mutations, copy number changes and structural variants on transcription at the gene or transcript level and at the transcriptome level, and to apply these approaches in a large-scale pan-cancer setting to understand the basic principles of cancer development and cancer evolution within and across tumour types. Publications listed on page 173 Copy number profile Expression in tumour samples Samples Genes Samples Genes Genes Samples Inferred expression in tumour cells Inferred expression in normal cells LINCOLN'S INN FIELDS 119 CELL FATE AND GENE REGULATION www.london-research-institute.org.uk/research/folkert-van-werven Group Leader Folkert van Werven Postdoctoral Scientist Fabien Moretto Graduate Student Minghao Chia Scientific Officer Gianpiero Spedale Masters Student Natalia Robert Figure 1 Schematic overview of how the mating type signal controls entry into gametogenesis. The aim of our research group is to elucidate the molecular mechanisms by which the cell integrates multiple signals to achieve a binary cell fate decision – whether or not to differentiate. Unfolding these mechanisms is critical for the understanding of how cell specialisation leads to multi-cellularity during development, and how impaired signalling can cause abnormal development and diseases such as cancer. The budding yeast S. cerevisiae is an ideal model system to study this problem. In response to a combination of extracellular and intracellular cues budding yeast undergoes a highly conserved cell differentiation programme called gametogenesis. Since entry into gametogenesis is controlled by only two master regulators in this model organism, there is the unique opportunity to study the molecular and quantitative aspects of this cell fate. Transcription of two long noncoding RNAs controls the cell fate decision leading to gametogenesis Expression of the master regulatory genes, IME1 and IME4, drive the cell fate decision leading to gametogenesis in budding yeast. This cell fate is also controlled by the mating-type locus. In order to initiate gametogenesis, diploid yeast cells need to express both mating type genes, MATa and MATα. The combined gene product of MATα and MATa, the a1-α2 heterodimer, inhibits RME1 transcription in diploid cells (Figure 1A), but how in A B 120 Scientific report 2014 LONDON RESEARCH INSTITUTE haploid cells Rme1 represses IME1 transcription was never understood. We discovered that in cells with the haploid mating-type, expression of IME1 is inhibited by a long noncoding RNA (lncRNA) called IRT1 that is located in the IME1 promoter and induced by the Rme1 transcription factor (Figure 1B). Transcription of this lncRNA recruits the Set2 histone methyltransferase and Set3 histone deacetylase complex to establish repressive chromatin in the IME1 promoter (Figure 1B). Figure 2 A. Overview of the signals controlling the IME1 promoter. B. Annotation of two lncRNAs in the IME1 promoter. C. Model describing the mechanism by which the lncRNA IRT1 represses IME1. A B C The chromatin remodelling enzymes Set2 and Set3 are highly conserved from yeast to human, and lncRNAs are also abundant in higher eukaryotes. This raises the interesting possibility that lncRNAs repress gene expression by a conserved mechanism. In addition to IME1, expression of the master regulator IME4 is antagonised by an antisense transcript in haploid cells (Figure 1B). This antisense transcript in turn is repressed by a1-α2 allowing expression of IME4 in diploid cells (Figure 1B). The mating-type dependence of IME4 expression led to a second key discovery from this study. When the expression of both the lncRNA in the IME1 promoter and the IME4 antisense transcript is inhibited, cells expressing the haploid mating-type enter gametogenesis with kinetics that are indistinguishable from cells expressing both mating-types (Figure 1). Thus, transcription of two lncRNAs governs mating-type control of gametogenesis in yeast. Current Research directions The promoter of the master transcription factor for entry into gametogenesis, IME1, integrates nutrient and mating-type signals to make the binary cell fate decision (Figure 2A). Whole genome transcriptome analysis identified two lncRNAs in the IME1 promoter (Figure 2B). In cells of the haploid mating-type, IME1 is repressed by the lncRNA IRT1 (Figure 2C). Induction of IRT1 by the transcription factor Rme1 recruits the methyltransferases Set1 and Set2 to methylate histone H3 at lysine 4 and 36. These marks are recognised by the histone deacetylase complexes Set3C and Rpd3(S) and establish a repressive chromatin state in the IME1 promoter (Figure 2C). The mechanism of cell fate control described here provides a starting point for further investigations. The goals are: Firstly, screen for novel factors required for gene repression by lncRNAs and, secondly, investigate how widespread gene repression by lncRNAs is across the genome. How do master regulatory genes integrate multiple signals? Multiple signals are required to initiate gametogenesis in budding yeast: nutrients such as nitrogen compounds and glucose need to be absent from the medium, cells need to respire (use a non-fermentable carbon source) and must be diploid. These signals drive entry into gametogenesis and all converge on the IME1 promoter (Figure 2A). This promoter is one of the largest and highly regulated promoters in budding yeast. To understand how these signals are integrated to make a binary cell fate decision, it is essential to identify all the molecular players and pathways involved in acting at the IME1 promoter. The lab will use proteomic and genetic approaches to identify these factors. Perspective The aims outlined here form the beginning of a systematic investigation on how multiple signals are integrated to drive cell fate decisions. It is often not well understood how, in higher eukaryotes, master regulatory genes make binary decisions that are important for development of an organism. Aberrant expression of these master genes due to impaired signalling can cause abnormal development and diseases such as cancer. The IME1 promoter serves as a basic model system for studying signal integration at master regulatory genes. The findings from these studies could shed light on how complex promoters are regulated in higher eukaryotes. LINCOLN'S INN FIELDS 121 CELL MOTILITY www.london-research-institute.org.uk/research/michael-way Group Leader Michael Way Postdoctoral Scientists Jasmine Abella David Barry Joseph Cockburn Flavia Leite Caitlin Tolbert Graduate Students Chiara Galloni Julia Pfanzelter Xenia Snetkov Scientific Officers Theresa Higgins Antonio Postigo Cell adhesion and motility play a critical role during the development and throughout the lifetime of multi-cellular organisms. Unfortunately, deregulation of these two fundamental cellular processes frequently occurs during pathological situations such as tumour cell metastasis. Our research focuses on understanding how signalling networks and the cytoskeleton regulate cytoplasmic transport, as well as cell adhesion and migration. We use a combination of quantitative imaging and biochemical approaches to study vaccinia virus as a model system to interrogate the regulation and function of Src and Rho GTPase signalling, actin and microtubule-based transport as well as cell migration. Outside the context of vaccinia infection, we also investigate the cellular function of Tes, a tumour suppressor that negatively regulates Menadependent cell migration, as well as the mechanisms regulating the assembly and function of invadopodia. Ena/VASP proteins regulate cell migration by promoting actin polymerisation at the plasma membrane by antagonising actin filament capping and acting as processive actin polymerases. The intracellular targeting of Ena/VASP proteins is mediated through the interaction of their N-terminal EVH1 domain with ‘FPPPP’ sequence motifs found in a variety of cytoskeletal proteins including lamellipodin and zyxin. Of all the proteins interacting with the EVH1 domain of Ena/VASP proteins, Tes, a tumour suppressor and focal adhesion protein, stands out as the only one known to lack an ‘FPPPP’ motif. Tes interacts with Mena via its C-terminal LIM3 domain and is also unique in being the only protein shown to bind a single Ena/VASP family member. Furthermore, Tes inhibits Mena-dependent cell migration by negatively regulating the localisation of Mena at focal adhesions and the leading edge. Given the interaction of Tes with Mena, we sought to identify additional Ena/VASP EVH1 binding partners lacking ‘FPPPP’ motifs. Using a combination of biochemical approaches and mass spectrometry, we found the EVH1 domain interacts directly with Abi in the WAVE regulatory complex (WRC), even though it lacks an ‘FPPPP’ motif. The WRC plays an essential role in promoting Arp2/3 complex-dependent actin polymerisation in response to Rac signalling. Using a Far-western approach, we found that the EVH1 domain interacts with residues 352-394 in the 122 Scientific report 2014 LONDON RESEARCH INSTITUTE proline rich region of human Abi1. With this information, we were able to generate an Abi mutant (termed AbiΔEVH1) that incorporates into the WAVE complex but cannot interact with Ena/ VASP proteins. When expressed in cells, GFPtagged AbiΔEVH1 was recruited to plasma membrane but was less effective than Abi at promoting cell migration. Given this phenotype, we collaborated with Michael Rosen (Howard Hughes Medical Institute and Department of Biophysics, UT Southwestern Medical Center, Dallas, USA) to investigate whether VASP modulates the ability of the WRC to promote Arp2/3-dependent actin polymerisation in vitro. We found that VASP increases the extent of actin polymerisation induced by Rac1-activated WRC. Importantly, this stimulation, which is over and above the intrinsic actin filament elongation activity of VASP, depends on the proline rich region of Abi. In contrast to the tetrameric full-length protein, monomeric VASP or its isolated EVH1 domain does not activate the WRC to stimulate Arp2/3-mediated actin polymerisation even at high concentrations. Loss of the VASP F-actin binding motif, which is essential for its actin filament elongation activity, also completely abolishes the ability of VASP to enhance actin assembly by the WRC. It is possible that the simultaneous engagement of a VASP tetramer with Abi and the ‘LPPPP’ motif in WAVE increases the activity of the WRC. However, oligomerisation interaction in Drosophila, we collaborated with Sven Bogdan (Institute of Neurobiology, University of Münster, Germany). Drosophila larval macrophages adhering to a substrate have a polarised actin cytoskeleton with a broad lamellipodial cell front (Figure 1). In contrast, abi mutant macrophages have a ‘spiky’ morphology. Re-expression of dAbi but not dAbi∆Ena rescues this defect in cell morphology. Live cell imaging also reveals that Abi∆Ena macrophages have a reduced rate of membrane protrusion compared to dAbi macrophages. Furthermore, Ena is no longer at the leading edge of lamellipodial protrusions but becomes re-localised to the tips of filopodia-like protrusions when it cannot bind dAbi. Figure 1 Maximum intensity projection SIM image of the actin cytoskeleton (black) in Drosophila hemocytes alone cannot account for our observations, since mutating the F-actin binding motif of VASP abrogates activity. While not definitive, our data are most consistent with a model in which VASP binds Rac-activated WRC with high affinity based on tetramerisation-mediated avidity as well as actin filaments, thus increasing the association of the WRC with filaments. To facilitate analysis of the physiological role of the Ena/VASP – WRC interaction, we switched to Drosophila, as it only has a single isoform for each protein. Drosophila Abi (dAbi) and the EVH1 domain of Ena have 38 and 72 % sequence identity to their respective human counterparts. Nevertheless, the EVH1 domain of Ena still binds dAbi. Using a Far-western approach, we found that Ena interacts with two ‘LPPPP’ motifs in dAbi. Mutation of both ‘LPPPP’ motifs does not affect the ability of dAbi (dAbi∆Ena) to incorporate into the WRC or co-localise with WAVE at the plasma membrane of Drosophila S2 cells. To investigate the consequences of the loss of the dAbi-Ena In vivo, dAbi and a functional WRC are required in the Drosophila larval visual system for the correct axonal targeting of photoreceptor neurons (R-cells) to their respective optic ganglions in the fly brain. We found that the loss of the ability of dAbi to interact with Ena resulted in a similar defect in R-cell targeting as the complete loss of dAbi. Ena and the WRC are also essential for normal Drosophila egg development, while the loss of abi in the germline results in small and abnormally shaped eggs. Re-expression of wild type Abi rescues the egg morphology defects and female sterility of abi mutant flies. In contrast, abi mutant flies expressing dAbi∆Ena are still completely sterile, containing smaller and abnormally round eggs. Late stage Abi∆Ena mutant egg chambers also have defects in nurse cell cortical actin integrity resulting in detached cytoplasmic actin bundles and ring canals. In summary, our in vitro analysis has now demonstrated that Ena/VASP proteins directly impact on the activity of the WAVE complex, while our observations in Drosophila have revealed that the function and activity of Ena/VASP proteins and the WAVE complex are intimately linked. Publications listed on page 173 LINCOLN'S INN FIELDS 123 Sub-nanometre resolution structure of a replicative helicase. Image: Alessandro Costa, Architecture and Dynamics of Macromolecular Machines Group. 124 Scientific report 2014 LONDON RESEARCH INSTITUTE CLARE HALL The London Research Institute Clare Hall Laboratories are located North of London. The main focus of the research for the laboratories housed on the Clare Hall campus is genome integrity; including DNA repair, recombination and replication, cell cycle control and transcription. The researchers are supported by an excellent range of Technology Core Facilities. CLARE HALL 125 DNA DAMAGE RESPONSE www.london-research-institute.org.uk/research/simon-boulton Group Leader Simon Boulton Postdoctoral Scientists Carrie Adelman Sara Garcia-Gomez Ana Maria Leon Ortiz Pol Margalef Paulina Marzac Kenichiro Matsuzaki Stephanie Panier Grzegorz Sarek Julien Stingele Jennifer Svendsen Jean-Baptiste Vannier Graduate Students Rafal Lolo Martin Taylor Scientific Officers Valerie Borel-Vannier Zuzana Licenikova-Horejsi Julie Martin Mark Petalcorin Tohru Takaki DNA is highly susceptible to damage and must be repaired correctly to prevent genome instability. Failure to correctly repair DNA damage is the underlying cause of a number of hereditary cancer predisposition syndromes such as Fanconi Anemia and Blooms. The long-term aim of my group is to understand how DNA double-strand break (DSB) repair pathways, such as non-homologous end joining (NHEJ) and homologous recombination (HR), are regulated in mitotic cells and during meiosis. We also have an active interest in understanding how these pathways impact on human diseases such as cancer. Phosphorylation-dependent PIH1D1 interactions define substrate specificity of the R2TP co-chaperone complex Molecular chaperones facilitate the folding and unfolding of polypeptides and are essential for the assembly of large protein complexes. The human R2TP complex consists of four subunits: RUVBL1, RUVBL2, PIH1D1 and RPAP3 and is essential for the assembly of a number of multi-subunit molecular machines including small nucleolar ribonucleoproteins (snoRNPs), spliceosomal snRNP U4, RNA polymerase II and mTORC1 and SMG1 complexes. However, the molecular basis of substrate recognition by the R2TP complex was unclear. Our previous study revealed that CK2 phosphorylation of the co-chaperone TEL2 is essential for direct binding to PIH1D1 and its disruption leads to destabilisation of mTOR and SMG1 and to a lesser extent ATM, ATR and DNA-PKcs. As PIH1D1 is not predicted to contain any of the known phospho-binding domains such as 14-3-3, FHA, BRCT, WD40, WW and Polo-box domains, it was unclear how it recognises phosphorylated TEL2. Furthermore, whether phosphorylation-dependent binding represents a universal substrate recognition mechanism for the R2TP complex had not previously been explored. In recent work in collaboration with the group of Steve Smerdon at Mill Hill (NIMR), we demonstrate that the N-terminal PIH1D1 region PIH-N is a novel phospho-binding domain required for recognition of phosphorylated substrates, while the C-terminal region of PIH1D1 binds to the other components of the R2TP complex. The crystal structure of PIH-N domain fragment bound to the phosphorylated TEL2 peptide revealed a highly specific phosphopeptide recognition mechanism in which Lys 57 126 Scientific report 2014 LONDON RESEARCH INSTITUTE and 64 in PIH1D1, together with a conserved DpSDD phospho-motif within TEL2, are essential and sufficient for binding. Proteomic analysis of PIH1D1 interactors identified R2TP complex substrates that are recruited by the PIH-N domain in a sequence-specific and phosphorylation-dependent manner suggestive of a common mechanism of substrate recognition. We proposed that protein complexes assembled by the R2TP complex are defined by phosphorylation of a specific motif and recognition by the PIH1D1 subunit. RTEL1 RTEL1 is a helicase that was originally identified by mapping of loci that control telomere length differences between M. musculus and M. spretus. RTEL1 plays a critical role in genome stability as knockout mice are embryonic lethal and cells derived from these mice exhibit telomere fragility and loss. We previously identified RTEL1 as a key regulator of homologous recombination (HR) in a genetic screen for anti-recombinases and biochemical studies revealed that human RTEL1 promotes the disassembly of D loop recombination intermediates in vitro. Based on our original study of RTEL1 we speculated that its role at telomeres might reflect a need to regulate HR. Indeed, we previously established that RTEL1 performs two distinct functions at vertebrate telomeres in promoting t-loop unwinding (requiring its D-loop disrupting activity) and counteracting the formation of telomeric G4-DNA structures to facilitate telomere replication (Vannier et al., 2012; Cell 149(4): 795-806). We proceeded to show that binding of RTEL1 to proliferating cell nuclear antigen (PCNA) is critical for unwinding telomeric G4-DNA structure but was found to be dispensable for t-loop disassembly (Vannier et al., 2013; Science 342(6155): 239-242). While it was known that RTEL1 transiently localises to telomeres during the cell cycle, the mechanism by which RTEL1 is recruited to telomeres to promote t-loop disassembly is totally unclear. In our most recent study, we make the unexpected discovery that a single telomere binding protein, TRF2, is responsible for both the assembly and S-phase specific disassembly of the t-loop, which is necessary to prevent catastrophic t-loop resolution by the SLX1/4 nuclease complex. We demonstrate that the ability of TRF2 to coordinate t-loop disassembly strictly depends on its ability to bind and recruit RTEL1 to telomeres in S-phase. We show that the TRF2-RTEL1 interaction requires a previously uncharacterised metal-coordinating C4C4 motif in RTEL1. Genetic studies in mouse cells revealed that the C4C4 mutant is a classic separation of function mutation, which uncouples the two distinct functions for RTEL1 at telomeres in t-loop disassembly and suppression of telomere fragility: the C4C4 domain (mediates the RTEL1- TRF2 interaction) is required for t-loop unwinding but is dispensable for suppressing telomere fragility. Thus, the PIP-box and C4C4 motifs in RTEL1 convey distinct and separable functions, which impact on the targeting of RTEL1 to replication forks and telomeres, respectively. The clinical importance of our work was highlighted by the fact that the TRF2-RTEL1 interaction is abolished by mutations in RTEL1 that are causal for Hoyeraal-Hreidarsson syndrome (HHS), a severe form of Dyskeratosis congenita. Specifically, the RTEL1 pR1264H mutation is causal for HHS and has a carrier frequency of 1% within the Ashkenazi Orthodox Jewish population and 0.45% in the general Ashkenazi Jewish population. We established that the pR1264H mutation, which resides within the C4C4, specifically disrupts the TRF2-RTEL1 interaction. These findings have major implications for the understanding of telomere function, genome instability and human disease. Publications listed on page 174 Figure 1 PIH1D1 and its phospho-binding PIH domain mediated substrate recognition for assembly by the R2TP chaperon complex. Clare Hall 127 CHROMATIN STRUCTURE AND MOBILE DNA www.london-research-institute.org.uk/research/peter-cherepanov Group Leader Peter Cherepanov Postdoctoral Scientists Jun He Paul Lesbats Daniel Maskell Graduate Student Samual Dick Scientific Officers Nicola Cook Valerie Pye Our research focuses on the structural biology of chromatin function and its interactions with the retroviral DNA integration machinery. Using X-ray crystallography and complementary approaches we aim to elucidate three-dimensional structures and mechanisms of biological machineries involved in regulation of gene expression, DNA replication and retroviral integration. This year we made substantial headway in several of our main projects, including the crystallography of human Cdc7 kinase and of the archaeal Orc1-DNA complex, which are described below. We have also made progress towards understanding how the retroviral integration machinery interfaces with chromatin. Structure of the essential S-phase kinase Cdc7 Eukaryotic chromosomal DNA replication is initiated at multiple origins at the onset of and throughout S phase. The replisome assembly and initiation of DNA synthesis at individual origins critically depend on activities of S phase cyclindependent kinases and Cdc7. Both types of kinases are regulated by their respective activating subunits (cyclins and Dbf4, respectively). Phosphorylation of MCM2-7 by Cdc7 allows recruitment of essential replication initiation factors en route to the replisome assembly. Cdc7 is overexpressed in many cancers and tumour cell lines. Due to its pivotal role in cell proliferation, this S phase kinase is emerging as a target for the development of cancer therapeutics. Two years ago we reported the first crystal structure of Cdc7, which revealed how Dbf4 activates the kinase (Hughes et al., 2012; Nat Struct Mol Biol. 19(11): 1101-1107). However, the crystallised Cdc7-Dbf4 construct was only partially active, and the initial structure did not explain how the kinase recognises its target substrates. The observed drop in activity could be explained by mutations that had to be introduced into the kinase construct in order to obtain well-ordered crystals. Looking closely at the amino acid sequence of Cdc7, in particular at the regions that were affected by the mutations in our initial structure, we identified a cluster of highly conserved Cys residues abutting of the activation loop of Cdc7. The presence of this region was required to restore activity of our recombinant kinase preparations. Following extensive screening, we were able to crystallise a fully active heterodimeric Cdc7-Dbf4 construct and refined the 128 Scientific report 2014 LONDON RESEARCH INSTITUTE structure to a resolution of 1.4 Å. The structure revealed a novel zinc-binding domain within the kinase insert 2 that pins the beginning of the activation loop to the motif-M of Dbf4 and the C-lobe of Cdc7 (Figure 1A). Importantly, these interactions lead to complete ordering of the activation loop, which is consistent with the recovery of the kinase activity. The breakthrough enabled us for the first time to crystallise the kinase bound to a substrate peptide. The co-crystal structure containing an MCM2-derived peptide, refined to a resolution of 1.7 Å, explained the known specificity of the kinase for Ser or Thr residues followed by a negatively-charged residue (Glu, Asp, or phospho-Ser). The new structure also revealed an intriguing possibility that Cdc7 may be programmed to recognise sites primed by phosphorylation with CDK2, whose target site consensus includes an Arg or a Lys residue at the P+3 position. We are now focusing on the mechanisms of regulation of Cdc7 activity throughout the cell cycle. Our results will aid in the development of more potent and specific inhibitors of Cdc7 kinase. Structure of an archaeal Orc1 in the ATP-bound state The molecular machinery involved in the initiation of DNA replication in archaea shares many common features with that of eukaryotes. Archaeal chromosomes replicate from multiple origins, which are marked by binding of conserved Orc/Cdc6 AAA+ proteins that load the hexameric MCM DNA helicase. The archaeal Orc/Cdc6 proteins display extremely tight nucleotide binding, and are invariably isolated in their ADP-bound states. Their active ATP-bound forms Figure 1 A. Crystal structure of Cdc7 kinase with fully ordered activation loop. The activation loop and the zinc-binding domain found within the kinase insert 2 (KI2) are shown in orange. The rest of Cdc7 and Dbf4 (motifs C and M) are shown in green and blue, respectively. B. Crystal structure of Sulfolobus islandicus Orc1-1 bound to an ATP mimetic and cognate DNA (ORB). The winged helix domain (WHD) and the initiator specific motif (ISM, magenta) are indicated. Weighted 2Fo-Fc electron density map for the bound nucleotide and the associated metal atom is contoured at 2σ (blue chicken wire). A B are thought to be short-lived, likely contributing to the temporal control of initiation of DNA replication by restricting it to a very narrow time window. Following a single round of MCM loading and ATP hydrolysis, the initiator would remain in an inactive state until the next round of cell division, to be replaced by the de novo synthetised ATPbound form. The structural rearrangements within Orc/Cdc6 proteins associated with ATP hydrolysis are therefore of particular interest. Our goals in this project are to characterise structural rearrangements in archaeal Orc1 associated with ATP hydrolysis and to determine the structural basis for MCM loading by archaeal Orc/Cdc6 proteins. We were able to optimise production of the apo form of Sulfolobus islandicus Orc1-1, which allowed us to crystallise and determine the structure of this protein bound to an ATP mimetic and its cognate DNA element. The structure has been refined to a resolution of 2.7 Å, and the nucleotide is defined very well in the electron density map (Figure 1B). Intriguingly, the overall conformation of the protein in our structure is substantially different from those in previously reported ADP-bound forms. We are now investigating the functional consequences of the conformational rearrangements revealed by our crystal structure. Publications listed on page 174 Clare Hall 129 ARCHITECTURE AND DYNAMICS OF MACROMOLECULAR MACHINES www.london-research-institute.org.uk/research/alessandro-costa Group Leader Alessandro Costa Postdoctoral Scientists Panchali Goswami Jin Chuan Zhou Graduate Students Ferdos Abid Ali Paolo Swuec Scientific Officers Adelina Davies Julian Gannon Ludovic Renault Errors in the mechanisms that maintain gene copy number give rise to genomic instability, which is a hallmark of cancer cells. Our research aims to understand how the biological nanomachines involved in chromosome replication function to maintain genome integrity. To this end, we employ single-particle cryo-electron microscopy and biochemistry to study how DNA molecules are duplicated and how this process is coordinated with DNA repair and other nucleic acid transactions. By describing the architecture and dynamics of the DNA replication machinery and its interactors we seek to establish a molecular framework to explain how eukaryotic cells respond to DNA damage and how cell proliferation is regulated to avoid tumourigenesis. DNA replication – Structure of the translocating eukaryotic replicative helicase The Cdc45/Mcm2-7/GINS (CMG) helicase unwinds the DNA double helix during replication in eukaryotes. How the CMG is assembled and how it engages DNA substrates remains only partially understood. Using negative-stain electron microscopy, we have determined the structure of the CMG (Figure 1) in the presence of a slowly hydrolysable ATP analogue and a DNA duplex substrate with a 3’ single-stranded tail. The structure shows that the Mcm motor subunits of the CMG bind single- and not double-stranded DNA, supporting a steric exclusion mechanism for replication fork unwinding. We used biotinstreptavidin labelling to establish the polarity by which DNA enters into the Mcm2-7 channel, and elucidate how Cdc45 keeps the helicase topologically linked to the translocation strand during DNA unwinding. The Mcm2-7 motor Figure 1 Architecture of the Cdc45/ Mcm2-7/GINS (CMG) complex. Adapted from Costa et al., 2014; eLife 3: e03273. 130 Scientific report 2014 LONDON RESEARCH INSTITUTE subcomplex forms a right-handed spiral when DNA-bound, revealing unexpected similarities between the CMG and other hexameric ATPases such as the bacterial DnaB helicase and the Rpt1-6 AAA+ motor of the eukaryotic proteasome. We identified a subpopulation of dimeric CMGs, which allowed us to establish the subunit register of Mcm2-7 double hexamers assembled onto DNA before replication origin firing. Altogether, our results provide novel important insights into the nucleoprotein architecture of the replication fork. We are now interested in describing the detailed molecular mechanism of DNA translocation by the activated Mcm2-7 AAA+ motor. To this end, we have optimised preparations of substrate-bound CMG molecules embedded in vitreous ice for high-resolution cryo-electron microscopy. By imaging our nucleoprotein preparations using a direct electron detector on a 300 kV electron These findings establish the architectural framework for further mechanistic studies of the elongation step of DNA replication in eukaryotic cells (Figure 3). Figure 2 The Ctf4 helicase-polymerase link can bind up to three client proteins concomitantly. Adapted from Simon et al., 2014; Nature. 510(7504): 293-97. We are currently working on reconstituting a larger protein assembly, where Ctf4 is linked to the CMG helicase as well as the Pol α/primase holo-enzyme. Describing the structure and function of a full helicase-polymerase super-complex will help us elucidate the mechanism of coupling DNA unwinding and synthesis in the eukaryotic replisome. microscopy instrument, we aim at determining the near-atomic resolution structure of the translocating CMG helicase in various stages of the ATP hydrolysis cycle. Our results will inform us on the mechanism of ATPase cycling and nucleic acid translocation by a hetero-hexameric motor. DNA replication – Helicase/polymerase coupling Genome duplication requires tight coordination between parental duplex-DNA unwinding and daughter-strand synthesis within the replication machinery, to prevent the accumulation of vulnerable single-stranded DNA segments and the onset of genomic instability. We recently employed single-particle electron microscopy coupled with biochemistry and crystallography (in collaboration with Luca Pellegrini at the University of Cambridge), to describe the architecture of the Ctf4 ‘helicase-polymerase bridging factor’, either alone or bound to components of the CMG helicase and the DNA Polymerase α/primase assemblies (Figure 2). We showed that budding yeast Ctf4 forms a homo-trimeric disk, suggesting that it has the ability to link multiple factors at replication forks. Indeed, the Ctf4 trimer contains three docking sites that recognise a conserved motif mapping within the Pol α catalytic subunit as well as one of the four GINS subunits of the CMG helicase. Importantly, we showed that the Ctf4 trimer is capable of simultaneously binding to GINS and the amino terminus of the Pol α catalytic subunit. Figure 3 Speculative representation of the helicase-primosome super-assembly in the eukaryotic replisome. Homologous Recombination Double-strand breaks can be repaired by homologous recombination in eukaryotic cells. At the end of this process Holliday Junctions, covalent linkages between donor sequences, can be resolved by the action of a nuclease. This event results in the exchange of genetic information between two DNA segments, which can lead to the rise of deleterious mutations. To prevent this, eukaryotes have developed a strategy to dissolve homologous recombination intermediates back to their pre-recombination state (DNA-crossover suppression). The key player in this process is the four-member dissolvasome complex, comprising Topoisomerase IIIα, the RMI1/2 factors and the BLM helicase, whose mutation is linked to genetic disease and cancer development (reviewed in Swuec and Costa, 2014; Cell Biosc. 4: 36). While the mechanism of Holliday Junction dissolution is still unclear, a wealth of information is available on various orthologs of the dissolvasome assembly. For example, Topoisomerase IIIα belongs to the type-IA class of topoisomerases, pad-lock shaped enzymes that effect changes in DNA topology through a ‘strand-passage’ mechanism. RMI1 and RMI2, OB-fold containing factors, have been implicated in nucleic acid engagement or in protein-protein interactions. BLM contains a RecQ-type DNA helicase domain, whose ATPase function is required for DNA opening and translocation. Many issues remain unresolved and a mechanistic understanding of Holliday Junction dissolution is still lacking. Using an integrated approach that combines biochemistry, crystallography and electron microscopy we are working to determine the architecture of the dissolvasome complex, to elucidate the molecular basis of double Holliday junction dissolution. Publications listed on page 174 Clare Hall 131 CHROMOSOME REPLICATION www.london-research-institute.org.uk/research/john-diffley Group Leader John FX Diffley Postdoctoral Scientists Corella Casas-Delucchi Gideon Coster Max Douglas Belén Gómez-González Stephanie Hills Kerstin Kinkelin Christoph Kurat Joseph Yeeles Mona Yekezare Graduate Students Tom Deegan Mohammed Raihaan Hassan Jake Hill Agnieszka Janska Scientific Officers Dominik Boos Lucy Drury Anne Early Jordi Frigola Khalid Siddiqui We each synthesise roughly 5x1011 metres of DNA every day – more than the distance from the earth to the sun and back. Despite this scale, and the large number of tumour suppressor genes and potential oncogenes in our genomes, 2/3 of the UK population will live cancer-free lives. Thus, DNA replication and the quality control mechanisms associated with it are remarkably efficient. Once cells become cancerous, however, genome instability becomes the norm, which can drive intratumour heterogeneity and tumour evolution. Oncogenes induce replicative stress, but we still do not understand the nature of this stress. Our goal is to understand how DNA replication initiates and how it is controlled, which will help us understand how high fidelity DNA replication is ensured in normal cells. This is of fundamental importance for understanding cell proliferation, and is also a prerequisite to understanding and exploiting the subversion of this process in cancer. This year we made significant progress in understanding some of the key biochemical reactions that underpin this process. Roles for ATP hydrolysis in origin licensing DNA replication in eukaryotes initiates from multiple chromosomal locations termed origins, and the stability of the genome is dependent upon each origin firing once and only once per cell cycle. This is achieved by the temporal separation of replication initiation into two distinct steps. The first step, origin licensing, involves the loading of the hexameric MCM helicase comprising the six related Mcm2-7 subunits into pre-replicative complexes (pre-RCs). This occurs during late mitosis and G1 phase. The second step, origin firing, involves the conversion of the inactive MCM double hexamer into two functional replisomes during S phase. Origin licensing occurs in an ordered fashion. First, the Origin Recognition Complex (ORC) binds to origin DNA. Origin binding requires ORC binding to (but not hydrolising) ATP. Cdc6 is then recruited to form an ORC/Cdc6 complex on origin DNA, which requires ATP binding by Cdc6. The budding yeast MCM forms a complex with the Cdt1 protein and this MCM/Cdt1 complex is recruited to ORC/Cdc6 via an essential C-terminal domain in Mcm3. The loading of MCM into salt-resistant double hexamers bound around double stranded DNA requires ATP and is not supported by the ATP analogue ATPγS, indicating a requirement for ATP hydrolysis. Once Figure 1 Roles of ATP binding and hydrolysis in MCM loading. Details are described in the text. ATP C 132 Scientific report 2014 LONDON RESEARCH INSTITUTE MCM is loaded, it no longer requires ORC, Cdc6 or Cdt1 to maintain its origin association. Using purified yeast proteins, we systematically analysed the role of ATP binding and hydrolysis in origin licensing. We found that ORC and Cdc6 mutants defective in ATP hydrolysis are competent for origin licensing, even when combined. This has allowed us to uncover a novel role for ATP hydrolysis by Cdc6 in release of non-productive loading intermediates. Cdc6 mutants that cannot hydrolyse ATP are lethal in vivo, suggesting that this ‘proofreading’ activity is an essential function of Cdc6. Surprisingly, we found that ATP binding and hydrolysis by MCM subunits play distinct and essential roles during pre-RC assembly: ATP binding is required for stability of the MCM complex under licensing reaction conditions and ATP hydrolysis is required for MCM loading and Cdt1 release. These results are summarised in Figure 1. The MCM double hexamer is a precursor for DNA replication Upon entry into S phase, increase in the activities of cyclin dependent kinase (CDK) and Dbf4 dependent kinase (DDK) promotes the activation of the inactive MCM double hexamers. These kinases, together with other firing factors, including Sld2,3,7, Dpb11, GINS, Mcm10 and DNA polymerase ε, convert each inactive MCM double hexamer into two Cdc45/MCM/GINS (CMG) Figure 2 Bidirectional DNA replication in vitro. Replication reactions were sequentially labelled with BrdU and biotin-dUTP. Reactions were stopped and spread on microscope coverslips by molecular combing. BrdU (red) was detected with anti-BrdU; biotin (green) was detected with fluorescently labelled streptavidin. Several representative images are shown. complexes each containing a hexamer of MCM. To accomplish this, the double hexamer must separate, origin DNA must melt, the MCM ring must open, the lagging strand template must be extruded and the MCM ring, along with Cdc45 and GINS must close around the leading strand template. Some firing factors, including Dpb11 and the key CDK substrates Sld2 and Sld3, are required for initiation but not elongation, whilst others, like Cdc45, GINS and the leading strand DNA polymerase ε, are required for initiation and then form part of the elongation machinery. We have developed a soluble, cell-free DNA replication system to study the initiation of DNA replication from MCM double hexamers loaded with purified proteins. In this system, a subset of firing factors (Dpb11, Sld2, Sld3, Sld7 and Cdc45) was overexpressed from galactose-inducible promoters. These cells, which harbour a cdc7 temperature-sensitive mutation, were first synchronised in G1 phase with α-factor and then released from α-factor at 37°C, inducing arrest of cells in an S phase-like state. MCM double hexamers loaded onto plasmids with purified proteins were then incubated in this extract with labelled nucleotide and products examined by gel electrophoresis. DNA replication of plasmids in these extracts is semiconservative, requires pre-assembly of pre-RCs and results in the formation of fully replicated, covalently closed circular product. Moreover, as shown in Figure 2, replication is bidirectional. Consequently, factors required for elongation and termination do not appear to be lacking from this extract. Using a proteomic approach, all three replicative polymerases as well as virtually all of the other identified components of the previously described replisome progression complex were found specifically associated with template DNA in a DDK-dependent manner suggesting that our extracts assemble replisomes similar to those generated in vivo. Surprisingly few proteins other than known replisome components and firing factors were found associated in a DDK-dependent manner, suggesting that most or all core factors involved in initiation and elongation have been identified. If true, it should be possible to reconstitute this process with this set of purified proteins in the future. Publications listed on page 175 Clare Hall 133 MAMMALIAN DNA REPAIR www.london-research-institute.org.uk/research/peter-karran Group Leader Peter Karran Postdoctoral Scientist Elizabeth McAdam Graduate Students Melisa Guven Matt Peacock Scientific Officers Reto Brem Peter MacPherson Figure 1 Protein oxidation and NER inhibition by a fluoroquinolone and UVA. A. Protein carbonylation. CCRF-CEM cells were treated with ciprofloxacin for 1h at the concentrations shown. After washing, they were irradiated with UVA (20 kJ/m2). Cell extracts were treated with AlexaFluor 647 Maleimide to derivatise carbonyls. Proteins were separated by PAGE. Green fluorescence is from derivatised protein carbonyls. Red fluorescence is Sypro Ruby stain for total protein. B. NER inhibition. Extracts prepared from cells treated with ciprofloxacin and UVA (20 kJ/m2) as indicated were incubated with circular DNA substrates containing a single site-specific NER substrate lesion. NER activity is monitored by the formation of excision products (27-31 nt oligonucleotides) that are radiolabelled and separated by PAGE. 134 Therapy-related cancer is a significant clinical problem. The approximately 100-fold increased skin cancer incidence in immunosuppressed organ transplant patients is an extreme example of cancer risk associated with medication. Sun exposure is a cofactor in this risk. Previous work in the Mammalian DNA Repair Laboratory has linked the photochemical properties of azathioprine, one of the most widely-prescribed immunosuppressants, to skin cancer. Azathioprine causes skin photosensitivity. Photosensitivity is a common side-effect of drug treatment and is often associated with a known, or suspected, increased risk of skin cancer. Based on our extensive studies of the photochemical effects of azathioprine, the group is investigating the mechanisms by which other drugs cause photosensitivity and whether this might be related to an increased skin cancer risk. These studies have led us to examine how different wavelengths of solar radiation might contribute to the development of skin cancer. Photosensitisation and DNA repair Work from the group previously demonstrated that azathioprine causes photosensitivity by embedding the thiopurine 6-thioguanine (6-TG) in patients’ DNA. DNA 6-TG is a photosensitiser because, unlike normal DNA bases, it can absorb energy from the UVA in sunlight. (UVA is generally considered to be relatively harmless but it comprises ≥ 90% of the UV radiation that we are exposed to). The UVA energy absorbed by DNA 6-TG is transferred to oxygen to generate singlet A Scientific report 2014 LONDON RESEARCH INSTITUTE oxygen (1O2), a highly reactive and damaging form of oxygen. We have identified numerous forms of damage to DNA and protein caused by the 1O2 generated from the DNA 6-TG/UVA interaction. Cellular proteins are particularly vulnerable to damage by 1O2. This protein oxidation reduces the efficiency of DNA repair, including nucleotide excision repair (NER), the main protection against mutation and cancer in the skin. One particularly hazardous product of oxidation is the crosslinking of proteins to DNA. Working with the Protein Analysis and Proteomics Laboratory, Melisa Guven has used SILAC to investigate whether important DNA repair factors are sequestered by covalent linkage to DNA under the oxidative conditions generated by combined DNA 6-TG and UVA. This analysis reveals that key proteins from all the major DNA repair pathways (NER, DNA double strand break rejoining, DNA mismatch repair and base excision repair) become crosslinked to DNA under these conditions. Many commonly prescribed drugs are photosensitisers and UVA chromophores. Unlike 6-TG, most do not become incorporated into DNA. Nevertheless, their effects can mirror those of DNA 6-TG/UVA. Matt Peacock has shown that the 1O2 that is generated when cells are treated with fluoroquinolone antibiotics and irradiated with UVA causes extensive protein oxidation. UVA irradiation of fluoroquinolone-treated cells impairs their ability to remove canonical sunlight-induced B Figure 2 Effects of combined UVA on NER and mutation. A. NER in vivo. HaCaT cells were irradiated with UVC to induce canonical UV DNA photoproducts. Half the cells also received a non-toxic dose of UVA (100 kJ/m2). Excision of UVC-induced NER substrates (6-4 Py:Py photoproducts) was measuredby ELISA. B. NER in vivo. Extracts prepared from HeLa cells that had been irradiated with UVA (left panel) or approximately equitoxic doses of UVA, UVB or UVC (right panel) were assayed for the ability to carry out NER on a circular DNA substrate that contained a single NER. C. Mutation induction. The frequency of TK mutation was measured in TK6 cells irradiated with UVB and/or UVA as indicated. White bars are the observed mutation frequencies induced by combined UVA and UVB treatment. Red bars are the expected values for mutation frequency if UVA and UVB treatment were simply additive. Means ± SD of three determinations. A B C mutagenic DNA lesions by NER. Together with Peter MacPherson, Matt showed that the effect on NER reflects damage to proteins, and extracts of treated cells have low levels of NER activity (Figure 1). He also showed that damage to DNA repair proteins is not confined to NER; other DNA repair pathways are also inhibited by oxidation of participating proteins. Sunlight, protein oxidation and skin cancer Sun exposure is a known risk factor for skin cancer. This is because the UVB (wavelengths 280-320 nm) in sunlight damages DNA. UVB-introduced DNA lesions are firmly implicated in skin cancer development; skin tumour genomes are dominated by mutations that bear the hallmark of UVBinduced damage. Paradoxically, UVB phototherapy for chronic skin conditions, which involves multiple rounds of DNA-damaging UVB treatment, is not associated with an increased skin cancer incidence. This surprising observation suggests that the UVA (wavelengths 320-400 nm) radiation that comprises ≥ 95% of the UV in incident sunlight is important for mutation and skin cancer development. Although it is generally accepted that UVA does contribute to cancer risk, how it does this is not understood. To investigate how UVB and UVA radiation might interact to influence mutation and cancer risk, Lizzy McAdam is comparing the effects of UVB alone and in combination with UVA in cultured human cells. She has confirmed that UVA irradiation of cultured cells generates 1O2 and increases their levels of oxidised proteins (carbonyls). The increased protein carbonylation is associated with less efficient NER and the removal by NER of canonical UV-induced DNA lesions is slower in cells irradiated with UVA (Figure 2A). Extracts prepared from UVA-irradiated cells are less efficient at NER, implicating damage to NER proteins in the reduced repair efficiency. The effects are specific for UVA and exposure of cells to UVB or UVC radiation does not affect NER efficiency (Figure 2B). The effect of combining UVB and UVA radiation on mutation has been studied in the well-characterised TK6 mutation system that detects mutational inactivation of the single TK locus in a cultured human cell line. Measurements of UVB and UVA-induced TK mutations in TK6 cells suggest that their mutagenic effects are partly synergistic. When cells are irradiated with a low dose of UVB together with UVA, the frequency of induced TK mutations is higher than the frequency that would be expected if UVA and UVB were contributing separately to mutation induction (Figure 2C). The effect of protein oxidation on human genome stability has not been extensively studied. In simpler organisms, protein oxidation is an important influence on survival and mutation. Certain extremophiles are protected against the lethal and mutagenic effects of radiation by highly efficient antioxidant systems that prevent protein oxidation. In addition, there are indications that the level of protein carbonyls directly influences both spontaneous and induced mutation in bacteria. Our findings are compatible with a similar relationship in human skin. They suggest that sunlight UVA contributes to skin cancer risk by increasing the level of protein oxidation. Because they point to an important role for UVA in mutation by sunlight, these findings also have implications for recommendations for appropriate sunscreen formulation. Publications listed on page 175 Clare Hall 135 CELL DIVISION AND ANEUPLOIDY www.london-research-institute.org.uk/research/mark-petronczki Graduate Students Anna Dowbaj Kristyna Kotynková Scientific Officer Tohru Takaki Figure 1 Depletion of splicing factors abrogates sister chromatid cohesion and the stable association of cohesion with chromatin. A. Transfection of HeLa Kyoto cells with siRNAs against splicing factors leads to a premature loss of sister chromatid cohesion. Images of DAPI stained chromosome spreads from cells reveal that, while chromosomes from control depleted cells display the characteristic ‘X’ shape, cells depleted of splicing factors have lost the connections between sister chromatids. B. Fluorescence recovery after photobleaching (FRAP) in HeLa Kyoto cells stably expressing a core cohesin subunit, SMC1-EGFP, reveal enhanced dissociation of cohesin from chromatin in MFAP1- depleted cells. The nuclear SMC1-EGFP fluorescence was repeatedly bleached in approximately half of the nucleus and the loss of fluorescent intensity in the unbleached area was plotted over time. These experiments demonstrate that depletion of splicing factors weakened the stability of cohesion-chromatin interaction in interphase cells. 136 The process of cell division is indispensable for life to flourish and diversify. It ensures equal segregation of cellular content including DNA to the two nascent daughter cells. Accurate segregation of chromosomes in eukaryotic organisms relies on connections between replicated sister chromatids, a phenomenon known as sister chromatid cohesion. Sister chromatid cohesion is mediated by a conserved ring-like protein complex, cohesin, that topologically entraps the sister chromatids until they are ready to be segregated. Cohesin lies at the heart of chromosome biology. It is essential for chromosome segregation, the proper repair of DNA lesions and important for gene regulation. Recent cancer genome studies have concluded that cohesin subunits are frequently mutated in human malignancies. Defects in cohesin function can give rise to aneuploidy and genome instability and thereby contribute to tumourigenesis in A A somatic cells or developmental defects and infertility in the germline. Using available functional genomic and proteomic data sets along with a focused RNAi approach, we identified a set of factors involved in pre-mRNA splicing whose depletion prevented successful cell division in human cells. Loss of these splicing factors abrogated chromosome alignment and trapped cells in mitosis. This mitotic phenotype was accompanied by a dramatic loss of sister chromatid cohesion that occurred soon after DNA replication (Figure 1A). While depletion of premRNA splicing mediators had no striking effect on bulk loading of cohesin onto chromatin, fluorescence recovery after photobleaching experiments (FRAP) revealed that depletion of splicing factors prevented the stable association of cohesin with chromatin in post-replicative cells (Figure 1B). Splicing factor depletion Control siRNA MFAP1 siRNA 14% B B SART1 siRNA NHP2L1 siRNA 98% 99% % spreads with split sister chromatids 81% Time (min) SMC1-EGFP Control Postdoctoral Scientists Laurent L’Epicier-Sansregret Murielle Serres Antonio Tedeschi Maria Dolores Vázquez Novelle 0 siRNA Mark Petronczki MFAP1 Group Leader Every second several hundred thousand cells in our body duplicate themselves through a process known as cell division. To generate healthy and viable cells the division process has to accurately partition all 46 chromosomes to daughter cells. Our group uses animal cell systems to investigate the molecular mechanisms underlying cell division and the consequences of genomic imbalances caused by cell division errors. We have recently discovered that pre-mRNA splicing is essential for sister chromatid cohesion in human cells (Sundaramoorthy et al., 2014; EMBO J. 33(22): 2623-2642). Photobleach Scientific report 2014 LONDON RESEARCH INSTITUTE 2 10 30 60 120 Figure 2 Model for the role of splicing factors in Sororin-mediated sister chromatid cohesion. In control cells (left), pre-mRNA splicing of Sororin ensures the presence of functional Sororin protein at the S phase of every cell cycle to counteract the destabilising activity of WAPL. Compromised splicing (right) leads to a sharp decline in levels of Sororin protein. The lack of Sororin, a factor necessary for cohesion maintenance leads to a reduced association between cohesin and chromatin and thereby to a premature loss of sister chromatid cohesion. Immunoblotting revealed that the depletion of splicing factors caused a 5-fold reduction in the protein levels of Sororin, a protein required for stable association of cohesin with chromatin. Bolstering our observation, erroneous splicing of Sororin pre-mRNA was detected upon depletion of splicing factors. Importantly, the sister chromatid cohesion loss caused by depletion of splicing factors could be suppressed by a Sororin transgene that does not depend on pre-mRNA splicing for its expression. Furthermore, we found that depletion of the cohesion release factor WAPL that is antagonised by Sororin also restores sister chromatid cohesion in cells lacking spliceosome components. Thus, the pre-mRNA splicing of Sororin is a rate-limiting step in the maintenance of sister chromatid cohesion in human cells (Figure 2). Our results highlight the loss of cohesion as an early cellular consequence of compromised splicing. Our work linking splicing and sister chromatid cohesion has potential implications for the pathology of Chronic Lymphocytic Leukemia (CLL). One of the splicing factors that we implicate in sister chromatid cohesion, SF3B1, is a gene that is one of the most frequently mutated genetic drivers found in CLL patients. Publications listed on page 175 Clare Hall 137 MECHANISMs OF GENE TRANSCRIPTION www.london-research-institute.org.uk/research/jesper-svejstrup Group Leader Jesper Q Svejstrup Postdoctoral Scientists Stefan Boeing Andreas Ehrensberger Lea Gregersen Theo Kantidakis Michael Ranes Marco Saponaro Yuming Wang Hannah Williams Laura Williamson Graduate Students Konstantin Fritz Michael Lim Kotryna Temcinaite Diana Zatreanu Scientific Officers Barbara Dirac-Svejstrup Michelle Harreman Jane Walker Maintaining genome integrity is of utmost importance for the longterm survival of cells and organisms. However, the key immediate cellular response to genotoxic insult is arguably to maintain gene expression. Indeed, without transcription, cells cannot proceed through the cell cycle, and even non-dividing cells will perish. The overall aim of our research is to understand the basic mechanisms underlying RNA polymerase II (RNAPII) transcript elongation, but in particular how elongation interfaces with other processes on DNA, such as DNA repair, replication and recombination. We also investigate the cellular and molecular consequences of transcription stress, such as that caused by DNA damage. We believe that a detailed insight into the cellular responses to transcription elongation impediments will make it possible for us to understand certain human diseases, and cancer in particular, and thereby eventually how to treat them. We use biochemical, genetic, and cell biological approaches, often combined with modern ‘omics’ technologies to understand the process of transcript elongation and its interface with other DNA-related processes such as DNA replication and repair. Our published work in 2014 included studies of the tumour suppressor RECQL5 and its effect on transcription stress, and on Cockayne Syndrome B (CSB) – the basis for Cockayne syndrome (CS). RECQL5 suppresses the genome destablising effects of transcription stress It has become increasingly evident that transcription is closely integrated with other DNA-related processes, such as DNA replication and repair. Indeed, the process of expressing genes comes at a cost: the movement of RNA polymerases through DNA is associated with genome instability (Helmrich et al., 2013; Nat Struct Mol Biol. 20(4): 412-418), and RNAPII stalling, pausing, arrest, and/or backtracking (collectively referred to as transcription stress) generates a cellular response akin to the DNA damage response (Wilson et al., 2013; Biochem Biophys Acta. 1829(1): 151-157). Transcribing polymerases are also potent modulators of DNA-related processes such as DNA replication. Indeed, transcription-associated DNA recombination involves clashes between transcription and replication, and transcription is 138 Scientific report 2014 LONDON RESEARCH INSTITUTE associated with mutagenesis and contraction of CAG repeats, as well as breaks at chromosome fragile sites (Helmrich et al., 2013). However, the mechanisms underlying transcription-associated genome instability remain largely obscure, and little is known about factors that might have evolved to counteract it. The RECQ proteins constitute a family of conserved DNA helicases that are important for maintaining genome stability from bacteria to humans (Chu and Hickson, 2009; Nat Rev Cancer. 9(9): 644-654). RECQL5 is unique among this family by interacting with RNAPII, and it harbours two RNAPII interaction domains which are relevant for the genome instability phenotypes of cells lacking RECQL5 (Aygün and Svejstrup, 2010; DNA Repair (Amst). 9(3): 345-353). Intriguingly, transcription reactions reconstituted with pure transcription factors and RNAPII suggested that RECQL5 acts as an inhibitor of transcription (Aygün et al., 2009; J Biol Chem. 284(35): 23197-23203). However, the physiological relevance, if any, of this observation was not investigated. Likewise, the mechanistic foundation for the well-known destabilising effects of RECQL5 mutation on genome integrity has also remained unclear. We investigated the effect of RECQL5 on transcript elongation and its role in suppressing genome instability (Saponaro et al., 2014; Cell. 157(5): Figure 1 Model for RECQL5 function. RECQL5 ‘dampens’ transcript elongation causing a slow down of overall average elongation rates while simultaneously avoiding excessive transcription stalling and arrest (i.e. transcription stress), which can otherwise lead to genome instability and, eventually, to cancer. 1037-1049). Our results indicate that RECQL5 is a general elongation factor, important for preserving genome stability during transcription. Depletion or overexpression of RECQL5 resulted in corresponding shifts in the genome-wide RNAPII density profile. Transcript elongation was particularly affected across most genes, with RECQL5 depletion causing a striking increase in the average elongation rate. Concurrently, increased transcription stress was observed in coding regions, together indicating that RECQL5 controls the movement of RNAPII across genes. Importantly, loss of RECQL5 also resulted in the loss or gain of genomic regions, with the breakpoints of lost regions located in genes and common fragile sites. Intriguingly, the chromosomal breakpoints overlapped with areas of elevated RNAPII transcription stress, suggesting that RECQL5 suppresses the detrimental effects of such stress, and thereby prevents genome instability in the transcribed region of genes (Saponaro et al., 2014). The molecular basis of Cockayne syndrome CS is an autosomal-recessive, multi-system disorder, characterised by severe neurologic disease, growth failure, developmental abnormalities, photosensitivity, and degeneration of different organ systems (Brooks, 2013; DNA Repair (Amst.). 12(8): 656-671). The majority of CS patients carry mutations in the gene encoding the DNA translocase CSB/ERCC6 (~80% of patients), or the gene encoding ubiquitin ligase-associated CSA/ ERCC8. These proteins are best known for their role in transcription-coupled nucleotide excision repair (TC-NER), a process whereby bulky DNA lesions, such as those generated by UV-irradiation, are preferentially removed from the transcribed strand of active genes. CS is thus frequently referred to as a TC-NER disease. However, CS cells are sensitive to a number of additional DNA-damaging agents, and oxidative damage in particular, implicating the CS proteins in other repair pathways as well. Indeed, the idea that CS results from inefficient repair of oxidative DNA damage has gained momentum over the last decade. Finally, studies from Weiner, Egly and other groups have reported evidence of a role for CSB in gene regulation, which might provide an alternative explanation for CS (Brooks, 2013). However, the relationship between deficiencies in molecular pathways affected by CS mutation and patient disease symptoms have generally remained tenuous, or unexplored. We investigated the connection between the neuropathology of CS and dysregulation of gene expression (Wang et al., 2014; Proc Natl Acad Sci USA. 111(40): 14454-14459). Transcriptome analysis of human fibroblasts revealed that, even in the absence of DNA damage, CSB affects the expression of thousands of genes, many of which are neuronal genes. CSB is present at a significant subset of these genes, suggesting that regulation is direct. Importantly, cellular reprogramming of CS fibroblasts to neuron-like cells was defective unless an exogenous CSB gene was introduced. Moreover, neuroblastoma cells from which CSB was depleted showed defects in gene expression programmes required for neuronal differentiation and failed to differentiate and extend neurites. Likewise, neuron-like cells could not be maintained without CSB. Finally, a number of disease symptoms may be explained by the marked gene expression changes observed in the brains of diseased CS patients. Together, our data point to dysregulation of gene regulatory networks, rather than DNA repair defects, as the cause of the neurological symptoms in CS (Wang et al., 2014). Publications listed on page 175 Clare Hall 139 GENETIC RECOMBINATION www.london-research-institute.org.uk/research/stephen-west Group Leader Stephen C West Postdoctoral Scientists Gary Chan Miguel Gonzalez Blanco Gregoriy Dokshin Kasper Fugger Maria Jose Martin Pereira Marieke Peuscher Kanagaraj Radhakrishnan Shriparna Sarbajna Joanna Soroka Haley Wyatt Graduate Student Kristyna Kotynkova Scientific Officers Michael McIlwraith Rajvee Shah Mammalian cells possess a large repertoire of DNA repair processes that maintain the integrity of our genetic material. Some individuals, however, carry mutations in genes required for DNA repair, and this often leads to inheritable disease. An important repair process involves recombination, and defects in this process are linked with cancer predisposition, in particular breast cancers caused by mutation of the BRCA2 gene, acute leukemias associated with Fanconi Anemia, and a wide range of cancers found in individuals with the chromosome instability disorder known as Bloom’s Syndrome. The focus of our research is to determine the molecular mechanisms of recombinational repair, and to define why defects in these processes cause cancer. Our genetic material (DNA) is continually subjected to damage, either from endogenous sources such as reactive oxygen species produced as byproducts of oxidative metabolism, from the breakdown of replication forks during cell growth, or by agents in the environment such as ionising radiation or carcinogenic chemicals. To cope with such damage, cells employ elaborate and effective repair processes that are each specialised to recognise different types of lesions in DNA. These repair systems are essential for the maintenance of genome integrity. Some individuals, however, are genetically predisposed to crippling diseases or cancers that are the direct result of mutations in genes involved in the DNA damage response. The breast cancer tumour suppressor BRCA2 For several years we have been interested in the mechanisms of homologous recombination, how they contribute to the repair of DNA double-strand breaks, and how they promote genome stability. Many of the proteins required for recombination have been purified in this laboratory, and we use biochemical and molecular and cell biological approaches to understand how they bring about the repair of DNA breaks. One of these proteins is the BRCA2 breast cancer tumour suppressor. Mutations in the BRCA2 gene lead to a greatly increased risk of breast and ovarian cancers, and biallelic BRCA2 mutations have been associated with the cancer-predisposition syndrome Fanconi Anemia (FA) sub-type FA-D1. We therefore want to understand the precise role of the product of this gene, the BRCA2 tumour suppressor, in DNA repair 140 Scientific report 2014 LONDON RESEARCH INSTITUTE mediated by recombination. Previous work from our group has shown that BRCA2 is required for the assembly of nucleoprotein filaments formed by the RAD51 recombinase, and in particular for the delivery of RAD51 to sites of DNA damage. But until recently the exact mechanism of how BRCA2 coordinates the RAD51-mediated steps of recombinational repair remained elusive. In new work, we obtained the first structural and mechanistic insights into the role that BRCA2 plays in promoting RAD51 nucleoprotein filament formation. In collaboration with Professor Xiaodong Zhang’s group at Imperial College London, we visualised full-length BRCA2 protein and the BRCA2-RAD51 complex using negative stain electron microscopy combined with single particle techniques to generate a threedimensional (3D) reconstruction of the protein’s structure (Shahid et al., 2014; Nat Struct Mol Biol. 21(11): 962-968). We found that BRCA2 forms dimers and that single-stranded DNA (ssDNA) binds across long axis of the dimeric protein. Our structural analysis of the BRCA2-RAD51 complex revealed that BRCA2 remains in a dimeric form upon binding to RAD51, and delivers RAD51 to the ssDNA thereby increasing the number of RAD51 filaments. Two 3D views of BRCA2 modelled into the structure of the BRCA2-RAD51 complex are shown in Figure 1. Further analysis of BRCA2RAD51-ssDNA complexes revealed that BRCA2 is present at one end of the RAD51 filament and directs unidirectional growth of the filaments along the ssDNA. In essence, it acts as a molecular chaperone. Together, our data has uncovered novel molecular insights into the mechanistic aspects of BRCA2 action in homologous recombination (HR) and have potential implications for designing more efficient therapeutic intervention of cancer. Figure 1 Two views of the BRCA2 dimer (the two subunits are coloured yellow and cyan) modelled into the BRCA2-RAD51 complex (pink mesh). Four RAD51 monomers are shown on each side of BRCA2. This BRCA2-RAD51 complex plays a key role in the initiation of DNA repair by recombination. Structure-selective nucleases resolve chromatid bridges to ensure chromatid segregation at mitosis Recombinational repair requires a reciprocal exchange of DNA strands between sister chromatids or homologous chromosomes, leading to the formation of DNA intermediates, such as Holliday junctions (HJs), in which the two interacting DNAs become covalently interlinked. The efficient processing of these joint molecules is essential for chromosome segregation at cell division, and is also important in determining the outcome of recombination: for example, crossovers (COs) between homologous chromosomes are required for meiotic division, whereas non-crossovers (NCOs) are favoured in mitotic cells in order to avoid ‘loss of heterozygosity’ (LOH), a known driver of tumourigenesis. The importance that the cell places on the efficient and timely processing of these intermediates is clear from our discovery of the involvement of three distinct pathways of HJ resolution, and the way that these pathways are regulated throughout Figure 2 The sub-cellular localisation of the GEN1 Holliday junction resolvase is directed by its nuclear export sequence (NES). In normal cells, GEN1 is cytoplasmic and can only resolve recombination intermediates when the nuclear envelope breaks down at mitosis. Mutation of the NES can be used to target GEN1 into the nucleus, but its presence causes increased sister chromatid exchanges (SCEs). The cytoplasmic localisation is therefore important to control the nuclease, and yet facilitate chromosome segregation in mitosis. the cell cycle (Matos et al., 2011; Cell. 147(1): 158-172, Wechsler et al., 2011; Nature. 471(7340): 642-646, Sarbajna et al., 2014; Genes Dev. 28(10): 1124-1136). Firstly, the human BTR complex (BLM-TOPIIIα-RMI1-RMI2) plays a major role in the removal of double HJs to generate NCO products. The preferential formation of NCOs is important to avoid LOH. Secondly, at G2/M phase of the cell cycle, our cells create a novel endonuclease that is capable of resolving persistent recombination intermediates that have escaped the attention of BTR. This nuclease, which forms in response to CDK/PLK1-dependent phosphorylation events, involves interactions between two structureselective endonucleases, MUS81-EME1 and SLX1-SLX4 (Wyatt et al., 2013; Mol Cell. 52(2): 234-247). Finally, at mitosis, any remaining intermediates are acted upon by GEN1, a classical Holiday resolvase (Chan et al., 2014; Nat Comm. 5: 4844). The MUS81-EME1-SLX1-SLX4 and GEN1 nucleases are potentially dangerous in S phase, since they generate COs (which can be measured by an increased frequency of sister chromatid exchanges) providing a good rationale why their activities are held in check until late in the cell cycle. Our recent work uncovered the mechanisms by which GEN1 (and it’s yeast ortholog Yen1) are regulated throughout the cell cycle in order to avoid CO formation and LOH. In yeast, Yen1 is held in an inactive state in the cytoplasm by virtue of its phosphorylation by Cdk. During mitosis, however, the protein is dephosphorylated by Cdc14, leading to its nuclear entry and activation (Blanco and West, 2014; Mol Cell. 54(1): 94-106). In human cells, the situation is similar, although mechanistically different from that in yeast: GEN1 is exported to the cytoplasm by a strong Nuclear Export Sequence (NES), and only gains access to the chromosomal DNA upon nuclear envelope breakdown at mitosis (Chan and West, 2014; Nat Commun. 5: 4844). Artificial localisation into the nucleus (by NES mutation and addition of nuclear localisation sequences) leads to inappropriate cleavage of replication forks and causes increased sister chromosome exchange (SCE) formation (Figure 2). The regulation of HJ processing pathways is therefore important for the maintenance of genome stability, both by limiting CO formation and for ensuring proper chromosome segregation. Publications listed on page 176 Clare Hall 141 SINGLE MOLECULE IMAGING www.london-research-institute.org.uk/research/hasan-yardimci Group Leader Hasan Yardimci Postdoctoral Scientists Daniel Burnham Melania Strycharska Graduate Student Hazal Busra Kose Scientific Officers Sherry Xie Sevim Yardimci Before a cell divides it has to duplicate its genome so that two identical copies of its DNA content can be partitioned into daughter cells. In eukaryotic cells, replication is initiated at thousands of origins on the DNA, each resulting in the assembly of two replisomes that travel away from the initiation site in opposite directions. Complete and high-fidelity duplication of the genome is essential for faithful transmission of genetic information. When DNA replication goes awry, the result could be cells with mutations, missing or extra genetic material, a hallmark of the genomic instability seen in most cancers. Our group aims to investigate processes involved in eukaryotic replication using a combination of conventional biochemistry and single-molecule imaging tools. Helicase mechanisms To copy their DNA in preparation for cell division, cells must separate the two strands of the double helix. All cells contain a ring-shaped hexameric DNA helicase, which performs this task. Bacteria use DnaB, whereas eukaryotes use the MCM2-7 complex. In G1 phase, two MCM2-7 hexamers assemble around double-stranded DNA (dsDNA) at each origin of replication in a head-to-head fashion (Figure 1). This structure, known as the prereplication complex (pre-RC), remains idle in the G1 phase. In S phase, MCMs are activated through the action of a number of proteins including Cdc45 and GINS, and multiple kinases to subsequently unwind DNA at the replication fork. Figure 1 Eukaryotic DNA replication. In G1 phase, double hexamers of the MCM2-7 complex are loaded onto origin DNA by ORC, Cdc6, and Cdt1. MCMs are activated in S phase via the action of a number of factors to initiate unwinding. Finally, polymerases replicate unwound DNA. 142 Scientific report 2014 LONDON RESEARCH INSTITUTE To understand the molecular mechanism by which MCM2-7 functions, we developed a single molecule assay in which a DNA template immobilised on the surface of a microfluidic flow cell is efficiently replicated in soluble extracts derived from Xenopus laevis eggs (Figure 2). Using this system, we showed that MCM2-7 complexes, which initially load as double hexamers on DNA, can physically uncouple and function as single hexamers during replication (Yardimci et al., 2012; Mol Cell. 40(5): 834-840), in contrast to some models. We also showed that MCM2-7 translocates along the leading strand template to unwind DNA, suggesting that the helicase goes through a conformational change during activation and transitions from a dsDNA to a single-stranded DNA (ssDNA) binding mode (Fu et al., 2011; Cell. 146(6): 931-941). In the future, we aim to gain an in depth understanding of the MCM2-7 dynamics through real-time visualisation of the helicase at the single molecular level. Simian Virus 40 (SV40), a mammalian DNA tumour virus, has served as a robust model system for investigating the mechanism of eukaryotic replication for several decades. The virus encodes its own replicative helicase, Large T-antigen (T-ag), which utilises host cell factors for replication of its genome. Our work indicated that T-ag activated at an origin functions as a single hexamer and translocates along ssDNA, similar to the MCM2-7 complex (Yardimci et al., 2012; Nature. 492(7428): 205-209). Importantly, we also discovered a surprising new property of T-ag. We found that T-ag can efficiently bypass a protein adduct covalently cross-linked to the translocation strand. Figure 2 Single molecule visualisation of eukaryotic replication. λ DNA was stretched and immobilised at both 3’ ends on the streptavidin-functionalised surface of a microfluidic flow cell. Immobilised DNA was exposed to Xenopus egg extracts to initiate MCM2-7-dependent replication. Finally, a second extract containing digoxigenin-modified dUTP (dig-dUTP) was withdrawn into the flow cell to confirm bidirectional replication. Extracts were removed via SDS containing buffer, dsDNA was labelled with SYTOX Orange and dig-dUTP was labelled with a fluorescent anti-dig antibody. A stretched λ DNA molecule that underwent replication in extracts (bottom). High intensity SYTOX tract corresponds to a replication bubble. Anti-dig tracts coincide with both ends of the bubble indicating bidirectional replication. This remarkable plasticity of T-ag may help the SV40 replisome overcome bulky barriers such as DNA-protein cross-links. Current work is focused on defining the molecular mechanism by which a ring-shaped hexamer can overcome large obstacles during translocation. Architecture and dynamics of the eukaryotic replication machinery An essential component of the replisome complex is the polymerase, which synthesises new DNA on unwound strands. First, a primase complex associates with unwound DNA and synthesises DNA/RNA primers. At the leading strand, the DNA primer is extended continuously the lagging strand is synthesised discontinuously as Okazaki fragments (Figure 1). It is important to understand how different polymerases coordinate DNA synthesis for accurate replication. Live cell imaging in bacteria showed that a single replisome contains three polymerases, one acting on the leading strand and two on the lagging as opposed to the previous assumption that there is one polymerase acting on each strand. The presence of two lagging strand polymerases was shown to be important for processive lagging strand synthesis. Unlike prokaryotes, eukaryotes employ different polymerases to synthesise leading and lagging strands. Upon priming by polymerase alphaprimase complex (Pol α), the leading strand is replicated by polymerase epsilon (Pol ε) while the lagging strand is replicated by the action of polymerase delta (Pol δ) (Figure 1). Currently, little is known about the stoichiometry and dynamics of eukaryotic replisome components including polymerases. How long does Pol α remain on DNA before Pol ε or Pol δ takes over? How many Pol ε and Pol δ molecules are associated with individual replisomes? How often do polymerases exchange at the fork while synthesising the leading and lagging strands? To address these questions, we will visualise individual molecules in real time during replication of stretched DNA molecules. Our work will also provide important insight into how the replication machinery acts upon encountering different types of DNA damage. Publications listed on page 176 Clare Hall 143 False coloured SEM of a human blood sample showing red blood cells and a single white blood cell surrounded by a mesh of fibrin. Image: Electron Microscopy, Technology Core Facility, LRI. 144 Scientific report 2014 LONDON RESEARCH INSTITUTE TECHNOLOGY CORE FACILITIES The London Research Institute benefits from access to a wide range of high quality research services. Scientific support for researchers at the LRI is provided by some nineteen core facility groups of various sizes run by the Institute. These aim to provide state of the art facilities for LRI researchers that are proactive in enabling the research groups to carry out world leading science. TECHNOLOGY CORE FACILITIES 145 The London Research Institute has benefited from access to a wide range of high quality research services. Scientific support for researchers at the LRI is provided by core facility laboratories of various sizes run by the Institute. These aim to provide state of the art facilities for LRI researchers that are proactive in enabling the research groups to carry out world leading science. Julian Downward Associate Director, LRI The past year has seen the final details of the planning of the space for the core facility labs at the Francis Crick Institute, where they will be named Science Technology Platforms (STPs). Plans have been made for combining the core facilities from the LRI and NIMR and transition leads for each facility were appointed in the spring. The move of STPs to the Crick building will start at the end of 2015 and will be arranged to minimise disruption to users during the transition period. At Crick we will maintain and build on the fantastic quality of the LRI core facilities, providing researchers with access to unparalleled technologies to pursue outstanding science. The quality and development of LRI core facilities has been driven by user committees, made up of representatives of the service provider, users and management, which provide advice on technical advances, prioritise projects when facilities are limited, and act as a focal point for interactions with researchers. In addition, a programme of review by external experts every three to four years has ensured that the LRI core technology facilities have remained cutting edge. Similar mechanisms will be put in place at the Crick to ensure the continued excellence of the STPs. Technology core facilities provided centrally at the LRI allow access to cutting edge equipment and instruction in its correct and effective usage. Within these facilities, services may either be run by dedicated staff or by researchers themselves with appropriate training. LRI core facilities include the following: Light Microscopy provides conventional, confocal, multiphoton and automated microscopy, time-lapse video and microinjection services. Electron Microscopy has field emission and serial block face scanning electron microscopes and transmission electron microscopes. The FACS Facility provides a comprehensive flow cytometry service, with several analysers and sorters, including a FlowSight imaging flow cytometer. Experimental Histopathology provide expertise in mouse and human histopathology, in situ 146 Scientific report 2014 LONDON RESEARCH INSTITUTE hybridisation techniques, laser capture microdissection and automated image acquisition. The Equipment Park provides DNA Sanger sequencing, robotic nucleic acid preparation, quantitative PCR, gel imaging systems and HPLC micro-purification. Advanced Sequencing provides next generation DNA sequencing with three Illumina HiSEQ 2000s and an Ion Torrent PGM. The High Throughput Screening Facility brings together the equipment, personnel and expertise needed to carry out and interpret large scale screening assays. It has recently acquired an Echo acoustic dispenser, enabling accurate automated delivery of nanolitre volumes. The Protein Purification Facility specialises in the production of pure recombinant proteins for structural studies, using baculoviral, bacterial and mammalian tissue culture systems. Bioinformatics and Biostatistics provide support for all the Institute’s bioinformatics needs, ranging from high throughput sequencing data analysis and high throughput screen interpretation to global gene expression analysis. Cell Services provides a wide range of quality controlled cells and media, as well as the production of monoclonal antibodies from hybridoma lines. The Peptide Chemistry Facility provides custom made peptides, peptide arrays and cross-linking reagents. Transgenics provide the latest methodology for the generation of genetically modified mice. The In Vivo Imaging facility was established over the past five years by Francois Lassailly to support visualisation of animal cancer models by high resolution ultrasound, micro CT, bioluminescence imaging and intravital multiphoton microscopy. The whole institute has been deeply saddened by the sudden and unexpected death of Francois in January 2015. We will all hugely miss his inspirational leadership, intelligence and charm. After nine years of overseeing the core technology facilities at LRI, I look forward to helping to establish the Science Technology Platforms at the Crick as world leading in their fields. ADVANCED SEQUENCING www.london-research-institute.org.uk/technologies/advanced-sequencing The past five years saw a significant increase in Next Generation Sequencing (NGS) activity at the LRI. The number of samples being analysed per project has increased from tens to thousands in some cases, supported by continued investment in technologies and protocols. Head Nik Matthews Staff Sharmin Begum Jennifer Biggs Ben Phillimore Adam Rabinowitz Figure 1 HiSeq sequencing report (near perfect) for a 101x2bp Paired End run with 7bp of index sequence in the centre. The figure shows the flowcell representation on the left. The other four diagrams are the cycle by cycle intensities (top left), cluster densities per lane (bottom left), passed filter Q-scored (top right) and Q-score heat map (bottom right) for the run. The Advanced Sequencing Facility (ASF) has three Illumina HiSeq systems and one Ion Torrent PGM system. As early adopters of these technologies the ASF has pushed the limits of the systems to include low concentration samples (down to single cell) and fragmented RNA. The facility offers four main methodologies (each with multiple protocols): – ChIP-seq – Paired End sequencing (e.g. whole genome, amplicon-seq) – RNA-seq (mRNA and total RNA) – Bait Targeted Enrichment ChIP-seq ChIP-seq is the evolution of ChIP-ChIP and involves the study of DNA-associated proteins. This technique can be used to study heritable information and epigenetic patterns in cancer cells and help validate RNA-seq experiments. This methodology includes also 4C and ChIA-PET. Paired End sequencing Paired End sequencing on the HiSeq is a method that makes it possible to sequence genomes of all model organisms to a high coverage. The HiSeq can be used to sequence these model organisms genomes, including human, to a level of accuracy essential to the study of rare variants. RNA-seq Unlike the genome, which is rather static apart from mutations, the transcriptome is very fluid and can change with internal and/or external conditions. An RNA-seq method can look simply at these expressed genes or more in-depth at e.g. splice variants. Other aspects of RNA biology can also be studied using shRNA-seq, RIP-seq and GRO-seq. Bait Target Enrichment Bait Target Enrichment is similar in many ways to amplicon-seq but is not a PCR method. Target enrichment is a method of pulling out specific contiguous or non-contiguous areas within any genome, e.g. exome, using complimentary oligo baits. This is a good alternative to larger whole genome sequencing as it allows reduction in costs and machine time. Figure 1 shows a whole exome targeted enrichment HiSeq sequencing report. In the time the ASF has worked at the LRI we have worked alongside some great PhD students, postdocs and group leaders on some fantastic studies and publications. We look forward to this continuing at the Crick. Publications listed on page 177 TECHNOLOGY CORE FACILITIES 147 BIOINFOMATICS AND BIOSTATISTICS www.london-research-institute.org.uk/technologies/bioinfomatics-and-biostatistics Head Aengus Stewart Staff Probir Chakravarty Philip East Mickael Escudero Stuart Horswell Gavin Kelly Anna Lobley Richard Mitter Harshil Patel Adam Rabinowitz Max Salm Figure 1 Circos plots depicting inter- and intrachromosomal translocations, as well as deletions and insertions for tumour regions R1 and R3 for samples L002 and L008; shared events are indicated in blue, events private to region R1 are indicated in red, and private to region R3 in green. The outer circle represents the integer copy number data for R1 and the inner circle for R3 for each tumor sample; copy number segments with an integer value greater than mean ploidy are in red and those less than mean ploidy in blue. 148 The Bioinformatics and Biostatistics Facility (BABS) collaborates with groups that require the analysis of large or complex biological datasets. The majority of this data is derived from genomic experiments that use sequence data as their readout and the application list is continually growing, e.g. transcript isoform profiling (TIF-Seq). After processing, the data is put in a biological context by combining statistical and mathematical methods for identifying significance with annotation and other biological datasets to provide informed insight. VarSLR – algorithm for assessing mutation calling in clinical samples Accurate mutation calling in clinical tumour samples remains a formidable challenge, confounded by sample complexity, experimental artefacts and algorithmic constraints. In particular, high sequencing error rates (~0.1-1x10-2 per base) entail costly manual review of putative mutations followed by orthogonal validation. Efficient filtering is currently required, given that most mutation callers identify many thousands (in exome sequencing), if not millions (in whole genome sequencing), of candidate mutations per experiment. To aid in this process, we developed the open-source VarSLR R package to identify somatic nucleotide and insertion-deletion mutations that are likely to be sequencing artefacts. The algorithm incorporates putative confounders of call accuracy (Koboldt et al., 2012; Genome Res. 22(3):568-576) into stepwise logistic regression models and subsequently classifies variants within a simple, 4-tiered quality schema. VarSLR is highly scalable and designed to be run in an ‘embarrassingly parallel’ fashion, thus benefiting from the LRI’s high-performance computing facility. Moreover, VarSLR performed with high precision when tested with synthetic and experimental data, and has been successfully applied to numerous projects (e.g. de Bruin EC et al., 2014; Science. 346(6206):251-256). Scientific report 2014 LONDON RESEARCH INSTITUTE Identifying genes involved in drug resistance using a genome transposon screen Given the correct sequence architecture it is possible to insert promoter containing DNA elements using transposons into the human genome. If inserted in the correct orientation and close to a gene’s transcription start site the inserted promoter can activate transcription. This provides a mechanism for a genome wide positive screen using elements inserted into randomly selected transposon sites within a population of cells. The transcriptional effect of a given insert on cell survival within the context of different environmental conditions such as drug treatment can then be assayed. To determine the specific genes involved, we first need to map the insertion sites within the population of cells before and after treatment. The inserts are sequenced within their genomic context and with software developed by BABS we remove adapter from within the sequenced reads, identify and map the genomic component of the read, orientate the insert, quantify the insertion event via the number of reads, cluster insertion events that lie in close proximity to one another and finally associate the insertion events to genes. The aim is to identify genes involved in drug resistance but it is also hoped that this powerful screening technique will be made more widely available to Crick Institute scientists once established. This work has been carried out in collaboration with Su Kit Chew and Charles Swanton. Publications listed on page 177 Biomolecular Modelling www.london-research-institute.org.uk/research/paul-bates Group Leader Paul Bates Staff Raphael Chaleil Postdoctoral Scientists Tammy Cheng Mieczyslaw Torchala Graduate Students Rudi Agius Sakshi Gulati Erik Pfeiffenberger We study fundamental and challenging problems in both structural and systems biology; in particular, how macromolecules interact at the atomic level to facilitate cellular events. Much of the work involves the design of novel computer algorithms that are based upon the principles of physics and evolutionary biology. These simulations are proving to be important in helping to interpret experimental data and suggest further experiments to probe complex molecular systems. Outlined below are two systems currently under investigation. Validating biomarkers for clear cell renal cell carcinoma (ccRCC) Candidate biomarkers have been identified for ccRCC patients, but most have not been validated. In collaboration with Charles Swanton (Translational Cancer Therapeutics) we have analysed 28 genetic or transcriptional biomarkers in 350 ccRCC patients in terms of cancer-specific survival (CSS). Our conclusion from the study is that only one biomarker, a gene expression set called ccB, could be considered to be an independent prognostic biomarker for CSS in ccRCC (Gulati et al., 2014; Eur Urol. 66: 936-948). Mapping the shape of protein-protein binding funnels with SwarmDock Predicting the effects of mutations on the kinetic rate constants of protein-protein interactions is Figure 1 Schematic diagram of the binding funnel between two proteins, actin and one of its binding partners, a vitamin D-binding protein. The left hand panel shows the complete search space between the two proteins as a connected graph of conformational states. Larger nodes represent more stable protein conformations. The highly connected set of nodes represents the true binding funnel. The right-hand panel shows an exploded view of the docking funnel, actin in magenta, and in green the final conformation of the vitamin D-binding protein. Moving from A (edge of the binding funnel) to D (near the final bound state) stabilising interactions between the two proteins can be seen to increase. important to both the modelling of complex diseases, such as cancer, and the design of effective protein inhibitors. To facilitate our understanding of how mutations affect binding kinetics we are mapping the conformational shapes of binding funnels for wild type and mutated binding partners. We are developing software to display the output of our publicly available macromolecular docking program SwarmDock (bmm.cancerresearchuk.org/~SwarmDock) to interpret these binding funnel shapes (Figure 1). Details of how to effectively use our docking program are given in a recent publication (Torchala and Bates, 2014; Methods Mol Biol. 1137:181-197). Publications listed on page 179 Transition Probility 0 1 Equilibrium Population 0 B A 15 D B A C D C TECHNOLOGY CORE FACILITIES 149 CELL SERVICES www.london-research-institute.org.uk/technologies/cell-services Our dedicated team provides LRI research groups with all their cell culture needs. We work together to fulfil cell culture requirements, purified antibody, cell authentication, customised media/plates and mycoplasma screening requests as well as providing washroom services at Clare Hall. We continue to offer the London Research Institute scientific community a high level of practical and advisory support. Head Ruth Peat Staff Susan Capon Trevor Cooper Warren Cooper Darren Haines Darren Harvey Marley Holding-Pillai Rachel Horton-Harpin Spencer Horton-Harpin Samantha Kenton Julie Morrin Christine Saunders Martin Saunders Robert Saunders Debbie Schofield Sonal Sheth Karen Stoughton Mark Thorlby Paul Willis Preparations for the transfer to the Crick Institute As we are preparing our own cell stocks for transition to the new Institute, we have undertaken to grow, mycoplasma screen and validate cell lines for groups within both the LRI and NIMR, this follows the cell survey carried out in 2013. The project will enable our scientists to have access to pre-prepared, clean, cell stocks and minimise ‘down-time’ during migration to the Crick. Currently we have processed over 500 cell lines. Mycoplasma screening developments We have recently changed our PCR-based mycoplasma screening kit for one supplied by the ATCC. This offers us the same sensitivity and reliability as before, at a much-reduced cost. We use this particular test if there is a disparity between our two regular screening processes (Agar culture and Fluorescence) and also to provide a quicker result, usually within 24 hours, for people waiting to use cells in the Containment Level 2 facility. Containment Level 2. Currently Cell Services are unable to grow these lines in our facility. Using our new ATCC Mycoplasma Screening kit, we are able to test the genomic DNA from Containment Level 2 lines, to ascertain the mycoplasma status – all we need is approximately 1μg of DNA to run on our test. FTA cards for STR Profiling We continually look for ways to improve all of our existing techniques. Currently we are investigating the use of FTA cards as an alternative to genomic DNA preparation for use in short tandem repeat (STR) Profiling. Our initial tests are very positive and if this method is fully adopted it will allow us to store DNA at room temperature. In addition, it makes the process of collecting DNA from Human cell lines much simpler – adjust the cell volume and drop a certain number of cells onto the card. We hope that this innovation will make it easier for our scientists to give us DNA prior to, during and after their experiments, enabling us to provide them with the STR profile data, so often required before journal publication. 150 Scientific report 2014 LONDON RESEARCH INSTITUTE Mycoplasma - ve control Mycoplasma + ve control Mycoplasma + ve Mycoplasma + ve Mycoplasma + ve Mycoplasma + ve Mycoplasma + ve Mycoplasma - ve Figure 1 ATCC Mycoplasma gel result showing both positive and negative results, along with controls. A positive result reveals a band between 434-468 bp. 100 bp Ladder For some time we have wanted to offer mycoplasma screening for lines that are ELECTRON MICROSCOPY www.london-research-institute.org.uk/technologies/electron-microscopy Head Lucy Collinson Staff Elisabeth Brama Raffaella Carzaniga Marie-Charlotte Domart Martin Jones Christopher Peddie Matthew Russell Anne Weston The Electron Microscopy Unit (EMU) is a Core Technology Facility providing the equipment and expertise necessary to image the structure of molecules, cells and tissues at high resolution. The main core of the EMU team consists of six experienced post-doctoral electron microscopists working closely with research groups at the LRI to plan, optimise and implement high-resolution imaging experiments. In addition, two post-doctoral scientists with image analysis and laser physics expertise joined the team this year, with a remit to develop and build new integrated light and electron microscopes for biomedical applications. The EMU has expertise in preparing, imaging and interpreting a wide range of samples, including: • Proteins, DNA, and protein:DNA complexes • Yeast • Viruses and virus-infected cells • Cultured cell lines • Tissues • Model organisms (fruit flies, worms and zebrafish) Figure 1 GFP-C1 fluorescence and HeLa cell ultrastructure, imaged from a single 200nm In-Resin Fluorescence (IRF) section in an integrated light and scanning electron microscope. We use a wide range of sample preparation and imaging techniques, including: • Negative staining and low angle rotary shadowing • Resin embedding and ultramicrotomy • Cryosectioning and immunolabelling • Plunge freezing • High pressure freezing and freeze substitution • Correlative light and electron microscopy (CLEM) • Transmission EM and Scanning EM • Electron tomography • Volume electron microscopy (Serial Block Face SEM, Focused Ion Beam SEM) • Integrated light and electron microscopy (ILEM) • Cryo-fluorescence microscopy • Cryo-soft X-ray tomography (at Diamond, ALBA and BESSY II synchrotrons) Figure 2 TEM micrograph of a HeLa cell prepared in one day: from living to imaging. The facility currently houses four electron microscopes, including: • FEI Tecnai Twin 120kV TEM 1 2 • FEI Tecnai BioTwin 120kV TEM with iCorrTM integrated light microscope • Zeiss Sigma SEM with 3View for automated 3D EM • FEI Quanta SEM with a DELMIC SECOM integrated light microscope New in 2014 This year we have continued to push the limits of electron microscopy… • We are now able to collect thousands of images from cells and tissues automatically using our 3View Serial Block Face SEM. We have adapted this technique for 3D correlative imaging of cells expressing fluorescent protein constructs. • We have developed and published a method for maintaining GFP fluorescence in cells and tissues prepared for EM (Figure 1). This means that we can perform in situ correlative imaging in an integrated light and electron microscope. We are the only site worldwide to have three integrated light and electron imaging systems (TEM, SEM and benchtop SEM). We are now developing integrated super-resolution light and electron microscopy for nanoscale accuracy in protein localisation. • We are one of ten sites worldwide to beta-test a new automated ultramicrotome called the ATUMtome, developed in Jeff Lichtman’s lab at Harvard, which can cut thousands of resin sections onto tape for array tomography in an SEM. • We are starting to apply new protocols that allow us to prepare cells and tissues extremely quickly, going from a live sample to TEM imaging in one day (as opposed to one week or longer) (Figure 2). Publications listed on page 179 TECHNOLOGY CORE FACILITIES 151 EQUIPMENT PARK www.london-research-institute.org.uk/technologies/equipment-park The Equipment Park provides access to state of the art molecular biology instrumentation and offers instruction in the correct and efficient use of the technologies involved. The range of equipment is constantly reviewed and specific requests from laboratory heads are encouraged. Head Graham MG Clark Staff Vicky Dearing Olga O’Neil David Philips Ramin Sadri The technologies include: Real-Time PCR quantitation – we have seven systems (4x96 well and 3x384 well format) designed to detect fluorescence during the thermal cycling of PCR. By plotting the increase in fluorescence versus the cycle number, the system produces amplification plots that provide a more complete picture of the PCR. PHERASTARPlus micro-plate reader – multidetection HTS micro-plate reader with Simultaneous Dual Emission in all modes. The reader is able to perform all leading non-isotopic detection technologies including: • Fluorescence Intensity • Fluorescence Polarisation • Time-Resolved Fluorescence • TR-FRET • Luminescence • Absorbance UV/Vis The QIAgility is a bench-top instrument for automated setup of PCR reactions that is able to handle a wide variety of tube and plate formats. Figure 1 LC3 lipidation assay to analyse autophagic flux. Lipidated LC3 (LC3 II) associates with autophagosomes and migrates faster in SDS-PAGE. Starvation (SM) induces protein degradation with autophagy and leads to an increase in LC3 II. Inhibition of lysosomal Bafilomycin A1 increases LC3 II under basal (FMB) and starvation (SMB) conditions. A stable inducible GFP-SCOC HeLa cell line was used for this experiment. Image courtesy of Dr Martina Wirth, Secretory Pathways Laboratory. 152 Scientific report 2014 LONDON RESEARCH INSTITUTE The system performs the preparation of master mix from individual reaction components and dilutions of standard series. Optionally available UV light and HEPA filter help to reduce the risk of sample carry-over. In addition to PCR setup, the flexibility of the system allows high-precision pipetting applications including: • Normalisation of DNA and RNA concentration • Transfer of liquid samples from one tube format to another • Serial dilutions with variable dilution ratios • Sample pooling Biomek FX Robot – advanced liquid-handling applications include: • High-density replication • Assay plate set-up (PCR, quantitative PCR (384 well), ELISA, kinase etc.) • PCR and sequencing reaction clean-up using paramagnetic technology. ImageQuant LAS 4000 – a digital (CCD) imaging system for sensitive, quantitative imaging of gels and blots without film, by chemiluminescence and fluorescence. The high sensitivity and wide dynamic range is designed to capture the signals from ECL western blotting reagents. A wide range of visible fluorescent dyes can also be imaged via red, green and blue epi-illumination. Li-COR Bioscience Odyssey Infrared Imager – the imaging system offers a different way to analyse blots and gels. Odyssey is uniquely equipped with two infrared channels for direct fluorescence detection enabling simultaneous probing of two separate targets on the same gel, e.g. Western Blots (Figure 1). EXPERIMENTAL HISTOPATHOLOGY www.london-research-institute.org.uk/technologies/experimental-histopathology The Experimental Histopathology (EHP) Facility provides advice, training and expertise in a range of techniques to analyse cells and tissues from experimental models and human tissue banks. Head Gordon Stamp Staff Janni Bertelsen Tamara Bunting Kornelia Fritsch Emma Nye Bradley Spencer-Dene Richard Stone Figure 1 Positive specific staining (brown) for glucagon mRNA, a secreted hormone found only in α-cells in the periphery of adult mouse pancreatic islets. Figure 2 Positive staining for ZFP36/ tristetraprolin in human lung tumour and adjacent stroma. 1 Technical procedures performed include: • Human & rodent histopathology: • MicroPix image capture teaching/discussion platform • Optimal handling/fixation of fresh tissue • Adult & embryonic mouse dissection • Mouse developmental analysis, including embryonic lethal phenotypes • Histological sectioning/staining of frozen/fixed tissue • High resolution photomicroscopy using NIS Elements platform with semi-automated morphometric analysis for object classification/ measurement • Immunohistochemistry (IHC) against >350 mouse orientated antibodies plus novel antibody optimisation (automated stainer for high throughput) • Non isotopic in situ hybridisation (ISH) on slides and whole-mount embryos and organs • 3D volume rendered reconstruction • Laser Capture Microdissection • Ariol SL50 Scanning system with morphometric software modules; – IHC: for quantitating membrane, nuclear and cytoplasmic expression – FISH and immunofluorescent capture – Tissue Microarray – General Morphometric Image Analysis module (ploidy, angiogenesis, area/volume etc.) EHP encourages participation by graduate students and postgraduate scientists, and we provide individual training and expert advice of laboratory techniques. 2 Technology Highlight RNAscope, now available in the EHP Facility, is an innovative non-radioisotopic in situ hybridisation assay for visualising the expression of any gene within any tissue to extremely low abundance mRNA. Results generated have provided highly specific detection of mRNA in cells in routinely fixed tissues. This provides a solution to demonstrating gene expression in tissues where there is no suitable antibody for IHC. RNAscope uses a methodology similar to FRET whereby two independent (double Z) probes must hybridise to the target sequence in tandem so that signal amplification occurs. Key features of the assay include: • Single RNA detection limit – most sensitive in situ assay. Unique ‘ZZ probe’ design for very high specificity • Manual workflow: 8 hours from slide to image analysis. 3 weeks from target sequence to probe for any gene, in any tissue, in any species • Integrated QC assay to confirm presence of detectable mRNA and degree of fixation • Duplex chromogenic and multiplex fluorescence readouts are available • Fully automated option available on the EHP lab’s Ventana Discovery ULTRA To date we have successfully localised mRNA in human cancer TMAs, formalin-fixed paraffin embedded tissues, and cell pellets, with various applications such as: • Demonstration of LOXL2 gene expression heterogeneity in human renal cancer • Determining specific location of low abundance biomarkers PDL-1 and ZFP36 in human lung cancers • Localising secreted proteins/transcription factors, e.g. Ngn3 and differentiation markers, e.g. glucagon, in mouse pancreas as well as cytokines in mouse models of lung cancer Publications listed on page 180 TECHNOLOGY CORE FACILITIES 153 FLUORESCENCE ACTIVATED CELL SORTING www.london-research-institute.org.uk/technologies/facs Head Derek Davies The FACS Facility at the London Research Institute is a dedicated scientific service offering an extensive flow cytometry analysis and sorting facility. Flow cytometry is a sophisticated form of fluorescence microscopy where cells in suspension pass one by one through a laser beam and emitted fluorescence can be captured and measured. Any part of a cell or any function of a cell that can be tagged by a fluorochrome may be measured by flow cytometry, which makes it an essential technique in many biological applications. Staff Laura Bazley Julfa Begum Joana Cerveira Andy Filby Carl Henderson Sukhveer Purewal Kirsty Sharrock Equipment Analytical cytometers The FACS Facility has 9 analytical cytometers including one plate-reading cytometer. These may all be user-operated and we offer a one to one training for all new users of the facility. Masters Student Thomas Scott • FACS Calibur: 4 fluorescence detectors, 2 lasers (488nm and 633nm) • LSRII: 13 fluorescence detectors, 4 lasers Cell sorters These are able to retrieve up to six specifically defined populations so that cells may be recovered for re-culture, functional assays or RNA or DNA recovery. Only members of the FACS Laboratory operate the sorters but experiments are scheduled and planned in close collaboration with our users. Three of the sorters are housed in Class 2 Microbiological Safety Cabinets. (355nm, 405nm, 488nm, 633nm) • LSRII-SORP: 16 fluorescence detectors, 4 lasers (405nm, 488nm, 561nm, 648nm) • LSR Fortessa: 15 fluorescence detectors, 5 lasers (355nm, 405nm, 488nm, 561nm, 638nm) • Two LSR Fortessas: 18 fluorescence detectors, 5 lasers (355nm, 405nm, 488nm, 561nm, 638nm) • MACSQuant VYB: 8 fluorescence detectors, 3 lasers (405nm, 488nm, 561nm) • FlowSight Imaging flow cytometer, 10 colour detection, 4 lasers (405nm, 488nm, 561nm, 640nm) • Image Stream X Imaging flow cytometer: 12 channel detection with 4 lasers (405nm, 488nm, 561nm, 640nm) Figure 1 The Amnis ImageStreamX Imaging Flow cytometer, which allows quantitation of fluorescence in conjunction with cell imagery. • MoFlo XDP: 9 fluorescence detectors, 4 lasers (355nm, 488nm, 561nm or 648nm) • FACS Aria III: 13 fluorescence detectors, 4 lasers (405nm, 488nm, 561nm, 640nm) • Influx: 14 fluorescence detectors, 4 lasers (405nm, 488nm, 561nm, 640nm) • FACS Aria Fusion: 13 fluorescence detectors, 4 lasers (405nm, 488nm, 561nm, 640nm) Other services The members of the FACS Facility are available to provide advice on the design of experiments, sourcing and supply of reagents, data analysis, presentation and interpretation as well as troubleshooting machines and experiments. We also develop and introduce new techniques and technologies that would be useful to our users. We collaborate closely with the facility’s users and this has led to several recent publications particularly with groups involved in stem cell investigation, imaging flow cytometry or where DNA analysis and cell kinetic information is required. Publications listed on page 181 154 Scientific report 2014 LONDON RESEARCH INSTITUTE HIGH THROUGHPUT SCREENING www.london-research-institute.org.uk/technologies/high-throughput-screening The High Throughput Screening Facility enables research groups to access large-scale screening technologies. Primarily this takes the form of genome-wide siRNA screens although other types of screening are increasingly popular particularly screening with our bespoke collection of well-characterised small molecule modulators. Head Michael Howell Staff Rachael Instrell Ming Jiang Rossella Rispoli Becky Saunders Summer Student Silvia Benito Last year we reported upgrades to our machinery aimed at increasing our capacity and throughput. Until now large-scale screening experiments have been conducted using 96- or 384-well plate formats. Even when using the 384-well format, a genome-wide siRNA screen requires 200 such plates and remains expensive. Although our improved machinery makes it possible to utilise the newer 1536-well format, there is still a question as to whether some biological responses could be compromised by this very small scale (Figure 1A). This year we tested this format in a real world screening setting. We have previously conducted a genome-wide siRNA screen in 384-well format for the Developmental Signalling Group aimed at identifying regulators of the TGF-β signalling pathway where the activity of the pathway can be monitored by the level of Smad2 accumulation in the nucleus (Figure 1B). We repeated this screen in the 1536-well plate format to see whether we could identify any siRNA reagents that inhibited the nuclear accumulation of Smad2 in response to A 1536well C More importantly, there was a 50% overlap between siRNA reagents significantly affecting Smad2 accumulation in the two formats (Figure 1D). This degree of overlap is similar to what we, and others, have observed when identical screens in the same format, but conducted on separate occasions, are compared. We conclude therefore that this new format does not inherently compromise the biology under observation. Moreover, this format offers a viable approach to screening cells with limited availability e.g. stem cells, or conducting screens across many related lines at reduced cost thereby opening up whole new research possibilities. Publications listed on page 181 B 384well 96well 2 R ~0.7 Raw cell no. replicate 2 Figure 1 A. The relative sizes of an individual well from a 96-, 384and 1536-well plate. B. Nuclear accumulation of Smad2 in HaCat cells in response to TGF-β monitored by immunofluorescence. From these images, parameters such as cell number per well (a measure of viability) and Smad2 nuclear intensity (a measure of Smad2 accumulation) are calculated. C. Scatter plot comparing raw ‘cell number per well’ measures for each well across two replicates from the 1536 genome screen. The correlation coefficient of 0.7 indicates a satisfactory degree of reproducibility. D. Venn-diagram indicating the overlap between siRNA reagents identified as significantly inhibiting TGF-β signalling (more than 3 S.D.) in either the 1536- or 384-well format screens. TGF-β in this format and if so whether hits from such a screen bore any similarity to those previously identified. When analysed in isolation, the 1536-well screen data was internally consistent and reproducible with replicates showing good correlation with each other (Figure 1C). Untreated D 1536-well screen 209 Raw cell no. replicate 1 TGF-β treated 384-well screen 208 206 Number of siRNA reagents inhibing TGF-β signalling TECHNOLOGY CORE FACILITIES 155 LIGHT MICROSCOPY www.london-research-institute.org.uk/technologies/light-microscopy We provide services in multi-dimensional imaging with fixed and live specimens using confocal microscopy and low-light-level imaging. Support is also available for image processing and motion analysis. In addition, we pursue collaborative research in application and development of light microscopy techniques. Head Daniel Zicha Staff Dominic Alibhai Deborah Aubyn Trevor Duhig Peter Jordan Alastair Nicol Matthew Renshaw Imaging technology overview Laser scanning confocal microscopes • LSM 880 (Zeiss) • LSM 780s (Zeiss) including a multiphoton system with a tuneable Chameleon Ultra II laser (Coherent) • LSM 710s (Zeiss) • SP5 (Leica) • LSM 510s (Zeiss) • Swept Field Confocal microscope Opterra (Bruker) • UltraVIEW spinning-disk confocal imaging system (PerkinElmer) • low-light-level imaging systems based on Metamorph software (Molecular Devices) including a confocal high content screening system Discovery 1 (Molecular Devices) and a microinjection system (Eppendorf) have been configured for contrast enhancement, high resolution 3D and dynamic imaging of biological specimens in multiple fluorescence channels with optical sectioning using motorised focus at multiple fields. Detailed information on functionality of individual imaging systems is presented on the Intranet. Image processing and statistical analysis can be employed for deconvolution, co-localisation, automatic or interactive segmentation of cells and intracellular structures, morphometry and tracking using Figure 1 Images of Biotium CF TM 488A labelled microtubules in type II pneumocytes fixed with 4% PFA produced with LSM 510 using a plan apochromat 63x/1.4 objective lens. Time-lapse sequence of Z-stacks was acquired while temperature in the microscope’s environmental incubator was increasing. XY and XZ projections from raw stacks showed spatial drift, which was corrected using the Huygens Object Stabilizer. Red arrows show positions of identical structures before and after correction. Plots of detected drift were automatically produced during the processing. Stack size is 20 x 21 x 6 μm. 156 Scientific report 2014 LONDON RESEARCH INSTITUTE Huygens (SVI), Volocity (Improvision), Imaris (Bitplane), AQM/ iQ (Kinetic Imaging/ Andor), Metamorph (Universal Imaging), Fiji-ImageJ, Mathematica (Wolfram Research), MATLAB (MathWorks) and C. New confocal imaging system This year we introduced a state of the art inverted laser scanning confocal microscope LSM 880 from Zeiss. We are the first laboratory in the UK with this technology. The LSM 880 has improved sensitivity and speed due to a water cooling system and upgraded electronics. Its updated software ZEN 2 features new flexible Experiment Designer. New stabiliser software This year we also introduced a new Huygens Object Stabilizer from Scientific Volume Imaging (Figure 1). A range of algorithms suitable for different situations is available. Collaborative research highlights We have developed bespoke software for quantitative analysis of Fluorescence Loss In Photobleaching (FLIP) for the specific requirements in a collaborative project (Brownlow et al., 2014; Nat Commun. 5:5658). Publications listed on page 182 PEPTIDE CHEMISTRY www.london-research-institute.org.uk/technologies/peptide-chemistry Head Nicola O’Reilly Staff Ganka Bineva-Todd Stefania Federico Dhira Joshi The Peptide Chemistry Facility provides peptides and peptide arrays to LRI scientists. We make peptides of many lengths and modifications including biotin addition, dye labels, phosphorylation, methylation, acetylation, isoprenylation, branched peptides and peptides linked by disulphide bridges. We are keen to make unusual peptides or peptide-based reagents, which can enable scientists to further their research. We can aid in design of peptides and conjugation and immunisation strategies of peptides for antibody generation. We have around 20 peptides in stock that are used to elute proteins from columns, prime immune cells and synchronise yeast. This year we have synthesised the yeast mating factor M-factor. Synthesis of M-factor Conjugation in Schizosaccharomyces pombe is controlled by the reciprocal action of mating pheromones, p-factor and M-factor. M-factor, released by cells of mating type Minus, is a nonapeptide in which the C-terminal cysteine residue is carboxyl-methylated and S-farnesylated. M-factor was synthesised following the method of synthesis for a-factor (O’Reilly et al., 2012; Yeast. 29:233-40). Method: The synthesis of M-factor was performed on a 433A synthesiser using Fmoc solid phase peptide chemistry on a 2-Chlorotrityl resin. The N-terminal amino acid was protected with a Boc Figure 1 Schematic representation of the synthesis of M-factor. Figure 2 FACS data on the M-factor of Schizosaccharomyces pombe cells shows that even at the lower concentration tested (0.1mg/ml) the pheromone induced G1 arrest (1c). In collaboration with Francisco Navarro (Cell Cycle Group). 1 group. The peptide was cleaved from the resin using 1% TFA in DCM whilst all side chains and the N-terminal were still protected. Next, the C-terminal end of the peptide was methylated using trimethylsilyldiazomethane. Following this, side chain and N-terminal protecting groups were removed using 95% TFA - 2.5% H2O - 2.5% TIS. Finally the peptide was farnesylated using farnesyl bromide then purified by RP-HPLC. A schematic representation of the synthesis is shown (Figure 1). Publications listed on page 182 2 Boc-Tyr(OtBu)-Thr(OtBu)-Pro-Lys(tBoc)-Val-ProTyr(OtBu)-Met-Cys(Trt)-Cltrt Resin 1% TFA in DCM Boc-Tyr(OtBu)-Thr(OtBu)-Pro-Lys(tBoc)-Val-ProTyr(OtBu)-Met-Cys(Trt)-OH Trimethylsilyldiazomethane Boc-Tyr(OtBu)-Thr(OtBu)-Pro-Lys(tBoc)-Val-ProTyr(OtBu)-Met-Cys(Trt)-OCH3 95% TFA - 2.5% H2O - 2.5% TIS NH2-Tyr-Thr-Pro-Lys-Val-Pro-Tyr-Met-Cys-OCH3 Farnesyl Bromide NH2-Tyr-Thr-Pro-Lys-Val-Pro-Tyr-Met-Cys(SFarnesyl)-OCH3 TECHNOLOGY CORE FACILITIES 157 PROTEIN ANALYSIS AND PROTEOMICS www.london-research-institute.org.uk/technologies/protein-analysis-and-proteomics Head Bram Snijders Staff Karin Barnouin Vesela Encheva-Yokoya Helen Flynn David Frith Andrew Jones The Protein Analysis and Proteomics (PAP) Facility at Clare Hall provides a wide variety of workflows for the analysis of proteins, peptides and their post-translational modifications. The laboratory houses two LTQ-Orbitrap instruments, a Q-Exactive and a 5800 Maldi TOF/TOF instrument. We specialise in the following key areas: 1) Interaction proteomics, 2) Post-translational modifications, 3) Global and subcellular proteomics, 4) Targeted approaches, 5) Computational proteomics. In order to extract the maximum amount of information from your sample we provide expertise in all key workflow aspects including analytical design, wet lab procedures, computational and statistical procedures. Post-translational modifications We have established workflows for the investigation of post-translational modifications in complex cell lysates or on purified proteins. Typically, these workflows require PTM enrichment steps using TiO2 or PTM specific antibodies. Depending on the desired result we are also able to employ fractionation strategies such as strong-cation-exchange (SCX) chromatography or off-line reverse phase chromatography. The resulting GBs of data can contain hundreds of thousands of spectra that are then subjected to high performance computation and rigorous statistical analysis for identification, quantification, PTM localisation and FDR control. Targeted approaches Once peptide properties such as transitions and retention times are known they can be used for confirmation or follow-up experiments through targeted SRM and PRM approaches. In 2014 PAP increased its capabilities in this area through an increased usage of Skyline software and the implementation of Panorama server for storage of spectral and chromatogram libraries. Further, Figure 1 Log2 Ratio vs intensity plot of the phosphoproteome of activated raw cells. Erk phosphorylation sites indicated in pink. Total number of quantified sites is 14922. Plots generated using Persues software. 1 Figure 2 Quantification of CDC7 peptides with attomole sensitivity as measured by a SIM/PRM scan. 158 Scientific report 2014 LONDON RESEARCH INSTITUTE targeted and multiplexed scans on the Q-Exactive instrument demonstrated sensitivities and specificities typical for western blot analysis without the requirement for affinity reagents. Interaction proteomics Co-immunoprecipitation of proteins followed by their identification by mass spectrometry is a powerful strategy for the discovery of novel protein complexes. Despite this, it can be challenging to recognise bona fide interactors and absence/ presence of protein identification criterion alone. Depending on the bait and the outcome of preliminary experiments we apply quantitative approaches based on chromatogram peak areas (iBAQ, Silac) or spectral counting. Further, we maintain databases of common contaminants that provide an additional quality filter. 2014 also saw an increased integration with NIMR and the development strategic model in preparation for relocation of the facility to the Crick Institute. Publications listed on page 182 2 PROTEIN PURIFICATION www.london-research-institute.org.uk/technologies/protein-purification The Protein Purification Facility (PPF) supports and collaborates with LRI-scientists in numerous aspects of research involving expression and characterisation of recombinant proteins. These activities span from early-stage project discussion relating to expression and purification strategies through to hands-on pilot and productionscale involvement and, finally, generation of biophysical data. Head Svend Kjær Staff Annabel Borg Roger Rajesh George Sara Kisakye-Nambozo Figure 1 Crystal structure of anti-RET Fab fragment. Collaboration with Neil McDonald and Andrew Purkiss. PPF specialises in high-end expression technologies such as baculovirus (BV)-mediated protein expression in silkworm-derived insect cells as well as transient transfection of suspension culture HEK293 cells. Additionally, an S2 cell-line (Drosophila melanogaster-derived) expression system has been added recently. The S2 expression system is particularly suited for secreted proteins. For large-scale expression, two Minifors bioreactors can be deployed. The purification process has been advanced through implementation of two AKTA Pure state of the art chromatography workstations, which accelerate workflows through automated and sequentially linked multi-column setups. To identify buffer conditions or small molecule additives, which stabilise the purified proteins, PPF now routinely applies the Thermofluor technology. Subsequently, the PPF can either educate users in, and/or perform various protein characterisation techniques such as Multi-Angle Light Scattering (MALS) for molecular weight determination, Isothermal Calorimetry (ITC), Biolayer Interferometry (BLI) and most recently Microscale Thermophoresis (MST) as various means of determining affinities of macromolecular interactions. The year 2014 at the PPF In line with previous years, 2014, the final year as PPF in its current incarnation, was very busy with the demand for PPF main deliverable – BV-related work – reaching 120-130 virus, including several MultiBac projects. Additionally, the trimming of certain upstream workflows has liberated resources for purification and protein characterisation and hence, the yearly output of well-characterised proteins for downstream applications such as crystallography, activity measurements and small molecule screens was unprecedented. Finally, the ‘Mab-to-Fab’ – pipeline was finalised with crystallisation and structural determination of an anti-RET Fab fragment (expressed in S2 cells) in collaboration with Neil McDonald (Figure 1). PPF at the Francis Crick Institute As of 1st of April 2015, the LRI becomes part of the Francis Crick Institute and the PPF will join the Structural Biology Science Technology Platform. The platform will provide support to the structural biology community and engage with nonstructural biologists in a variety of aspects of biochemical and biophysical characterisation of proteins. Publications listed on page 183 TECHNOLOGY CORE FACILITIES 159 TRANSGENICS www.london-research-institute.org.uk/technologies/transgenics Transgenic Services are a Core Facility of seven people providing the skills, services and techniques relevant to supporting mouse genetics at the institute. This can involve genetic modification techniques or involve cryopreservation and rederivation to optimise the use of available space and resource for this work. Head Ian Rosewell Staff Jessica Gruninger Mary Ann Haskings Natalia Karzakova Natalia Moncaut Jaroslaw Narloch Sunita Varsani-Brown Genetic modification has traditionally involved two routes. One, the injection of the pronucleus of a one cell stage embryo with DNA solution, providing for random integration of sequence. Then, targeted modifications have been possible via the embryonic stem (ES) cell route that adds the possibility of screening multiple clones to isolate relatively rare homologous recombinants. In the last two years genetic modification via targeted delivery of nucleases has added a further option. Nucleases can create double strand breaks (DSB) in a pre-determined DNA sequence that could for example remove gene expression. If the DSB is repaired from a co-injected repair template, scope for all forms of complex genetic modifications become possible. This is an exciting development that is already in use across many research laboratories within the LRI. In particular the use of CRISPR/Cas9 to deliver nucleases to the genome suggests significant advantages over current technologies and we have been keen to advance its use with mice at the LRI. Figure 1 Cytoplasmic injection of a one cell stage fertilised mouse embryo to achieve CRISPR/Cas9 gene editing. Last year in collaboration with the Translational Cancer Therapeutics laboratory a gene targeting project in ES cells was the first to use CRISPR/Cas9 for generating a gene-targeted mouse. This year has seen the first mice created by direct injection of CRISPR/Cas9 RNA into the cytoplasm of a one cell fertilised mouse embryo creating a ‘knockout’ mouse. Several further projects, for example to create conditional knockout mice, are underway. Another strand of development was prompted by a seminar given in 2012 at Lincolns Inn Fields. Dr Jos Jonkers from the Netherlands Cancer Institute presented work that involved the generation of ‘chimera cohorts’. This work is highly relevant to the use of mouse cancer model research at the LRI. Where using such animal models would normally have involved breeding multiple alleles into a background strain requiring extensive, time consuming breeding programmes. The alternative is to use ES cells derived from strains bearing complex allele combinations, to generate experimental cohorts of chimeric mice from blastocyst injection. This approach builds on our expertise in generating and further modifying ES cells, enabling experimental cohorts to be produced with the potential for repeated cohorts. An archive and discussion of such cell lines is available at www.infrafrontier.eu. Publications listed on page 183 160 Scientific report 2014 LONDON RESEARCH INSTITUTE FERMENTATION IN VIVO IMAGING www.london-research-institute.org.uk/ technologies/in-vivo-imaging Fermentation Alireza Alidoust Namita Patel The Fermentation Service is based at Clare Hall Laboratories and provides pilot-plant scale production of microbiological organisms for LRI scientists. Batches of 10-100 L of yeast, bacteria or nematodes can be produced under stringently controlled conditions using four state of the art fermenters. It is possible to run batch or fed-batch cultures where additional nutrient or induction chemicals can be added at any point during the run. Computer control of the main growth parameters (pH, temperature, dissolved oxygen, aeration and agitation) allows a wide spectrum of growth conditions and reproducibility. The types of organisms we grow routinely are yeasts (Saccharomyces cerevisiae, Schizosaccharomyces pombe and Pichia pastoris), the bacteria Escherichia coli and the nematode Caenorhabditis elegans for whole cell production or protein purification. The service also offers breakage of cells using one of three available methods: • cell disrupter • ball mill • freezer mill Structural, functional, cellular and molecular imaging technologies have become pivotal to better understand the development and homeostatic regulation of normal and diseased tissues and to assess the efficacy of new therapeutic strategies in the context of longitudinal studies. This is particularly true in oncology but also applies to virtually any biomedical research field. The facility is equipped with 5 cutting edge imaging modalities providing complementary information: • Whole body bioluminescence and fluorescence (IVIS Spectrum; PerkinElmer) • Hybrid multiphoton/confocal microscope (LSM710-NLO; Zeiss) • X-ray micro-computed tomography (micro-CT; Skyscan1176; Bruker) • High resolution ultrasound (US; Vevo2100; Visualsonics) A number of exciting projects have developed this year to image different types of cancer models as well as new collaborations (Figure 1). We are delighted to announce the establishment of a partnership with the UCL Centre for Advanced Biomedical Imaging (CABI) for the development of the In Vivo Imaging capabilities of the Francis Crick Institute. The CABI is a world class imaging centre, which will contribute its expertise, notably to establish MRI and nuclear imaging at the Francis Crick Institute. In Vivo Imaging Francois Lassailly May Zaw Thin Publications listed on page 183 Fermentation Figure 1 New Brunswick fermenter In Vivo Imagining Figure 1 A. Visualisation of individual human acute myeloid leukaemia (AML) cells (green) developing around blood vessels (red) within an intact bone (cyan) thanks to intravital microscopy. Collaboration with Diana Passaro, Haematopoietic Stem Cell. B. Image-guided injection for precise delivery of cells or (bio) chemicals at a specific anatomical localisation using ultrasound imaging. Collaboration with Rute Ferreira, Mammalian Genetics. C. Micro-CT phenotyping of pups (left: whole body; right: calcified tissues (bones), where bone density has been pseudocoloured). Collaboration with Prof. Paul Gissen – GOSH/ICH-UCL. D. A pilot micro-CT study performed in collaboration with Owen Arthurs (GOSH and ICH-UCL) demonstrated the use of new technology to improve perinatal, post-mortem clinical diagnostic practice, showing detailed views of the morphology of the heart chambers and great vessels. A C B D TECHNOLOGY CORE FACILITIES 161 PROTEIN STRUCTURE www.london-research-institute.org.uk/ technologies/protein-structure X-ray crystallography can provide highly detailed molecular structure of large molecular assemblies. Elucidating the atomic details of molecular interactions is particularly important to understand the biological function of a protein/ enzyme at an atomic level. Protein Structure Stéphane Mouilleron The Protein Structure Facility provides to any biology group from the LRI the opportunity to undertake structural studies of their favourite proteins in-house at the LRI. This allows groups, collaborative access to the state of the art high throughput crystallisation robots and regular access to the ESRF (Grenoble, France) and Diamond synchrotron (Oxford, UK) to collect high quality data from protein crystals. We are currently collaborating with five LRI groups and, so far, four of those collaborations lead to the resolution of one or more crystal structures. Publications listed on page 183 SUPER-RESOLUTION MICROSCOPY www.london-research-institute.org.uk/ technologies/super-resolution-microscopy Super-resolution microscopy continued to be an important tool for research projects that study how chromosomes segregate during cell division. Errors in this process lead to too few or too many chromosomes in daughter cells, which is a hallmark of cancer progression in humans. Some of these studies are done using yeast cells. Yeast is an important model system. However, due to the small size of yeast cells, it is difficult to image molecules in these cells using conventional, diffraction-limited microscopes. Super-resolution microscopy has been used e.g. to measure the amount of native chromatin condensation in yeast cells. This is not possible using a confocal microscope. One of the most recent super-resolution microscopy projects studies centrosomal proteins such as Msd1 and Msd2. Anomalous centrosomes are often present in tumour cells so it is important to better understand the structure and function of centrosomes. High-resolution imaging has revealed new features in how the centrosome is organised at the molecular level. Super-Resolution Microscopy Anne Vaahtokari Protein Structure Figure 1 Protein crystal structures solved by the Protein Structure Facility. Super-Resolution Microscopy Figure 1 Centrosomes of asynchronously growing HeLa cells stained with the indicated combinations of antibodies and visualised using either a structured illumination super-resolution (OMX) or deconvolution (DeltaVision) microscope. Scale bar, 1 μm. Courtesy of Akiko Nishi, Cell Regulation Laboratory. 162 Scientific report 2014 LONDON RESEARCH INSTITUTE RESEARCH PUBLICATIONS RESEARCH PUBLICATIONS THESES SUBMITTED 2014 RESEARCH PUBLICATIONS 163 RESEARCH PUBLICATIONS Facundo Batista (page 66) Lymphocyte Interaction Primary Research Paper Burbage M, Keppler SJ, Gasparrini F, Martinez-Martín N, Gaya M, Feest C, Domart MC, Brakebusch C, Collinson L, Bruckbauer A, Batista FD. Cdc42 is a key regulator of B cell differentiation and is required for antiviral humoral immunity. J Exp Med. 2014;doi:10.1084/jem.20141143 Schülein-Völk C, Wolf E, Jing Zhu, Xu W, Taranets L, Hellmann A, Jänicke LA, Diefenbacher ME, Behrens A, Eilers M, Popov N. Dual regulation of Fbw7 function and oncogenic transformation by Usp28. Cell Rep. 2014;9(3):1099-109 Zhang T, Cronshaw J, Kanu N, Snijders AP, Behrens A. UBR5-mediated ubiquitination of ATMIN is required for ionizing radiation-induced ATM signaling and function. Proc Natl Acad Sci USA. 2014;111(33):12091-6 Other Publication Feest C, Bruckbauer A, Batista FD. B Cell Receptor Signaling. Cell Membrane Nanodomains: From Biochemistry to Nanoscopy by Cambi A and Lidke DS. CRC Press. 2014;ISBN 9781482209891 Other Publication Behrens A, van Deursen JM, Rudolph KL, Schumacher B. Impact of genomic damage and ageing on stem cell function. Nat Cell Biol. 2014;16(3):201-7 Axel Behrens (page 68) Dominique Bonnet (page 70) Mammalian Genetics Haematopoietic Stem Cell Primary Research Papers Diefenbacher ME, Popov N, Blake SM, Schülein-Völk C, Nye E, Spencer-Dene B, Jaenicke LA, Eilers M, Behrens A. The deubiquitinase USP28 controls intestinal homeostasis and promotes colorectal cancer. J Clin Invest. 2014;124(8):3407-18 Primary Research Papers Griessinger E, Anjos-Afonso F, Pizzitola I, Rouault-Pierre K, Vargaftig J, Taussig D, Gribben J, Lassailly F, Bonnet D. A niche-like culture system allowing the maintenance of primary human acute myeloid leukemia-initiating cells: A new tool to decipher their chemoresistance and self-renewal mechanisms. Stem Cells Transl Med. 2014;3(4):520-9 Sancho R, Gruber R, Gu G, Behrens A. Loss of Fbw7 reprograms adult pancreatic ductal cells into α, δ, and β cells. Cell Stem Cell. 2014;15(2):139-53 Pizzitola I, Anjos-Afonso F, Rouault-Pierre K, Lassailly F, Tettamanti S, Spinelli O, Biondi A, Biagi E, Bonnet D. Chimeric Antigen Receptors against CD33/ CD123 antigens efficiently target primary Acute Myeloid Leukemia cells in vivo. Leukemia. 2014;28(8):1596-605 Other Publications Anjos-Afonso F, Bonnet D. Forgotten gems: human CD34(-) hematopoietic stem cells. Cell Cycle. 2014;3(4):503-4 Tettamanti S, Biondi A, Biagi E, Bonnet D. CD123 AML targeting by chimeric antigen receptors: A novel magic bullet for AML therapeutics? Oncoimmunology. 2014;3:e28835 Zhao W, Phinney DG, Bonnet D, Dominici M, Krampera M. Mesenchymal stem cell biodistribution, migration, and homing in vivo. Stem Cells Int. 2014;2014;292109 Co-staining for CK19 (red) and insulin (green) in an Fbw7-deficient pancreatic duct. 164 Scientific report 2014 LONDON RESEARCH INSTITUTE Julian Downward (page 74) Signal Transduction Primary Research Papers de Bruin EC, Cowell C, Warne PH, Jiang M, Saunders RE, Melnick MA, Gettinger S, Walther Z, Wurtz A, Heynen GJ, Heideman DA, Gómez-Román J, García-Castaño A, Gong Y, Ladanyi M, Varmus H, Bernards R, Smit EF, Politi K, Downward J. Reduced NF1 expression confers resistance to EGFR inhibition in lung cancer. Cancer Discov. 2014;4(5)606-19 Endesfelder D, Burrell R, Kanu N, McGranahan N, Howell M, Parker PJ, Downward J, Swanton C, Kschischo M. Chromosomal instability selects gene copy number variants encoding core regulators of proliferation in ER+ breast cancer. Cancer Res. 2014;74(17):4853-63 Murillo MM, Zelenay S, Nye E, Castellano E, Lassailly F, Stamp G, Downward J. RAS interaction with PI3K p110α is required for tumor-induced angiogenesis. J Clin Invest. 2014;124(8):3601-11 Wang Y, Bu F, Royer C, Serres S, Larkin JR, Soto MS, Sibson NR, Salter V, Fritzsche F, Turnquist C, Koch S, Zak J, Zhong S, Wu G, Liang A, Olofsen PA, Moch H, Hancock DC, Downward J, Goldin RD, Zhao J, Tong X, Guo Y, Lu X. ASPP2 controls epithelial plasticity and inhibits metastasis through β-catenin-dependent regulation of ZEB1. Nat Cell Biol. 2014;16(11):1092-104 Other Publications Downward J. RAS’s cloak of invincibility slips at last? Cancer Cell. 2014;25(1):5-6 Geraghty RJ, Capes-Davis A, Davis JM, Downward J, Freshney RI, Knezevic I, Lovell-Badge R, Masters JR, Meredith J, Stacey GN, Thraves P, Vias M. Guidelines for the use of cell lines in biomedical research. Br J Cancer. 2014;111(6):1021-46 Holger Gerhardt (page 76) Vascular Biology Primary Research Papers Bentley K, Franco CA, Philippides A, Blanco R, Dierkes M, Gebala V, Stanchi F, Jones M, Aspalter IM, Cagna G, Weström S, Claesson-Welsh L, Vestweber D, Gerhardt H. The role of differential VE-cadherin dynamics in cell rearrangement during angiogenesis. Nat Cell Biol. 2014;16(4):309-21 Bernabeu MO, Jones ML, Nielsen JH, Krüger T, Nash RW, Groen D, Schmieschek S, Hetherington J, Gerhardt H, Franco CA, Coveney PV. Computer simulations reveal complex distribution of haemodynamic forces in a mouse retina model of angiogenesis. J R Soc Interface. 2014;11(99):20140543 Fan J, Ponferrada VG, Sato T, Vemaraju S, Fruttiger M, Gerhardt H, Ferrara N, Lang RA. Crim1 maintains retinal vascular stability during development by regulating endothelial cell Vegfa autocrine signaling. Development. 2014;141(2):448-59 Maes H, Kuchnio A, Peric A, Moens S, Nys K, De Bock K, Quaegebeur A, Schoors S, Georgiadou M, Wouters J, Vinckier S, Vankelecom H, Garmyn M, Vion AC, Radtke F, Boulanger C, Gerhardt H, Dejana E, Dewerchin M, Ghesquière B, Annaert W, Agostinis P, Carmeliet P. Tumor vessel normalization by chloroquine independent of autophagy. Cancer Cell. 2014;26(2):190-206 Mleynek TM, Chan AC, Redd M, Gibson CC, Davis CT, Shi DS, Chen T, Carter KL, Ling J, Blanco R, Gerhardt H, Whitehead K, Li DY. Lack of CCM1 induces hypersprouting and impairs response to flow. Hum Mol Genet. 2014;23(23):6223-34 Schoors S, De Bock K, Cantelmo AR, Georgiadou M, Ghesquière B, Cauwenberghs S, Kuchnio A, Wong BW, Quaegebeur A, Goveia J, Bifari F, Wang X, Blanco R, Tembuyser B, Cornelissen I, Bouché A, Vinckier S, Diaz-Moralli S, Gerhardt H, Telang S, Cascante M, Chesney J, Dewerchin M, Carmeliet P. Partial and transient reduction of glycolysis by PFKFB3 blockade reduces pathological angiogenesis. Cell Metab. 2014;19(1):37-48 Wälchli T, Mateos JM, Weinman O, Babic D, Regli L, Hoerstrup SP, Gerhardt H, Schwab ME, Vogel J. Quantitative assessment of angiogenesis,perfused blood vessel and endothelial tip cells in postnatal mouse brain. Nat Protoc. 2014;doi: 10.1038/ nprot.2015.002 Other Publication Wacker A, Gerhardt H, Phng LK. Tissue guidance without filopodia. Commun Integr Biol. 2014;7:e28820 RESEARCH PUBLICATIONS 165 Nathan Goehring (page 78) Nicholas Luscombe (page 84) Developmental Systems Computational Biology Primary Research Paper Trong PK, Nicola EM, Goehring NW, Kumar KV, Grill SW. Parameter-space topology of models for cell polarity. New J Phys. 2014;16:065009 Primary Research Paper Castelnuovo M, Zaugg JB, Guffanti E, Maffioletti A, Camblong J, Xu Z, Clauder-Münster S, Steinmetz LM, Luscombe NM, Stutz F. Role of histone modifications and early termination in pervasive transcription and antisense-mediated gene silencing in yeast. Nucleic Acids Res. 2014;42(7):4348-62 Other Publication Goehring NW. PAR polarity: From complexity to design principles. Exp Cell Res. 2014;328(2):258-66 Adrian Hayday (page 80) Ilaria Malanchi (page 86) Tumour Host Interaction Immuno Surveillance Primary Research Papers Gibbons D, Fleming P, Virasami A, Michel ML, Sebire NJ, Costeloe K, Carr R, Klein N, Hayday A. Interleukin-8 (CXCL8) production is a signatory T cell effector function of human newborn infants. Nat Med. 2014;20(10):1206-10 Other Publication Ombrato L, Malanchi I. The EMT universe: Space between cancer cell dissemination and metastasis initiation. Crit Rev Oncog. 2014;19(5):349-61 Vantourout P, Willcox C, Turner A, Swanson CM, Haque Y, Sobolev O, Grigoriadis A, Tutt A, Hayday A. Immunological visibility: posttranscriptional regulation of human NKG2D ligands by the EGF receptor pathway. Sci Transl Med. 2014;6(231):231ra49 Other Publications Hayday A, Vantourout P. A long-playing CD about the γδ TCR repertoire. Immunity. 2014;39(6):994-6 Vantourout P, Hayday A. Regulation of immunological visibility by the EGF receptor. Med Sci (Paris). 2014;30(8-9):742-4 Caroline Hill (page 82) Developmental Signalling Other Publications Gaarenstroom T, Hill CS. TGF-β signaling to chromatin: How Smads regulate transcription during self-renewal and differentiation. Semin Cell Dev Biol. 2014;32:107-18 Vizán P, Miller DSJ, Schmierer B, Hill CS. Response to comment on ‘Controlling long-term signaling: Receptor dynamics determine attenuation and refractory behavior of the TGF-β pathway’-Smad2/3 activity does not predict the dynamics of transcription. Sci Signal. 2014;7(344):lc2 166 Scientific report 2014 LONDON RESEARCH INSTITUTE The murine small intestinal epithelium is normally rich in γδ T cells expressing T cell receptors (TCRs) composed of Vγ7Vδ4 chains not found anywhere else in the body and detected by confocal microscopy of the gut (pink-stained cells; top panel). Conversely, an almost total loss of such signatory intestinal T cells (lower panel) in mice mutant in a single intestinal epithelium-specific member of the B7 supergene family, permits the identification of the first, epithelial regulator of a gut-specific T cell compartment potentially involved in immune-surveillance. Key: pink – Vγ7+Vδ4+ cells; orange – Vγ7-Vδ4+ cells; blue – Vγ7+Vδ4- cells; white – Vγ7-Vδ4- γδ cells; green – αβ T cells Neil McDonald (page 88) Structural Biology Peter Parker (page 92) Protein Phosphorylation Primary Research Papers Fisher R, Horswell S, Rowan A, Salm MP, de Bruin EC, Gulati S, McGranahan N, Stares M, Gerlinger M, Varela I, Crockford A, Favero F, Quidville V, André F, Navas C, Grönroos E, Nicol D, Hazell S, Hrouda D, O Brien T, Matthews N, Phillimore B, Begum S, Rabinowitz A, Biggs J, Bates PA, McDonald NQ, Stamp G, Spencer-Dene B, Hsieh JJ, Xu J, Pickering L, Gore M, Larkin J, Swanton C. Development of synchronous VHL syndrome tumors reveals contingencies and constraints to tumor evolution. Genome Biol. 2014;15(8):433 Primary Research Papers Brownlow N, Pike T, Zicha D, Collinson L, Parker PJ. Mitotic catenation is monitored and resolved by a PKCepsilon-regulated pathway. Nat Commun. 2014; 5:5685 Goodman KM, Kjær S, Beuron F, Knowles PP, Nawrotek A, Burns EM, Purkiss AG, George R, Santoro M, Morris EP, McDonald NQ. RET recognition of GDNF-GFRα1 ligand by a composite binding site promotes membrane-proximal self-association. Cell Rep. 2014;8(6):1894-904 Gatliff J, East D, Crosby J, Abeti R, Harvey R, Craigen W, Parker PJ, Campanella M. TSPO interacts with VDAC1 and triggers a ROS-mediated inhibition of mitochondrial quality control. Autophagy. 2014;10(12):2279-96 Plaza-Menacho I, Barnouin K, Goodman K, Martínez-Torres RJ, Borg A, Murray-Rust J, Mouilleron S, Knowles P, McDonald NQ. Oncogenic RET kinase domain mutations perturb the autophosphorylation trajectory by enhancing substrate presentation in trans. Mol Cell. 2014;53(5):738-51 Other Publications Linch M, Riou P, Claus J, Cameron AJ, de Naurois J, Larijani B, Ng T, McDonald NQ, Parker PJ. Functional implications of assigned, assumed and assembled PKC structures. Biochem Soc Trans. 2014;42(1):35-41 Plaza-Menacho I, Mologni L, McDonald NQ. Mechanisms of RET signaling in cancer: current and future implications for targeted therapy. Cell Signal. 2014;26(8):1743-52 Paul Nurse/Jacqueline Hayles (page 90) Cell Cycle Primary Research Paper Wu PY, Nurse P. Replication origin selection regulates the distribution of meiotic recombination. Mol Cell. 2014;53(4):655-62 Other Publication Nurse P. EMBO at 50. Science. 2014;343(6167):117 Endesfelder D, Burrell RA, Kanu N, McGranahan N, Howell M, Parker PJ, Downward J, Swanton C, Kschischo M. Chromosomal instability selects gene copy-number variants encoding core regulators of proliferation in ER+ breast cancer. Cancer Res. 2014;74(17):4853-63 Kiuchi T, Kiuchi T, Ortiz-Zapater E, Monypenny J, Matthews DR, Nguyen LK, Barbeau J, Coban O, Lawler K, Burford B, Rolfe DJ, de Rinaldis E, Dafou D, Simpson MA, Woodman N, Pinder S, Gillett CE, Devauges V, Poland SP, Fruhwirth G, Marra P, Boersma YL, Plückthun A, Gullick WJ, Yarden Y, Santis G, Winn M, Kholodenko BN, Martin-Fernandez ML, Parker PJ, Tutt A, Ameer-Beg SM, Ng T. The ErbB4 CYT2 variant protects EGFR from ligand-induced degradation to enhance cancer cell motility. Sci Signal. 2014; 7(339): ra78 Ménard L, Parker PJ, Kermorgant S. Receptor tyrosine kinase c-Met controls the cytoskeleton from different endosomes via different pathways. Nat Commun. 2014;5:3907 Rossé C, Lodillinsky C, Fuhrmann L, Nourieh M, Monteiro P, Irondelle M, Lagoutte E, Vacher S, Waharte F, Paul-Gilloteaux P, Romao M, Sengmanivong L, Linch M, van Lint J, Raposo G, Vincent-Salomon A, Bièche I, Parker PJ, Chavrier P. Control of MT1-MMP transport by atypical PKC during breast-cancer progression. Proc Natl Acad Sci USA. 2014;111(18):E1872-9 Veeriah S, Leboucher P, de Naurois J, Jethwa N, Nye E, Bunting T, Stone R, Stamp G, Calleja V, Jeffrey SS, Parker PJ, Larijani B. High-throughput time-resolved FRET reveals Akt/PKB activation as a poor prognostic marker in breast cancer. Cancer Res. 2014;74(18):4983-95 RESEARCH PUBLICATIONS 167 Other Publications Brownlow N, Pike T, Crossland V, Claus J, Parker PJ. Regulation of the cytokinesis cleavage furrow by PKCepsilon. Biochem Soc Trans. 2014;42(6):1534-37 Calleja V, Laguerre M, de Las Heras-Martinez G, Parker PJ, Requejo-Isidro J, Larijani B. Acute regulation of PDK1 by a complex interplay of molecular switches. Biochem Soc Trans. 2014;42(5):1435-40 Claus J, Patel G, Ng T, Parker PJ. A role for the pseudokinase HER3 in the acquired resistance against EGFR- and HER2-directed targeted therapy. Biochem Soc Trans. 2014;42(4):831-6 Linch M, Riou P, Claus J, Cameron AJ, de Naurois J, Larijani B, Ng T, McDonald NQ, Parker PJ. Functional implications of assigned, assumed and assembled PKC structures. Biochem Soc Trans. 2014;42(1):35-41 Martin-Liberal J, Cameron AJ, Claus J, Judson IR, Parker PJ, Linch M. Targeting protein kinase C in sarcoma. Biochim Biophys Acta. 2014;1846(2):547-59 Parker PJ, Justilien V, Riou P, Linch M, Fields AP. Atypical protein kinase Cι as a human oncogene and therapeutic target. Biochem Pharmacol. 2014;88(1):1-11 Caetano Reis e Sousa (page 94) Immunobiology Primary Research Papers Acton SE, Farrugia AJ, Astarita JL, Mourão-Sá D, Jenkins RP, Nye E, Hooper S, van Blijswijk J, Rogers NC, Snelgrove KJ, Rosewell I, Moita LF, Stamp G, Turley SJ, Sahai E, Reis e Sousa C. Dendritic cells control fibroblastic reticular network tension and lymph node expansion. Nature. 2014;14(7523):498-502 Bär E, Whitney PG, Moor K, Reis e Sousa C, LeibundGut-Landmann S. IL-17 regulates systemic fungal immunity by controlling the functional competence of NK cells. Immunity. 2014;40(1): 117-27 Deddouche S, Goubau D, Rehwinkel J, Chakravarty P, Begum S, Maillard PV, Borg AP, Matthews N, Feng Q, van Kuppeveld FJM, Reis e Sousa C. Identification of an LGP2-associated MDA5 agonist in picornavirusinfected cells. Elife. 2014;3:e01535 Goubau D, Schlee M, Deddouche S, Pruijssers AJ, Zillinger T, Goldeck M, Schuberth C, Van der Veen AG, Fujimura T, Rehwinkel J, Iskarpatyoti JA, Barchet W, Ludwig J, Dermody TS, Hartmann G, Reis e Sousa C. Antiviral immunity via RIG-I-mediated recognition of RNA bearing 5’-diphosphates. Nature. 2014;514(7522):372-5 Li J, Ahmet F, Sullivan LC, Brooks AG, Kent SJ, De Rose R, Salazar AM, Reis e Sousa C, Shortman K, Lahoud MH, Heath WR, Caminschi I. Antibodies targeting Clec9A promote strong humoral immunity without adjuvant in mice and non-human primates. Eur J Immunol. 2014;doi: 10.1002/eji.201445127 van Blijswijk J, Schraml BU, Rogers NC, Whitney PG, Zelenay S, Acton SE, Reis e Sousa C. Altered lymph node composition in diphtheria toxin receptor-based mouse models to ablate dendritic cells. J Immunol. 2014;doi: 10.4049/jimmunol.1401999 Whitney PG, Bär E, Osorio F, Rogers NC, Schraml BU, Deddouche S, LeibundGut-Landmann S, Reis e Sousa C. Syk signaling in dendritic cells orchestrates innate resistance to systemic fungal infection. PLoS Pathog. 2014;10(7):e1004276 Breast cancer (bottom right area) and adjacent normal tissue (top left). 168 Scientific report 2014 LONDON RESEARCH INSTITUTE Other Publications Reis e Sousa C, Unanue ER. Antigen processing. Curr Opin Immunol. 2014;26:138-9 Martin Singleton (page 100) Macromolecular Structure and Function Schraml BU, Reis e Sousa C. Defining dendritic cells. Curr Opin Immunol. 2014;doi: 10.1016/j. coi.2014.11.001 Primary Research Paper Drechsler H, McHugh T, Singleton MR, Carter NJ, McAinsh AD. The Kinesin-12 Kif15 is a processive track-switching tetramer. Elife. 2014;3,e01724 Erik Sahai (page 96) Thomas Surrey (page 102) Tumour Cell Biology Primary Research Papers Acton SE, Farrugia AJ, Astarita JL, Mourão-Sá D, Jenkins RP, Nye E, Hooper S, van Blijswijk J, Rogers NC, Snelgrove KJ, Rosewell I, Moita LF, Stamp G, Turley SJ, Sahai E, Reis e Sousa C. Dendritic cells control fibroblastic reticular network tension and lymph node expansion. Nature. 2014;514(7523):498-502 Manning CS, Hooper S, Sahai EA. Intravital imaging of SRF and Notch signalling identifies a key role for EZH2 in invasive melanoma cells. Oncogene. 2014;doi:10.1038/onc.2014.362 Other Publications Charras G, Sahai E. Physical influences of the extracellular environment on cell migration. Nat Rev Mol Cell Biol. 2014;15(12):813-24 Hirata E, Park D, Sahai E. Retrograde flow of cadherins in collective cell migration. Nat Cell Biol. 2014;16(7):621-3 Huttenlocher A, Sahai E. Editorial overview: cell adhesion and migration. Curr Opin Cell Biol. 2014; 30:v-vi Microtubule Cytoskeleton Primary Research Papers Baumann H, Surrey T. Motor-mediated cortical versus astral microtubule organisation in lipid-monolayered droplets. J Biol Chem. 2014;89(32):22524-35 Duellberg C, Trokter M, Jha R, Sen I, Steinmetz MO, Surrey T. Reconstitution of a hierarchical +TIP interaction network controlling microtubule end tracking of the human dynein complex. Nat Cell Biol. 2014;16(8):804-11 Maurer SP, Cade NI, Bohner G, Gustafsson N, Boutant E, Surrey T. EB1 accelerates two conformational transitions important for microtubule maturation and dynamics. Curr Biol. 2014;24(4):372-84 Other Publications Fourniol FJ, Li TD, Bieling P, Mullins RD, Fletcher DA, Surrey T. Micropattern-guided assembly of overlapping pairs of dynamic microtubules. Methods Enzymol. 2014;540:339-60 Maurer SP, Fourniol FJ, Hoenger A, Surrey T. Seeded microtubule growth for cryoelectron microscopy of end-binding proteins. Methods Mol Biol. 2014;1136:247-60 Paola Scaffadi (page 98) Cancer Epigenetics Primary Research Papers Ben-David U, Biran A, Scaffidi P, Herold-Mende C, Boehringer M, Meshorer E, Benvenisty N. Elimination of undifferentiated cancer cells by pluripotent stem cell inhibitors. J Mol Cell Biol. 2014;6(3):267-9 Fernandez P, Scaffidi P, Markert E, Lee J, Rane R, Misteli T. Transformation resistance in a premature aging disorder identifies a tumor-protective function of BRD4. Cell Rep. 2014;9(1):248-60 RESEARCH PUBLICATIONS 169 Charles Swanton (page 104) Translational Cancer Therapeutics Primary Research Papers de Bruin E, McGranahan N, Mitter R, Salm M, Wedge DC, Yates L, Jamal-Hanjani M, Shafi S, Murugaesu N, Rowan AJ, Gronroos E, Muhammad MA, Horswell S, Gerlinger M, Varela I, Jones D, Marshall J, Voet T, Van Loo P, Rassl DM, Rintoul RC, Janes SM, Lee S, Forster M, Ahmed T, Lawrence D, Falzon M, Capitanio A, Harkins TT, Lee CC, Tom W, Teefe E, Chen SC, Begum S, Rabinowitz A, Phillimore B, Spencer-Dene B, Stamp G, Szallasi Z, Matthews N, Stewart A, Campbell P, Swanton C. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science. 2014; 346(6206):251-6 Dewhurst SM, McGranahan N, Burrell RA, Rowan AJ, Grönroos E, Endesfelder D, Joshi T, Mouradov D, Gibbs P, Ward RL, Hawkins NJ, Szallasi Z, Sieber OM, Swanton C. Tolerance of whole-genome doubling propagates chromosomal instability and accelerates cancer genome evolution. Cancer Discov. 2014;4(2):175-85 Endesfelder D, Burrell R, Kanu N, McGranahan N, Howell M, Parker PJ, Downward J, Swanton C, Kschischo M. Chromosomal instability selects gene copy number variants encoding core regulators of proliferation in ER+ breast cancer. Cancer Res. 2014;74(17):4853-63 Fisher R, Horswell S, Rowan A, Salm MP, de Bruin EC, Gulati S, McGranahan N, Stares M, Gerlinger M, Varela I, Crockford A, Favero F, Quidville V, André F, Navas C, Grönroos E, Nicol D, Hazell S, Hrouda D, O Brien T, Matthews N, Phillimore B, Begum S, Rabinowitz A, Biggs J, Bates PA, McDonald NQ, Stamp G, Spencer-Dene B, Hsieh JJ, Xu J, Pickering L, Gore M, Larkin J, Swanton C. Development of synchronous VHL syndrome tumors reveals contingencies and constraints to tumor evolution. Genome Biol. 2014;15(8):433 Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I, Fisher R, McGranahan N, Matthews N, Santos CR, Martinez P, Phillimore B, Begum S, Rabinowitz A, Spencer-Dene B, Gulati S, Bates PA, Stamp G, Pickering L, Gore M, Nicol DL, Hazell S, Futreal PA, Stewart A, Swanton C. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet. 2014;46(3):225-33 170 Scientific report 2014 LONDON RESEARCH INSTITUTE Gulati S, Martinez P, Joshi T, Birkbak N, Santos CR, Rowlan AJ, Pickering L, Gore M, Larkin J, Szallasi Z, Bates PA, Swanton C, Gerlinger M. Systematic evaluation of the prognostic impact and intratumour heterogeneity of clear cell renal cell carcinoma biomarkers. Eur Urol. 2014;66(5):936-48 Lönnstedt IM, Caramia F, Li J, Fumagalli D, Salgado R, Rowan A, Salm M, Kanu N, Savas P, Horswell S, Gade S, Loibl S, Neven P, Sotiriou C, Swanton C, Loi S, Speed TP. Deciphering clonality in aneuploid breast tumors using SNP array and sequencing data. Genome Biol. 2014;15(9):470. Molnár J, Póti A, Pipek O, Krzystanek M, Kanu N, Swanton C, Tusnády GE, Szállási Z, Csabai I, Szüts D. The genome of the chicken DT40 bursal lymphoma cell line. G3 (Bethesda). 2014;4(11):2231-40 Ng CK, Weigelt B, A’Hern R, Bidard FC, Lemetre C, Swanton C, Shen R, Reis-Filho JS. Predictive performance of microarray gene signatures: impact of tumor heterogeneity and multiple mechanisms of drug resistance. Cancer Res. 2014;74(11):2946-61 Roylance R, Endesfelder D, Jamal-Hanjani M, Burrell RA, Gorman P, Sander J, Murphy N, Birkbak NJ, Hanby AM, Speirs V, Johnston SR, Kschischo M, Swanton C. Expression of regulators of mitotic fidelity are associated with intercellular heterogeneity and chromosomal instability in primary breast cancer. Breast Cancer Res Treat. 2014;148(1):221-9 Staples CJ, Myers KN, Beveridge RD, Patil AA, Howard AE, Barone G, Lee AJ, Swanton C, Howell M, Maslen S, Skehel JM, Boulton SJ, Collis SJ. Ccdc13 is a novel human centriolar satellite protein required for ciliogenesis and genome stability. J Cell Sci. 2014;127(Pt 13):2910-9 Other Publications Bakhoum SF, Swanton C. Chromosomal instability, aneuploidy, and cancer. Front Oncol. 2014;4:161 Burrell RA, McClelland SE, Bartek J, Swanton C. Response to Bakhoum et al. Curr Biol. 2014;24(4):R150 Burrell RA, Swanton C. The evolution of the unstable cancer genome. Curr Opin Genet Dev. 2014;24:61-7 Burrell RA, Swanton C. Tumour heterogeneity and the evolution of polyclonal drug resistance. Mol Oncol. 2014;8(6):1095-111 Crockford A, Jamal-Hanjani M, Hicks J, Swanton C. Implications of intratumour heterogeneity for treatment stratification. J Pathol. 2014;232(2):264-73 Gerlinger M, Catto JW, Orntoft TF, Real FX, Zwarthoff EC, Swanton C. Intratumour Heterogeneity in Urologic Cancers: From Molecular Evidence to Clinical Implications. Eur Urol. 2014;doi: 10.1016/j. eururo.2014.04.014 Gerlinger M, McGranahan N, Dewhurst SM, Burrell RA, Tomlinson I, Swanton C. Cancer: evolution within a lifetime. Annu Rev Genet. 2014;48:215-36. Hiley C, de Bruin EC, McGranahan N, Swanton C. Deciphering intratumor heterogeneity and temporal acquisition of driver events to refine precision medicine. Genome Biol. 2014;15(8):453 Infante JR, Swanton C. Combined inhibition of BRAF and MEK in melanoma patients. Lancet Oncol. 2014;15(9):908-10 Jamal-Hanjani M, Hackshaw A, Ngai Y, Shaw J, Dive C, Quezada S, Middleton G, de Bruin E, Le Quesne J, Shafi S, Falzon M, Horswell S, Blackhall F, Khan I, Janes S, Nicolson M, Lawrence D, Forster M, Fennell D, Lee SM, Lester J, Kerr K, Muller S, Iles N, Smith S, Murugaesu N, Mitter R, Salm M, Stuart A, Matthews N, Adams H, Ahmad T, Attanoos R, Bennett J, Birkbak NJ, Booton R, Brady G, Buchan K, Capitano A, Chetty M, Cobbold M, Crosbie P, Davies H, Denison A, Djearman M, Goldman J, Haswell T, Joseph L, Kornaszewska M, Krebs M, Langman G, MacKenzie M, Millar J, Morgan B, Naidu B, Nonaka D, Peggs K, Pritchard C, Remmen H, Rowan A, Shah R, Smith E, Summers Y, Taylor M, Veeriah S, Waller D, Wilcox B, Wilcox M, Woolhouse I, McGranahan N, Swanton C. Tracking genomic cancer evolution for precision medicine: The lung TRACERx study. PLoS Biol. 2014;12(7):e1001906 Stewart GD, Harrison DJ, Swanton C, Lewis R, Bliss J, Larkin J, Nicol DL; A‐PREDICT TMG. Multidisciplinary urological engagement in translational renal cancer research. BJU Int. 2014;114(4):474-5 Swanton C. Cancer Evolution: The Final Frontier of Precision Medicine? Ann Oncol. 2014;25(3):549-51 SIM image of the actin cytoskeleton (white) in Drosophila hemocytes. Swanton C, André F, Mardis E. Deciphering root causes of intrinsic BRAF inhibitor resistance in melanoma: ushering in a new genomics case reports feature for Annals of Oncology. Ann Oncol. 2014;25(5):917-8 Swanton C, Beck S. Epigenetic noise fuels cancer evolution. Cancer Cell. 2014;26(6):775-6 Nicolas Tapon (page 106) Apoptosis and Proliferation Control Primary Research Papers Doughton G, Wei J, Tapon N, Welham, MJ, Chalmers AD. Formation of a polarised primitive endoderm layer in embryoid bodies requires fgfr/erk signalling. PloS One. 2014;9(4):e95434 Ribeiro P, Holder M, Frith D, Snijders AP, Tapon N. Crumbs promotes expanded recognition and degradation by the SCFSlimb/beta-TrCP ubiquitin ligase. Proc Nat Acad Sci USA. 2014;111(19):E1980-9 Other Publication Gaspar P, Tapon N. Sensing the local environment: actin architecture and Hippo signalling. Curr Opin Cell Biol. 2014;31C:74-8 Swanton C. SAFIR01: steps towards precision treatment in breast cancer. Lancet Oncol. 2014;15(3):242-3 RESEARCH PUBLICATIONS 171 Barry Thompson (page 108) Sharon Tooze (page 112) Epithelial Biology Secretory Pathways Primary Research Papers Bell GP, Fletcher G, Brain R, Thompson BJ. Aurora kinases phosphorylate Lgl to induce mitotic spindle orientation in Drosophila epithelia. Curr Biol. 2014;doi: 10.1016/j.cub.2014.10.052 Primary Research Papers Dooley HC, Razi M, Polson HEJ, Girardin SE, Wilson MI, Tooze SA. WIPI2 links LC3-conjugation with PI3P, Autophagosome formation and pathogen clearance by recruiting Atg12–5-16L1. Mol Cell. 2014;55(2):238-52 Rodrigues-Campos M, Thompson BJ. The ubiquitin ligase FbxL7 regulates the Dachsous-Fat-Dachs system in Drosophila. Development. 2014;141(21):4098-103 Other Publication Bell GP, Thompson BJ. Colorectal cancer progression: Lessons from Drosophila? Semin Cell Dev Biol. 2014;28:70-7 Takashi Toda (page 110) Cell Regulation Primary Research Papers Hori A, Ikebe C, Tada M, Toda T. Msd1/SSX2IPdependent microtubule anchorage ensures spindle orientation and primary cilia formation. EMBO Rep. 2014;15(2):175-84 Okada O, Toda T, Yamamoto M, Sato M. CDK-dependent phosphorylation of Alp7–Alp14 (TACC–TOG) promotes its nuclear accumulation and spindle microtubule assembly. Mol Biol Cell. 2014;25(13):1969-82 Tang NH, Okada N, Fong CS, Arai K, Sato M, Toda T. Targeting Alp7/TACC to the spindle pole body is essential for mitotic spindle assembly in fission yeast. FEBS Lett. 2014;588(17):2814-21 Tang NH, Toda T. Alp7/TACC recruits kinesin-8-PP1 to the Ndc80 kinetochore protein for timely mitotic progression and chromosome movement. J Cell Sci. 2014;doi: 10.1242/jcs.160036 Duke EM, Razi M, Weston A, Guttmann P, Werner S, Henzler K, Schneider G, Tooze SA, Collinson LM. Imaging endosomes and autophagosomes in whole mammalian cells using correlative cryo-fluorescence and cryo-soft X-ray microscopy (cryo-CLXM). Ultramicroscopy. 2014;143:77-87 Ellis RA, Horswell S, Ness T, Lumsdon J, Tooze SA, Kirkham N, Armstrong JL, Lovat PE. Prognostic impact of p62 expression in cutaneous malignant melanoma. J Invest Dermatol. 2014;134(5):1476-8 Henson SM, Lanna A, Riddell NE, Franzese O, Macaulay R, Griffiths SJ, Puleston DJ, Watson AS, Simon AK, Tooze SA, Akbar AN. p38 signaling inhibits mTORC1-independent autophagy in senescent human CD8+ T cells. J Clin Invest. 2014;124(9):4004-16 Talaber G, Miklossy G, Oaks Z, Liu Y, Tooze SA, Chudakov DM, Banki K, Perl A. HRES-1/Rab4 promotes the formation of LC3(+) autophagosomes and the accumulation of mitochondria during autophagy. PLoS One. 2014;9(1):e84392 Other Publications Tooze SA, Abada A, Elazar Z. Endocytosis and autophagy: exploitation or cooperation? Cold Spring Harb Perspect Biol. 2014;6(5):a018358 Wilson MI, Dooley HC, Tooze SA. WIPI2b and Atg16L1: setting the stage for autophagosome formation. Biochem Soc Trans. 2014;42(5):1327-34 Richard Treisman (page 114) Signalling and Transcription Primary Research Paper Esnault C, Stewart A, Gualdrini F, East P, Horswell S, Matthews N, Treisman R. Rho-actin signaling to the MRTF coactivators dominates the immediate transcriptional response to serum in fibroblasts. Genes Dev. 2014;28(9):943-58 172 Scientific report 2014 LONDON RESEARCH INSTITUTE Other Publication Yu-Wai-Man C, Treisman R, Bailly M, Khaw PT. The role of the MRTF-A/SRF pathway in ocular fibrosis. Invest Ophthalmol Vis Sci. 2014;55(7):4560-7 Frank Uhlmann (page 116) Chromosome Segregation Primary Research Papers Lopez-Serra L, Kelly G, Patel H, Stewart A, Uhlmann F. The Scc2-Scc4 complex acts in sister chromatid cohesion and transcriptional regulation by maintaining nucleosome-free regions. Nat Genet. 2014;46(10):1147-51 Murayama Y, Uhlmann F. Biochemical reconstitution of topological DNA binding by the cohesin ring. Nature. 2014;505(7483):367-71 Other Publication Uhlmann F. A silent revolution in chromosome biology. Nat Rev Mol Cell Biol. 2014;15(7):431 Drosophila wing imaginal discs expressing expanded-GFP (green) and ubi-RFP (red). Michael Way (page 122) Cell Motility Peter Van Loo (page 118) Cancer Genomics Primary Research Papers de Bruin EC, McGranahan N, Mitter R, Salm M, Wedge DC, Yates L, Jamal-Hanjani M, Shafi S, Murugaesu N, Rowan AJ, Grönroos E, Muhammad MA, Horswell S, Gerlinger M, Varela I, Jones D, Marshall J, Voet T, Van Loo P, Rassl DM, Rintoul RC, Janes SM, Lee SM, Forster M, Ahmad T, Lawrence D, Falzon M, Capitanio A, Harkins TT, Lee CC, Tom W, Teefe E, Chen SC, Begum S, Rabinowitz A, Phillimore B, Spencer-Dene B, Stamp G, Szallasi Z, Matthews N, Stewart A, Campbell P, Swanton C. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science. 2014;346(6206):251-6. Rashid NU, Sperling AS, Bolli N, Wedge DC, Van Loo P, Tai YT, Shammas MA, Fulciniti M, Samur MK, Richardson PG, Magrangeas F, Minvielle S, Futreal PA, Anderson KC, Avet-Loiseau H, Campbell PJ, Parmigiani G, Munshi NC. Differential and limited expression of mutant alleles in multiple myeloma. Blood. 2014;124(20):3110-7 Primary Research Papers Chen XJ, Squarr JA, Stephan R, Chen B, Higgins TE, Barry DJ, Martin MC, Rosen MK, Bogdan S, Way M. Ena/VASP proteins cooperate with the WAVE complex to regulate the actin cytoskeleton. Dev Cell. 2014;30(5):569-84 Gill MB, Turner R, Stevenson PG, Way M. KSHV-TK is a tyrosine kinase that disrupts focal adhesions and induces Rho-mediated cell contraction. EMBO J. 2014;pii:e201490358 Humphries AC, Donnelly SK, Way M. Cdc42 and the RhoGEF Intersectin-1 collaborate with Nck to promote N-WASP-dependent actin polymerization. J Cell Sci. 2014;127(Pt 3):673-685 Young JC, Clements A, Lang AE, Garnett JA, Munera D, Arbeloa A, Pearson J, Hartland EL, Matthews SJ, Mousnier A, Barry DJ, Way M, Schlosser A, Aktories K, Frankel G. The Escherichia coli effector EspJ blocks Src kinase activity via amidation and ADP ribosylation. Nat Commun. 2014; 5:5887 Other Publication Way M. JCS Prize. 2013 winner: Liam Cheeseman. J Cell Sci. 2014;127(Pt 10):2121 RESEARCH PUBLICATIONS 173 Simon Boulton (page 126) DNA Damage Response Primary Research Papers Beveridge RD, Staples CJ, Patil AA, Myers KN, Maslen S, Skehel JM, Boulton SJ, Collis SJ. The leukemia-associated Rho guanine nucleotide exchange factor LARG is required for efficient replication stress signaling. Cell Cycle. 2014;13(21):3450-9 Frizzell A, Nguyen JH, Petalcorin MI, Turner KD, Boulton SJ, Freudenreich CH, Lahue RS. RTEL1 inhibits trinucleotide repeat expansions and fragility. Cell Rep. 2014;6(5):827-35 Hořejší Z, Stach L, Flower TG, Joshi D, Flynn H, Skehel JM, O’Reilly N, Ogrodowicz RW, Smerdon SJ, Boulton SJ. Phosphorylation-dependent PIH1D1 interactions define substrate specificity of the R2TP co-chaperone complex. Cell Rep. 2014;6(5):827-35 Staples CJ, Myers KN, Beveridge RDD, Patil AA, Howard AE, Giancarlo B, Lee AJX, Swanton C, Howell M, Maslen S, Skehel JM, Boulton SJ, Collis SJ. Ccdc13 is a novel human centriolar satellite protein required for ciliogenesis and genome stability. J Cell Sci. 2014;127(Pt 13):2910-9 Other Publications León-Ortiz AM, Svendsen J, Boulton SJ. Metabolism of DNA secondary structures at the eukaryotic replication fork. DNA Repair (Amst). 2014;19:152-62 Panier S, Boulton SJ. Double-strand break repair: 53BP1 comes into focus. Nat Rev Mol Cell Biol. 2014;15(1):7-18 Vannier JB, Sarek G, Boulton SJ. RTEL1: functions of a disease-associated helicase. Trends Cell Biol. 2014;24(7):416-25 Peter Cherepanov (page 128) Chromatin Structure and DNA Primary Research Papers Maertens GN, Cook NJ, Wang W, Hare S, Gupta SS, Öztop I, Lee K, Pye VE, Cosnefroy O, Snijders AP, KewalRamani VN, Fassati A, Engelman A, Cherepanov P. Structural basis for nuclear import of splicing factors by human Transportin 3. Proc Natl Acad Sci USA. 2014;111(7):2728-33 174 Scientific report 2014 LONDON RESEARCH INSTITUTE Serrao E, Krishnan L, Shun MC, Li X, Cherepanov P, Engelman A, Maertens GN. Integrase residues that determine nucleotide preferences at sites of HIV-1 integration: implications for the mechanism of target DNA binding. Nucleic Acids Res. 2014;42(8):5164-76 Wang H, Shun M-C, Li X, Di Nunzio F, Hare S, Cherepanov P, Engelman A. Efficient transduction of LEDGF/p75 mutant cells by complementary gain-offunction HIV-1 integrase mutant viruses. Mol Ther Methods Clin Dev. 2014;1: 10.1038/mtm.2013.2. Other Publication Engelman A, Cherepanov P. Retroviral Integrase Structure and DNA Recombination Mechanism. Microbiol Spectr. 2014;2(6):1-22 Alessandro Costa (page 130) Architecture and Dynamics of Macromolecular Machines Primary Research Papers Costa A, Renault L, Swuec P, Petojevic T, Pesavento J, Ilves I, MacLellan-Gibson K, Fleck RA, Botchan MR, Berger JM. DNA binding polarity, dimerization, and ATPase ring remodeling in the CMG helicase of the eukaryotic replisome. Elife. 2014;3:e03273 Simon AC, Zhou JC, Perera RL, van Deursen F, Evrin C, Ivanova ME, Kilkenny ML, Renault L, Kjær S, Matak-Vinković D, Labib K, Costa A*, Pellegrini L*. A Ctf4 trimer couples the CMG helicase to DNA polymerase α in the eukaryotic replisome. Nature. 2014;510(7504):293-7. (*co-corresponding authors). Other Publications Swuec P, Costa A. Molecular mechanism of double Holliday junction dissolution. Cell Biosci. 2014;4:36 Zhou JC, Costa A. Preparing to unwind. Elife. 2014;3:e02618. John Diffley (page 132) Chromosome Replication Primary Research Papers Coster G, Frigola J, Beuron F, Morris EP, Diffley JF. Origin licensing requires ATP binding and hydrolysis by the MCM replicative helicase. Mol Cell. 2014;55(5):666-77 On KF, Beuron F, Frith D, Snijders AP, Morris EP, Diffley JF. Prereplicative complexes assembled in vitro support origin-dependent and independent DNA replication. EMBO J. 2014;33(6):605-20 Other Publication Hills SA, Diffley JF. DNA replication and oncogeneinduced replicative stress. Curr Biol. 2014;24(10):R435-44 Drosophila larval central nervous system marked with ELAV (red) and 91Y-GFP (green). Peter Karran (page 134) Mammalian DNA Repair Primary Research Papers Grasso F, Ruggieri V, De Luca G, Leopardi P, Mancuso MT, Casorelli I, Pichierri P, Karran P, Bignami M. MUTYH mediates the toxicity of combined DNA 6-thioguanine and UVA radiation. Oncotarget. 2014 Gueranger Q, Li F, Peacock M, Larnicol-Fery A, Brem R, Macpherson P, Egly JM, Karran P. Protein oxidation and DNA repair inhibition by 6-Thioguanine and UVA radiation. J Invest Dermatol. 2014;134(5):1408-17 Peacock M, Brem R, Macpherson P, Karran P. DNA repair inhibition by UVA photoactivated fluoroquinolones and vemurafenib. Nucleic Acids Res. 2014;42(22):13714-22 Other Publication Gueranger Q, Li F, Peacock M, Larnicol-Fery A, Brem R, Macpherson P, Egly JM, Karran P. Response to Emanuele. J Invest Dermatol. 2014;134(7):2053-5 Mark Petronczki (page 136) Cell Division and Aneuploidy Primary Research Papers Su KC, Bement WM, Petronczki M, von Dassow G. An astral simulacrum of the central spindle accounts for normal, spindle-less, and anucleate cytokinesis in echinoderm embryos. Mol Biol Cell. 2014;25(25):4049-62 Sundaramoorthy S, Vázquez-Novelle MD, Lekomtsev S, Howell M, Petronczki M. Functional genomics identifies a requirement of pre-mRNA splicing factors for sister chromatid cohesion. EMBO J. 2014;33(22):2623-42 Vázquez-Novelle MD, Sansregret L, Dick AE, Smith CA, McAinsh AD, Gerlich DW, Petronczki M. Cdk1 inactivation terminates mitotic checkpoint surveillance and stabilizes kinetochore attachments in anaphase. Curr Biol. 2014;24(6):638-45 Jesper Svejstrup (page 138) Mechanisms of Gene Transcription Primary Research Papers Saponaro M, Kantidakis T, Mitter R, Kelly GP, Heron M, Williams H, Söding J, Stewart A, Svejstrup JQ. RECQL5 controls transcript elongation and suppresses genome instability associated with transcription stress. Cell. 2014;157(5):1037-49 Wang Y, Chakravarty P, Ranes M, Kelly G, Brooks P, Neiland E, Stewart A, Schiavo G, Svejstrup JQ. Dysregulation of gene expression as a cause of Cockayne syndrome neurological disease. Proc Natl Acad Sci USA. 2014;111(40):14454-9 RESEARCH PUBLICATIONS 175 Stephen West (page140) Genetic Recombination Primary Research Papers Blanco MG, Matos J, West SC. Dual control of Yen1 nuclease activity and cellular localization by Cdk and Cdc14 prevents genome instability. Mol Cell. 2014;54(1):94-106 Chan YW, West SC. Spatial control of the GEN1 Holliday junction resolvase ensures genome stability. Nat Commun. 2014;5:4844 Pepe A, West SC. MUS81-EME2 promotes replication fork restart. Cell Rep. 2014;7(4):1048-55 The spread of vaccinia infected cells (green) in a cell monolayer labelled for actin (red). Pepe A, West SC. Substrate specificity of the MUS81EME2 structure selective endonuclease. Nucleic Acids Res. 2014;42(6):3833-45 Sarbajna S, Davies D, West SC. Roles of SLX1-SLX4, MUS81-EME1 and GEN1 in avoiding genome instability and mitotic catastrophe. Genes Dev. 2014;28(10):1124-36 Shahid T, Soroka J, Kong EH, Malivert L, McIlwraith MJ, Pape T, West SC, Zhang X. Structure and mechanism of action of the BRCA2 breast cancer tumor suppressor. Nat Struct Mol Biol. 2014;21(11):962-8 Other Publications Matos J, West SC. Holliday junction resolution: regulation in space and time. DNA Repair. 2014;7(4):1048-55 Hasan Yardimici (page 142) Sarbajna T, West SC. Holliday junction processing enzymes as guardians of mammalian genome stability. Trends Biochem Sci. 2014;39(9):409-19 Primary Research Paper Duxin JP, Dewar JM, Yardimci H, Walter JC. Repair of a DNA-protein crosslink by replication-coupled proteolysis. Cell. 2014;159(2):346-57 Wyatt HDM, West SC. Holliday junction resolvases. Cold Spring Harb Perspect Biol. 2014;6(9):a023192 176 Scientific report 2014 LONDON RESEARCH INSTITUTE Single Molecule Imaging Other Publication Yardimci H, Walter JC. Prereplication-complex formation: a molecular double take? Nat Struct Mol Biol. 2014;21(1):20-5 Advanced Sequencing (page 147) Nik Matthews Primary Research Papers Chandler H, Patel H, Palermo R, Brookes S, Matthews N, Peters G. Role of polycomb group proteins in the DNA damage response – a reassessment. PLoS One. 2014;9(7):e102968 de Bruin E, McGranahan N, Mitter R, Salm M, Wedge DC, Yates L, Jamal-Hanjani M, Shafi S, Murugaesu N, Rowan AJ, Gronroos E, Muhammad MA, Horswell S, Gerlinger M, Varela I, Jones D, Marshall J, Voet T, Van Loo P, Rassl DM, Rintoul RC, Janes SM, Lee S, Forster M, Ahmed T, Lawrence D, Falzon M, Capitanio A, Harkins TT, Lee CC, Tom W, Teefe E, Chen SC, Begum S, Rabinowitz A, Phillimore B, Spencer-Dene B, Stamp G, Szallasi Z, Matthews N, Stewart A, Campbell P, Swanton C. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science. 2014; 346(6206):251-6 Deddouche S, Goubau D, Rehwinkel J, Chakravarty P, Begum S, Maillard PV, Borg A, Matthews N, Feng Q, van Kuppeveld FJ, Reis e Sousa C. Identification of an LGP2-associated MDA5 agonist in picornavirus-infected cells. Elife. 2014;3:e01535 Esnault C, Stewart A, Gualdrini F, East P, Horswell S, Matthews N, Treisman R. Rho-actin signaling to the MRTF coactivators dominates the immediate transcriptional response to serum in fibroblasts. Genes Dev. 2014;28(9):943-58 Fisher R, Horswell S, Rowan A, Salm MP, de Bruin EC, Gulati S, McGranahan N, Stares M, Gerlinger M, Varela I, Crockford A, Favero F, Quidville V, André F, Navas C, Grönroos E, Nicol D, Hazell S, Hrouda D, O Brien T, Matthews N, Phillimore B, Begum S, Rabinowitz A, Biggs J, Bates PA, McDonald NQ, Stamp G, Spencer-Dene B, Hsieh JJ, Xu J, Pickering L, Gore M, Larkin J, Swanton C. Development of synchronous VHL syndrome tumors reveals contingencies and constraints to tumor evolution. Genome Biol. 2014;15(8):433 Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I, Fisher R, McGranahan N, Matthews N, Santos CR, Martinez P, Phillimore B, Begum S, Rabinowitz A, Spencer-Dene B, Gulati S, Bates PA, Stamp G, Pickering L, Gore M, Nicol DL, Hazell S, Futreal PA, Stewart A, Swanton C. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet. 2014;46(3):225-33 Jamal-Hanjani M, Hackshaw A, Ngai Y, Shaw J, Dive C, Quezada S, Middleton G, de Bruin E, Le Quesne J, Shafi S, Falzon M, Horswell S, Blackhall F, Khan I, Janes S, Nicolson M, Lawrence D, Forster M, Fennell D, Lee SM, Lester J, Kerr K, Muller S, Iles N, Smith S, Murugaesu N, Mitter R, Salm M, Stuart A, Matthews N, Adams H, Ahmad T, Attanoos R, Bennett J, Birkbak NJ, Booton R, Brady G, Buchan K, Capitano A, Chetty M, Cobbold M, Crosbie P, Davies H, Denison A, Djearman M, Goldman J, Haswell T, Joseph L, Kornaszewska M, Krebs M, Langman G, MacKenzie M, Millar J, Morgan B, Naidu B, Nonaka D, Peggs K, Pritchard C, Remmen H, Rowan A, Shah R, Smith E, Summers Y, Taylor M, Veeriah S, Waller D, Wilcox B, Wilcox M, Woolhouse I, McGranahan N, Swanton C. Tracking genomic cancer evolution for precision medicine: the lung TRACERx study. PLoS Biol. 2014;12(7):e1001906 Pemberton H, Anderton E, Patel H, Brookes S, Chandler H, Palermo R, Stock J, Rodriguez-Niedenführ M, Racek T, de Breed L, Stewart A, Matthews N, Peters G. Genome-wide co-localization of Polycomb orthologs and their effects on gene expression in human fibroblasts. Genome Biol. 2014;15(2):R23 Bioinformatics and Biostatistics (page 148) Aengus Stewart Primary Research Papers Andre F, Mardis E, Salm M, Soria JC, Siu LL, Swanton C. Prioritizing targets for precision cancer medicine. Ann Oncol. 2014;25(12):2295-303 Bullock MD, Mellone M, Pickard KM, Sayan AE, Mitter R, Primrose JN, Packham GK, Thomas G, Mirnezami AH. Molecular profiling of the invasive tumor microenvironment in a 3-dimensional model of colorectal cancer cells and ex vivo fibroblasts. J Vis Exp. 2014;(86). doi: 10.3791/51475 RESEARCH PUBLICATIONS 177 Chandler H, Patel H, Palermo R, Brookes S, Matthews N, Peters G. Role of polycomb group proteins in the DNA damage response – a reassessment. PLoS One. 2014;9(7):e102968 de Bruin E, McGranahan N, Mitter R, Salm M, Wedge DC, Yates L, Jamal-Hanjani M, Shafi S, Murugaesu N, Rowan AJ, Gronroos E, Muhammad MA, Horswell S, Gerlinger M, Varela I, Jones D, Marshall J, Voet T, Van Loo P, Rassl DM, Rintoul RC, Janes SM, Lee S, Forster M, Ahmed T, Lawrence D, Falzon M, Capitanio A, Harkins TT, Lee CC, Tom W, Teefe E, Chen SC, Begum S, Rabinowitz A, Phillimore B, Spencer-Dene B, Stamp G, Szallasi Z, Matthews N, Stewart A, Campbell P, Swanton C. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science. 2014;346(6206):251-6 Deddouche S, Goubau D, Rehwinkel J, Chakravarty P, Begum S, Maillard PV, Borg A, Matthews N, Feng Q, van Kuppeveld FJ, Reis e Sousa C. Identification of an LGP2-associated MDA5 agonist in picornavirusinfected cells. Elife. 2014;3:e01535 di Martino E, Kelly G, Roulson JA, Knowles MA. Alteration of cell-cell and cell-matrix adhesion in urothelial cells: an oncogenic mechanism for mutant FGFR3. Mol Cancer Res. 2014; doi: 10.1158/1541-7786. MCR-14-0022. Ellis RA, Horswell S, Ness T, Lumsdon J, Tooze SA, Kirkham N, Armstrong JL, Lovat PE. Prognostic impact of p62 expression in cutaneous malignant melanoma. J Invest Dermatol. 2014;134(5):1476-8 Esnault C, Stewart A, Gualdrini F, East P, Horswell S, Matthews N, Treisman R. Rho-actin signaling to the MRTF coactivators dominates the immediate transcriptional response to serum in fibroblasts. Genes Dev. 2014;28(9):943-58 Fisher R, Horswell S, Rowan A, Salm MP, de Bruin EC, Gulati S, McGranahan N, Stares M, Gerlinger M, Varela I, Crockford A, Favero F, Quidville V, André F, Navas C, Grönroos E, Nicol D, Hazell S, Hrouda D, O Brien T, Matthews N, Phillimore B, Begum S, Rabinowitz A, Biggs J, Bates PA, McDonald NQ, Stamp G, Spencer-Dene B, Hsieh JJ, Xu J, Pickering L, Gore M, Larkin J, Swanton C. Development of synchronous VHL syndrome tumors reveals contingencies and constraints to tumor evolution. Genome Biol. 2014;15(8):433 178 Scientific report 2014 LONDON RESEARCH INSTITUTE Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I, Fisher R, McGranahan N, Matthews N, Santos CR, Martinez P, Phillimore B, Begum S, Rabinowitz A, Spencer-Dene B, Gulati S, Bates PA, Stamp G, Pickering L, Gore M, Nicol DL, Hazell S, Futreal PA, Stewart A, Swanton C. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet. 2014;46(3):225-33 Hayashi R, Wainwright SM, Liddell SJ, Pinchin SM, Horswell S, Ish-Horowicz D. A genetic screen based on in vivo RNA imaging reveals centrosome-independent mechanisms for localizing gurken transcripts in Drosophila. G3 (Bethesda). 2014;4(4):749-60 Jamal-Hanjani M, Hackshaw A, Ngai Y, Shaw J, Dive C, Quezada S, Middleton G, de Bruin E, Le Quesne J, Shafi S, Falzon M, Horswell S, Blackhall F, Khan I , Janes S, Nicolson M, Lawrence D, Forster M, Fennell D, Lee SM, Lester J, Kerr K, Muller S, Iles N, Smith S, Murugaesu N, Mitter R, Salm M, Stuart A, Matthews N, Adams H, Ahmad T, Attanoos R, Bennett J, Birkbak NJ, Booton R, Brady G, Buchan K, Capitano A, Chetty M, Cobbold M, Crosbie P, Davies H, Denison A, Djearman M, Goldman J, Haswell T, Joseph L, Kornaszewska M, Krebs M, Langman G, MacKenzie M, Millar J, Morgan B, Naidu B, Nonaka D, Peggs K, Pritchard C, Remmen H, Rowan A, Shah R, Smith E, Summers Y, Taylor M, Veeriah S, Waller D, Wilcox B, Wilcox M, Woolhouse I, McGranahan N, Swanton C. Tracking genomic cancer evolution for precision medicine: The lung TRACERx study. PLoS Biol. 2014;12(7):e1001906 Lönnstedt IM, Caramia F, Li J, Fumagalli D, Salgado R, Rowan A, Salm M, Kanu N, Savas P, Horswell S, Gade S, Loibl S, Neven P, Sotiriou C, Swanton C, Loi S, Speed TP. Deciphering clonality in aneuploid tumors using SNP array and sequencing data. Genome Biol. 2014;15(9):470 Lopez-Serra L, Kelly G, Patel H, Stewart A, Uhlmann F. The Scc2-Scc4 complex acts in sister chromatid cohesion and transcriptional regulation by maintaining nucleosome-free regions. Nat Genet. 2014;46(10):1147-51 Luxton HJ, Barnouin K, Kelly G, Hanrahan S, Totty N, Neal DE, Whitaker HC. Regulation of the localisation and function of the oncogene LYRIC/AEG-1 by ubiquitination at K486 and K491. Mol Oncol. 2014;8(3):633-41 Pemberton H, Anderton E, Patel H, Brookes S, Chandler H, Palermo R, Stock J, Rodriguez-Niedenführ M, Racek T, de Breed L, Stewart A, Matthews N, Peters G. Genome-wide co-localization of Polycomb orthologs and their effects on gene expression in human fibroblasts. Genome Biol. 2014;15(2):R23 Roden DL, Sewell GW, Lobley A, Levine AP, Smith AM, Segal AW. ZODET: software for the identification, analysis and visualisation of outlier genes in microarray expression data. PLoS One. 2014;8;9(1):e81123 Saponaro M, Kantidakis T, Mitter R, Kelly GP, Heron M, Williams H, Söding J, Stewart A, Svejstrup JQ. RECQL5 controls transcript elongation and suppresses genome instability associated with transcription stress. Cell. 2014;157(5):1037-49 Sawers L, Ferguson MJ, Ihrig BR, Young HC, Chakravarty P, Wolf CR, Smith G. Glutathione S-transferase P1 (GSTP1) directly influences platinum drug chemosensitivity in ovarian tumour cell lines. Br J Cancer. 2014;111(6):1150-8 Wang Y, Chakravarty P, Ranes M, Kelly G, Brooks PJ, Neilan E, Stewart A, Schiavo G, Svejstrup JQ. Dysregulation of gene expression as a cause of Cockayne syndrome neurological disease. Proc Natl Acad Sci USA. 2014;111(40):14454-9 Biomolecular Modelling Research (page 149) Paul Bates Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I, Fisher R, McGranahan N, Matthews N, Santos CR, Martinez P, Phillimore B, Begum S, Rabinowitz A, Spencer-Dene B, Gulati S, Bates PA, Stamp G, Pickering L, Gore M, Nicol DL, Hazell S, Futreal PA, Stewart A, Swanton C. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet. 2014;46(3):225-33 Gulati S, Martinez P, Joshi T, Birkbak N, Santos CR, Rowlan AJ, Pickering L, Gore M, Larkin J, Szallasi Z, Bates PA, Swanton C, Gerlinger M. Systematic evaluation of the prognostic impact and intratumour heterogeneity of clear cell renal cell carcinoma biomarkers. Eur Urol. 2014;66(5):936-48 Torchala M, Bates PA. Predicting the structure of protein-protein complexes using the SwarmDock Web Server. Methods Mol Biol. 2014;1137:181-97 Electron Microscopy (page 151) Lucy Collinson Primary Research Papers Brownlow N, Pike T, Zicha D, Collinson L, Parker PJ. Mitotic catenation is monitored and resolved by a PKCε-regulated pathway. Nature Commun. 2014;5:5685 Burbage M, Keppler SJ, Gasparrini F, Martinez-Martín N, Gaya M, Feest C, Domart MC, Brakebusch C, Collinson L, Bruckbauer A, Batista FD. Cdc42 is a key regulator of B cell differentiation and is required for antiviral humoral immunity. J Exp Med. 2014;doi: 10.1084/jem.20141143 Primary Research Papers Bush M, Ghosh T, Sawicka M, Moal IH, Bates PA, Dixon R, Zhang X. The structural basis for enhancerdependent assembly and activation of the AAA transcriptional activator NorR. Mol Microbiol. 2014; doi: 10.1111/mmi.12844 Fisher R, Horswell S, Rowan A, Salm MP, de Bruin EC, Gulati S, McGranahan N, Stares M, Gerlinger M, Varela I, Crockford A, Favero F, Quidville V, André F, Navas C, Grönroos E, Nicol D, Hazell S, Hrouda D, O Brien T, Matthews N, Phillimore B, Begum S, Rabinowitz A, Biggs J, Bates PA, McDonald NQ, Stamp G, Spencer-Dene B, Hsieh JJ, Xu J, Pickering L, Gore M, Larkin J, Swanton C. Development of synchronous VHL syndrome tumors reveals contingencies and constraints to tumor evolution. Genome Biol. 2014;15(8):433 False coloured SEM of cell cultured lung cancer cells. RESEARCH PUBLICATIONS 179 Peddie CJ, Blight K, Wilson E, Melia C, Marrison J, Carzaniga R, Domart MC, O’Toole P, Larijani B, Collinson LM. Correlative and integrated light and electron microscopy of in-resin GFP fluorescence, used to localise diacylglycerol in mammalian cells. Ultramicroscopy. 2014;143:3-14 Russell MRG, West M, Peddie CJ, Collinson LM. 3D electron microscopy of cells across scales: electron tomography and serial block face scanning electron microscopy. Cell Imaging: Methods Express. Ed. Stephens, Scion Publishing. 2014. Simão D, Pinto C, Piersanti S, Weston A, Peddie CJ, Bastos AE, Licursi V, Schwarz SC, Collinson L, Salinas S, Serra M, Teixeira AP, Saggio I,Lima PA, Kremer EJ, Schiavo G, Brito C, Alves PM. Modeling human neural functionality in vitro: 3D culture for dopaminergic differentiation. Tissue Eng Part A. 2014;doi:10.1089/ten.tea.2014.0079 Experimental Histopathology (page 153) Terenzio M, Golding M, Russell MRG, Wicher K, Rosewell I, Spencer-Dene B, Ish-Horowicz D, Schiavo G. Bicaudal-D1 regulates the intracellular sorting and signalling of neurotrophin receptors. EMBO J. 2014;33(14):1582-98 Other Publications Carzaniga R, Domart MC, Duke E, Collinson LM. Correlative cryo-fluorescence and cryo-soft X-ray tomogarphy of adherent cells at European synchrotrons. Methods Cell Biol. 2014;124:151-78 Duke E, Dent K, Razi M, Collinson LM. Biological applications of cryo-soft X-ray tomography. J Microsc. 2014;255(2):65-7 Larijani B, Hamati F, Kundu A, Chung GC, Domart MC, Collinson LM, Poccia DL. Principle of duality in phospholipids: regulators of membrane morphology and dynamics. Biochem Soc Trans. 2014;42(5):1335-42 Patwardhan A, Ashton A, Brandt R, Butcher S, Carzaniga R, Chiu W, Collinson LM, Doux P, Duke E, Ellisman M, Franken E, Grünewald K, Heriche JK, Koster A, Kühlbrandt W, Lagerstedt I, Larabell C, Lawson CL, Saibil HR, Sanz-García E, Subramaniam S, Verkade P, Swedlow JR, and Kleywegt GJ. A 3D cellular context for the macromolecular world. Nat Struct Mol Biol. 2014;21(10):841-5 Peddie CJ, Liv N, Hoogenboom JP, Collinson LM. Integrated light and scanning electron microscopy of GFP-expressing cells. Methods Cell Biol. 2014;124:363-89 Peddie CJ, Collinson LM. Exploring the third dimension: volume electron microscopy comes of age. Micron. 2014;61:9-19 180 Scientific report 2014 LONDON RESEARCH INSTITUTE Gordon Stamp Primary Research Papers Acton SE, Farrugia AJ, Astarita JL, Mourão-Sá D, Jenkins RP, Nye E, Hooper S, van Blijswijk J, Rogers NC, Snelgrove KJ, Rosewell I, Moita LF, Stamp G, Turley SJ, Sahai E, Reis e Sousa C. Dendritic cells control fibroblastic reticular network tension and lymph node expansion. Nature. 2014;514(7523):498-502 de Bruin E, McGranahan N, Mitter R, Salm M, Wedge DC, Yates L, Jamal-Hanjani M, Shafi S, Murugaesu N, Rowan AJ, Gronroos E, Muhammad MA, Horswell S, Gerlinger M, Varela I, Jones D, Marshall J, Voet T, Van Loo P, Rassl DM, Rintoul RC, Janes SM, Lee S, Forster M, Ahmed T, Lawrence D, Falzon M, Capitanio A, Harkins TT, Lee CC, Tom W, Teefe E, Chen SC, Begum S, Rabinowitz A, Phillimore B, Spencer-Dene B, Stamp G, Szallasi Z, Matthews N, Stewart A, Campbell P, Swanton C. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science. 2014; 346(6206):251-6 Diefenbacher ME, Popov N, Blake SM, Schülein-Völk C, Nye E, Spencer-Dene B, Jaenicke LA, Eilers M, Behrens A. The deubiquitinase USP28 controls intestinal homeostasis and promotes colorectal cancer. J Clin Invest. 2014;124(8):3407-18 Fisher R, Horswell S, Rowan A, Salm MP, de Bruin EC, Gulati S, McGranahan N, Stares M, Gerlinger M, Varela I, Crockford A, Favero F, Quidville V, André F, Navas C, Grönroos E, Nicol D, Hazell S, Hrouda D, O Brien T, Matthews N, Phillimore B, Begum S, Rabinowitz A, Biggs J, Bates PA, McDonald NQ, Stamp G, Spencer-Dene B, Hsieh JJ, Xu J, Pickering L, Gore M, Larkin J, Swanton C. Development of synchronous VHL syndrome tumors reveals contingencies and constraints to tumor evolution. Genome Biol. 2014;15(8):433 Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I, Fisher R, McGranahan N, Matthews N, Santos CR, Martinez P, Phillimore B, Begum S, Rabinowitz A, Spencer-Dene B, Gulati S, Bates PA, Stamp G, Pickering L, Gore M, Nicol DL, Hazell S, Futreal PA, Stewart A, Swanton C. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet. 2014;46(3):225-33 Murillo MM, Zelenay S, Nye E, Castellano E, Lassailly F, Stamp G, Downward J. RAS interaction with PI3K p110α is required for tumor-induced angiogenesis. J Clin Invest. 2014;124(8):3601-11 Terenzio M, Golding M, Russell MR, Wicher KB, Rosewell I, Spencer-Dene B, Ish-Horowicz D, Schiavo G. Bicaudal-D1 regulates the intracellular sorting and signalling of neurotrophin receptors. EMBO J. 2014;33(14):1582-98 Veeriah S, Leboucher P, de Naurois J, Jethwa N, Nye E, Bunting T, Stone R, Stamp G, Calleja V, Jeffrey SS, Parker PJ, Larijani B. High throughput time-resolved-FRET reveals Akt/PKB activation as a poor prognostic marker in breast cancer. Cancer Res. 2014;74(18):4983-95 Zhuang Z, Frerich JM, Huntoon K, Yang C, Merrill MJ, Abdullaev Z, Pack SD, Shively SB, Stamp G, Lonser RR. Tumor derived vasculogenesis in von Hippel-Lindau disease-associated tumors. Sci Rep. 2014;4:4102 Fluorescence-Activated Cell Sorting (page 154) Derek Davies Primary Research Papers Filby A. “Mega” cytometry for a “mega” challenging cell type. Cytometry A. 2014;85(4):289-91 Sage EK, Kolluri KK, McNulty K, Lourenco Sda S, Kalber TL, Ordidge KL, Davies D, Gary Lee YC, Giangreco A, Janes SM. Systemic but not topical TRAIL-expressing mesenchymal stem cells reduce tumour growth in malignant mesothelioma. Thorax. 2014;69(7):638-47 Sarbajna S, Davies D, West SC. Roles of SLX1-SLX4, MUS81-EME1, and GEN1 in avoiding genome instability and mitotic catastrophe. Genes Dev. 2014;28(10):1124-36 Drosophila wing imaginal discs expressing expanded-GFP (green) and ubi-RFP (red) and stained with DAPI (blue). High Throughput Screening (page 155) Michael Howell Primary Research Papers de Bruin EC, Cowell C, Warne PH, Jiang M, Saunders RE, Melnick MA, Gettinger S, Walther Z, Wurtz A, Heynen GJ, Heideman DA, Gómez-Román J, García-Castaño A, Gong Y, Ladanyi M, Varmus H, Bernards R, Smit EF, Politi K, Downward J. Reduced NF1 expression confers resistance to EGFR inhibition in lung cancer. Cancer Discov. 2014;4(5):606-19 Endesfelder D, Burrell R, Kanu N, McGranahan N, Howell M, Parker PJ, Downward J, Swanton C, Kschischo M. Chromosomal instability selects gene copy number variants encoding core regulators of proliferation in ER+ breast cancer. Cancer Res. 2014;74(17):4853-63 Staples CJ, Myers KN, Beveridge RD, Patil AA, Howard AE, Barone G, Lee AJ, Swanton C, Howell M, Maslen S, Skehel JM, Boulton SJ, Collis SJ. Ccdc13 is a novel human centriolar satellite protein required for ciliogenesis and genome stability. J Cell Sci. 2014;127(Pt 13):2910-9 Sundaramoorthy S, Vázquez-Novelle MD, Lekomtsev S, Howell M, Petronczki M. Functional genomics identifies a requirement of pre-mRNA splicing factors for sister chromatid cohesion. EMBO J. 2014;33(22):2623-42 RESEARCH PUBLICATIONS 181 Luxton HJ, Barnouin K, Kelly G, Hanrahan S, Totty N, Neal DE, Whitaker HC. Regulation of the localisation and function of the oncogene LYRIC/AEG-1 by ubiquitination at K486 and K491. Mol Oncol. 2014;(3):633-41 Maertens GN, Cook NJ, Wang W, Hare S, Gupta SS, Öztop I, Lee K, Pye VE, Cosnefroy O, Snijders AP, KewalRamani VN, Fassati A, Engelman A, Cherepanov P. Structural basis for nuclear import of splicing factors by human Transportin 3. Proc Natl Acad Sci USA. 2014;111(7):2728-33 On KF, Beuron F, Frith D, Snijders AP, Morris EP, Diffley JF. Prereplicative complexes assembled in vitro support origin-dependent and independent DNA replication. EMBO J. 2014;33(6):605-20 Drosophila early larval central nervous system marge with GFP (green), c855a (red) and DAPI (blue). Light Microscopy (page 156) Daniel Zicha Primary Research Paper Brownlow N, Pike T, Zicha D, Collinson L, Parker PJ. Mitotic catenation is monitored and resolved by a PKCε-regulated pathway. Nature Commun. 2014;5:5685 Peptide Chemistry (page 157) Nicola O’Reilly Primary Research Paper Hořejší Z, Stach L, Flower TG, Joshi D, Flynn H, Skehel JM, O’Reilly NJ, Ogrodowicz RW, Smerdon SJ, Boulton SJ. Phosphorylation-dependent PIH1D1 interactions define substrate specificity of the R2TP cochaperone complex. Cell Rep. 2014;7(1):19-26 Protein Analysis and Proteomics (page 158) Bram Snijders Primary Research Papers Hořejší Z, Stach L, Flower TG, Joshi D, Flynn H, Skehel JM, O’Reilly NJ, Ogrodowicz RW, Smerdon SJ, Boulton SJ. Phosphorylation-dependent PIH1D1 interactions define substrate specificity of the R2TP cochaperone complex. Cell Rep. 2014;7(1):19-26 182 Scientific report 2014 LONDON RESEARCH INSTITUTE Pitcher DS, de Mattos-Shipley K, Wang Z, Tzortzis K, Goudevenou K, Flynn H, Bohn G, Rahemtulla A, Roberts I, Snijders AP, Karadimitris A, Kleijnen MF. Nuclear proteasomes carry a constitutive posttranslational modification which derails SDSPAGE (but not CTAB-PAGE). Biochim Biophys Acta. 2014;1844(12):2222-8 Plaza-Menacho I, Barnouin K, Goodman K, Martínez-Torres RJ, Borg A, Murray-Rust J, Mouilleron S, Knowles P, McDonald NQ. Oncogenic RET kinase domain mutations perturb the autophosphorylation trajectory by enhancing substrate presentation in trans. Mol Cell. 2014;53(5):738-51 Ribeiro P, Holder M, Frith D, Snijders AP, Tapon N. Crumbs promotes expanded recognition and degradation by the SCF(Slimb/β-TrCP) ubiquitin ligase. Proc Natl Acad Sci USA. 2014;111(19):E1980-9 Swarts DC, Jore MM, Westra ER, Zhu Y, Janssen JH, Snijders AP, Wang Y, Patel DJ, Berenguer J, Brouns SJ, van der Oost J. DNA-guided DNA interference by a prokaryotic Argonaute. Nature. 2014;507(7491):258-61 Zhang T, Cronshaw J, Kanu N, Snijders AP, Behrens A. UBR5-mediated ubiquitination of ATMIN is required for ionizing radiation-induced ATM signaling and function. Proc Natl Acad Sci USA. 2014;111(33):12091-6 Protein Purification (page 159) Svend Kjær Primary Research Papers Ben-Addi A, Mambole-Dema A, Brender C, Martin SR, Janzen J, Kjær S, Smerdon SJ, Ley SC. IκB kinase-induced interaction of TPL-2 kinase with 14-3-3 is essential for Toll-like receptor activation of ERK-1 and -2 MAP kinases. Proc Natl Acad Sci USA. 2014;111(23):E2394-403 Deddouche S, Goubau D, Rehwinkel J, Chakravarty P, Begum S, Maillard PV, Borg A, Matthews N, Feng Q, van Kuppeveld FJ, Reis e Sousa C. Identification of an LGP2-associated MDA5 agonist in picornavirusinfected cells. Elife. 2014;3:e01535 Goodman KM, Kjær S, Beuron F, Knowles PP, Nawrotek A, Burns EM, Purkiss AG, George R, Santoro M, Morris EP, McDonald NQ. RET recognition of GDNF-GFRα1 ligand by a composite binding site promotes membrane-proximal selfassociation. Cell Rep. 2014;8(6):1894-904 Terenzio M, Golding M, Russell MR, Wicher KB, Rosewell I, Spencer-Dene B, Ish-Horowicz D, Schiavo G. Bicaudal-D1 regulates the intracellular sorting and signalling of neurotrophin receptors. EMBO J. 2014;33(14):1582-98 In vivo imaging (page 161) Francois Lassailly Primary Research Papers Griessinger E, Anjos-Afonso F, Pizzitola I, Rouault-Pierre K, Vargaftig J, Taussig D, Gribben J, Lassailly F, Bonnet D. A niche-like culture system allowing the maintenance of primary human acute myeloid leukemia-initiating cells: a new tool to decipher their chemoresistance and self-renewal mechanisms. Stem Cells Transl Med. 2014;(4):520-9 Murillo MM, Zelenay S, Nye E, Castellano E, Lassailly F, Stamp G, Downward J. RAS interaction with PI3K p110α is required for tumor-induced angiogenesis. J Clin Invest. 2014;124(8):3601-11 Plaza-Menacho I, Barnouin K, Goodman K, Martínez-Torres RJ, Borg A, Murray-Rust J, Mouilleron S, Knowles P, McDonald NQ. Oncogenic RET kinase domain mutations perturb the autophosphorylation trajectory by enhancing substrate presentation in trans. Mol Cell. 2014;53(5):738-51 Pizzitola I, Anjos-Afonso F, Rouault-Pierre K, Lassailly F, Tettamanti S, Spinelli O, Biondi A, Biagi E, Bonnet D. Chimeric antigen receptors against CD33/ CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo. Leukemia. 2014;28(8):1596-605 Simon AC, Zhou JC, Perera RL, van Deursen F, Evrin C, Ivanova ME, Kilkenny ML, Renault L, Kjær S, Matak-Vinković D, Labib K, Costa A, Pellegrini L. A Ctf4 trimer couples the CMG helicase to DNA polymerase α in the eukaryotic replisome. Nature. 2014;510(7504):293-7 Protein Structure (page 162) Transgenics (page 160) Ian Rosewell Stephane Mouilleron Primary Research Paper Plaza-Menacho I, Barnouin K, Goodman K, Martínez-Torres RJ, Borg A, Murray-Rust J, Mouilleron S, Knowles P, McDonald NQ. Oncogenic RET kinase domain mutations perturb the autophosphorylation trajectory by enhancing substrate presentation in trans. Mol Cell. 2014;53(5):738-51 Primary Research Papers Acton SE, Farrugia AJ, Astarita JL, Mourão-Sá D, Jenkins RP, Nye E, Hooper S, van Blijswijk J, Rogers NC, Snelgrove KJ, Rosewell I, Moita LF, Stamp G, Turley SJ, Sahai E, Reis e Sousa C. Dendritic cells control fibroblastic reticular network tension and lymph node expansion. Nature. 2014;514(7523):498-502 RESEARCH PUBLICATIONS 183 Other Research Papers Vincenzo Costanzo Bellelli R, Castellone MD, Guida T, Limongello R, Dathan NA, Merolla F, Cirafici AM, Affuso A, Masai H, Costanzo V, Grieco D, Fusco A, Santoro M, Carlomagno F. NCOA4 transcriptional coactivator inhibits activation of DNA replication origins. Mol Cell. 2014;55(1):123-37. Errico A, Aze A, Costanzo V. Mta2 promotes Tipindependent maintenance of replication fork integrity. Cell Cycle. 2014;13(13):2120-8 Nancy Hogg De Filippo K, Neill DR, Mathies M, Bangert M, McNeill E, Kadioglu A, Hogg N. A new protective role for S100A9 in regulation of neutrophil recruitment during invasive pneumococcal pneumonia. FASEB J. 2014;28(8):3600-8 McNeill E, Hogg N. S100A9 has a protective role in inflammation-induced skin carcinogenesis. Int J Cancer. 2014;135(4):798-808 Wang Y, Fang C, Gao H, Bilodeau ML, Zhang Z, Croce K, Liu S, Morooka T, Sakuma M, Nakajima K, Yoneda S, Shi C, Zidar D, Andre P, Stephens G, Silverstein RL, Hogg N, Schmaier AH, Simon DI. Platelet-derived S100 family member myeloid-related protein-14 regulates thrombosis. J Clin Invest. 2014;124(5):2160-71 David Ish-Horowicz Hayashi R, Wainwright SM, Liddell SJ, Pinchin SM, Horswell S, Ish-Horowicz D. A genetic screen based on in vivo RNA imaging reveals centrosome-independent mechanisms for localizing gurken transcripts in Drosophila. G3 (Bethesda). 2014;4(4):749-60 Soza-Ried C, Öztürk E, Ish-Horowicz D, Lewis J. Pulses of Notch activation synchronise oscillating somite cells and entrain the zebrafish segmentation clock. Development. 2014;141(8):1780-8 Terenzio M, Golding M, Russell MR, Wicher KB, Rosewell I, Spencer-Dene B, Ish-Horowicz D, Schiavo G. Bicaudal-D1 regulates the intracellular sorting and signalling of neurotrophin receptors. EMBO J. 2014;33(14):1582-98 184 Scientific report 2014 LONDON RESEARCH INSTITUTE Banafshe Larijani Byrne RD, Veeriah S, Applebee CJ, Larijani B. Conservation of proteo-lipid nuclear membrane fusion machinery during early embryogenesis. Nucleus. 2014;5(5):441-8 Calleja V, Laguerre M, de Las Heras-Martinez G, Parker PJ, Requejo-Isidro J, Larijani B. Acute regulation of PDK1 by a complex interplay of molecular switches. Biochem Soc Trans. 2014;42(5):1435-40 Larijani B, Hamati F, Kundu A, Chung GC, Domart MC, Collinson L, Poccia DL. Principle of duality in phospholipids: regulators of membrane morphology and dynamics. Biochem Soc Trans. 2014;42(5):1335-42 Linch M, Riou P, Claus J, Cameron AJ, de Naurois J, Larijani B, Ng T, McDonald NQ, Parker PJ. Functional implications of assigned, assumed and assembled PKC structures. Biochem Soc Trans. 2014;42(1):35-41 Peddie CJ, Blight K, Wilson E, Melia C, Marrison J, Carzaniga R, Domart MC, O’Toole P, Larijani B, Collinson LM. Correlative and integrated light and electron microscopy of in-resin GFP fluorescence, used to localise diacylglycerol in mammalian cells. Ultramicroscopy. 2014;143:3-14 Veeriah S, Leboucher P, de Naurois J, Jethwa N, Nye E, Bunting T, Stone R, Stamp G, Calleja V, Jeffrey SS, Parker PJ, Larijani B. High-throughput time-resolved FRET reveals Akt/PKB activation as a poor prognostic marker in breast cancer. Cancer Res. 2014;74(18):4983-95 Zhang H, Cohen AL, Krishnakumar S, Wapnir IL, Veeriah S, Deng G, Coram MA, Piskun CM, Longacre TA, Herrler M, Frimannsson DO, Telli ML, Dirbas FM, Matin AC, Dairkee SH, Larijani B, Glinsky GV, Bild AH, Jeffrey SS. Patient-derived xenografts of triple-negative breast cancer reproduce molecular features of patient tumors and respond to mTOR inhibition. Breast Cancer Res. 2014;16(2):R36 Julian Lewis Soza-Ried C, Öztürk E, Ish-Horowicz D, Lewis J. Pulses of Notch activation synchronise oscillating somite cells and entrain the zebrafish segmentation clock. Development. 2014;141(8):1780-8 Taija Makinen Lutter S, Makinen T. Regulation of lymphatic vasculature by extracellular matrix. Adv Anat Embryol Cell Biol. 2014;214:55-65 Gordon Peters Chandler H, Patel H, Palermo R, Brookes S, Matthews N, Peters G. Role of polycomb group proteins in the DNA damage response - a reassessment. PLoS One. 2014;9(7):e102968 Mikawa T, Maruyama T, Okamoto K, Nakagama H, Lleonart ME, Tsusaka T, Hori K, Murakami I, Izumi T, Takaori-Kondo A, Yokode M, Peters G, Beach D, Kondoh H. Senescence-inducing stress promotes proteolysis of phosphoglycerate mutase via ubiquitin ligase Mdm2. J Cell Biol. 2014;204(5):729-45 O’Loghlen A, Martin N, Krusche B, Pemberton H, Alonso MM, Chandler H, Brookes S, Parrinello S, Peters G, Gil J. The nuclear receptor NR2E1/TLX controls senescence. Oncogene. 2014;doi: 10.1038/ onc.2014.335 Pemberton H, Anderton E, Patel H, Brookes S, Chandler H, Palermo R, Stock J, Rodriguez-Niedenführ M, Racek T, de Breed L, Stewart A, Matthews N, Peters G. Genome-wide co-localization of Polycomb orthologs and their effects on gene expression in human fibroblasts. Genome Biol. 2014;15(2):R23 Giampietro Schiavo Bercsenyi K, Schmieg N, Bryson JB, Wallace M, Caccin P, Golding M, Zanotti G, Greensmith L, Nischt R, Schiavo G. Tetanus toxin entry. Nidogens are therapeutic targets for the prevention of tetanus. Science. 2014;346(6213):1118-23 Hislop JN, Islam TA, Eleftheriadou I, Carpentier DC, Trabalza A, Parkinson M, Schiavo G, Mazarakis ND. Rabies virus envelope glycoprotein targets lentiviral vectors to the axonal retrograde pathway in motor neurons. J Biol Chem. 2014; 289(23):16148-63 Schmieg N, Menendez G, Schiavo G, Terenzio M. Signalling endosomes in axonal transport: travel updates on the molecular highway. Semin Cell Dev Biol. 2014;27:32-43 Simão D, Pinto C, Piersanti S, Weston A, Peddie CJ, Bastos AE, Licursi V, Schwarz SC, Collinson LM, Salinas S, Serra M, Teixeira AP, Saggio I, Lima PA, Kremer EJ, Schiavo G, Brito C, Alves PM. Modeling human neural functionality in vitro: three-dimensional culture for dopaminergic differentiation. Tissue Eng Part A. 2014;doi: 10.1089/ten.TEA.2014.0079 Terenzio M, Golding M, Russell MR, Wicher KB, Rosewell I, Spencer-Dene B, Ish-Horowicz D, Schiavo G. Bicaudal-D1 regulates the intracellular sorting and signalling of neurotrophin receptors. EMBO J. 2014;33(14):1582-98 Terenzio M, Golding M, Schiavo G. siRNA screen of ES cell-derived motor neurons identifies novel regulators of tetanus toxin and neurotrophin receptor trafficking. Front Cell Neuroscience. 2014;8:140 Wang Y, Chakravarty P, Ranes M, Kelly G, Brooks PJ, Neilan E, Stewart A, Schiavo G, Svejstrup JQ. Dysregulation of gene expression as a cause of Cockayne syndrome neurological disease. Proc Natl Acad Sci U S A. 2014;111(40):14454-9 Almut Schulze Bensaad K, Favaro E, Lewis CA, Peck B, Lord S, Collins JM, Pinnick KE, Wigfield S, Buffa FM, Li JL, Zhang Q, Wakelam M, Karpe F, Schulze A, Harris AL. Fatty acid uptake and lipid storage induced by HIF-1α contribute to cell growth and survival after hypoxiareoxygenation. Cell Rep. 2014;9(1):349-65 Peck B, Schulze A. Cholesteryl esters: fueling the fury of prostate cancer. Cell Metab. 2014;19(3):350-2 Yizhak K, Devedec SE, Rogkoti VM, Baenke F, de Boer VC, Frezza C, Schulze A, van de Water B, Ruppin E. A computational study of the Warburg effect identifies metabolic targets inhibiting cancer migration. Mol Syst Biol. 2014;10:744 Helen Walden Hodson C, Purkiss A, Miles JA, Walden H. Structure of the human FANCL RING-Ube2T complex reveals determinants of cognate E3-E2 selection. Structure. 2014;22(2):337-44 RESEARCH PUBLICATIONS 185 THESES Rudi Agius Biomolecular Modelling Understanding the Stability of Protein-Protein Complexes Susan Ahrens Immunobiology Extracellular actin in innate immunity Graham Bell Alessandra Audia Haematopoietic Stem Cell In vitro model to study the role of Notch pathway in the interaction between Haematopoietic Stem Cells (HSCs) and their microenvironment. Hella Baumann Microtubule Cytoskeleton In vitro reconstruction of confined microtubule cytoskeleton self-organisation Emily Burns Graham Bell Epithelial Biology The roles and regulation of the Drosophila Lgl tumour suppressor in cell division Kinga Bercsenyi Molecular Neuropathobiology Nidogens are essential for the entry of tetanus toxins into motor neurons Mariana Campos Emily Burns Structural Biology Structural and functional aspects of RET receptor tyrosine kinase maturation, signalling and chemical inhibition Mariana Campos Epithelial Biology Bul and Kul are novel components of the DachsousFat planar polarity system Heike Miess Rahul Thadani Gary Hong Chun Chung Cell Biophysics The role of fusogenic vesicles in the regulation of nuclear envelope assembly Alex Fennell Telomere Biology Centromeres and telomeres display unanticipated and interchangeable roles in promoting nuclear division in fission yeast Francesco Gualdrini Signalling and Transcription Mechanisms of transcriptional regulation by SRF co-factors Heike Miess Gene Expression Analysis Identification of metabolic genes essential for proliferation of clear cell Renal Cell Carcinoma (ccRCC) cells Richard Panayiotou Signalling and Transcription Phosphorylation mediated regulation of MRTF-A Matt Peacock Mammalian DNA Repair UVA Photosensitisers, Protein Oxidation and DNA Repair Thibaud Perriches Macromolecular Structure and Function The CBF3 complex structure and function during point centromere establishment Ngang Heok Tang Cell Regulation A study on Kinetochore-Spindle Microtubule Attachment: Ndc80 and TACC-TOG/MAPs Martin Taylor DNA Damage Response Mechanism of action of Rad51 paralogs Jeroen Claus Protein Phosphorylation The pseudokinase HER3: Structure/function relationships and inhibitor-induced signalling Rahul Thadani Chromosome Segregation The condensin ATPase: towards a mechanistic view of chromosome condensation Tom Deegan Chromosome Replication Investigating the Mechanism of Activation of the Mcm2-7 Replicative Helicase Martin Wallace Molecular Neuropathobiology Phosphorylation of Rab7 at serine 72 and its role in the regulation of the late endocytic pathway Stefanie Derzsi Tumour Cell Biology Investigation of heterotypic interactions between cancer cells and cancer-associated fibroblasts Tianyi Zhang Mammalian Genetics The role of ATMIN in regulating ATM signalling Hannah Dooley Secretory Pathways Investigation of the role of WIPI2 in autophagosome formation: Functional characterisation of the WIPI2-Atg16L1 interaction 186 Christian Duellberg Microtubule Cytoskeleton Mechanism and control of microtubule dynamic instability probed by in vitro reconstitutions and microfluidics approaches Scientific report 2014 LONDON RESEARCH INSTITUTE Yanxiang Zhou Apoptosis and Proliferation Control Developmental functions of Drosophilia ASPP and RASSF8 INSTITUTE INFORMATION Administration Academic Programme Seminars and Conferences External Funding Institute Management INSTITUTE INFORMATION 187 ADMINISTRATION Director of Operations Ava Yeo PhD Administration Team Charis Ashton David Bacon Andrew Brown Lucy Davinson Sabina Ebbols Nicola Hawkes PhD Rachel Hawkes* David Hudson PhD Sophie Kidane* Sally Leevers PhD Ivana Petrovska PhD Kim Rowan* Emma Rainbow* Michelle Trowsdale* Tom Wallace* Electronics Department Peter Nash Paul Boyles David Martin Finance Team Claire MacIver* Catherine Cunningham PhD* Betty Chen* Curtis Okonkwo* Azizur Rahman Fly Facility Terrance Gilbank Shaun Malloney Paul Moulder Francis Crick Transition Team Caroline Doran Cate Dixon Georgie Nightingall Fay Oetegenn Fatima Sulaiman PhD Karen Vincent Grants Team Holly Elphinstone* Justin Wilson An Tyrrell* PhD Health and Safety Tim Budd PhD Mandy Marshall John Richmond Nicholas Tidman HR Team Emma Collins Heather Campbell Frederick Giffard* Kayleigh Smith* 188 LRI administration The LRI Administration team provides the Director with the administrative infrastructure and support to ensure the smooth running of the institute. The team led by the LRI Director of Operations is responsible for the academic infrastructure through the administration of the academic committees for students and postdocs, management of LRI Technology Core Facilities, IT, finance, co-ordinating institute-wide initiatives and providing general administrative support to the Research Laboratories. The LRI also manages the Cancer Research UK Library. Graduate student administration This year saw the launch of the new Crick PhD Programme, with 27 students, selected from 1060 candidates from all over the world, starting their PhDs based at the LRI. Together with students from the NIMR, they will form the first cohort of Crick PhD students, following a newly devised PhD programme that will provide continual training, development and support throughout their four-year programme. Specialised training programme The LRI Software Tutor, David Bacon, runs specially designed courses to enable students to use commercially available software efficiently and effectively in the course of their research. Classes are held regularly at Clare Hall and Lincoln’s Inn Fields and include Adobe Illustrator, PowerPoint, Endnote, Word etc. Targeted support is given to students as they write up their thesis in their final year. All students attend an internal seminar where the LRI’s image processing guidelines are outlined. The seminar introduces the concept of image integrity, and highlights the importance of correct processing techniques for digital editing. Postdoctoral fellows administration Postdocs at the LRI continue to be supported by the LRI Postdoctoral Training Programme, developed collaboratively by postdocs, group leaders and the Academic Director. The programme supports the postdocs throughout their time at the LRI, from their induction and introduction to all the facilities and activities available, to their annual career development reviews and discussions as they prepare to move onto the next stage of their career. Postdocs attend Postdoc Consultative Meetings (PDCMs) several times a year enabling communication between the Scientific report 2014 LONDON RESEARCH INSTITUTE institute and postdocs. Postdocs are also represented on committees such as the Technology Core Facility User Group ensuring that they contribute to institute activities. For the third year running, LRI postdocs joined with postdocs from the National Institute for Medical Research (NIMR) to organise the Francis Crick Institute Postdoc Retreat. Postdocs from the LRI, NIMR, King’s College London, Imperial College London and University College London attended the retreat, which was held at the British Library. Administrative support for group leaders Group leaders receive comprehensive secretarial and administrative support from research administrators. The procedures and processes are continually assessed and improvements implemented to ensure a smooth and efficient service is provided. Laboratory management services The Laboratory Services and Support team works closely with the research laboratories and Core Technology Facilities, playing a role in support of their scientific activities. As well as looking after refurbishment projects, communal equipment and the Containment Facility, the team also includes the Electronics Department, Stores Team and the Fly Facility Service. The upkeep and replacement of communal equipment is an important part of our work, ensuring scientists are provided with the appropriate facilities. The team liaises closely with both the Health and Safety and Property Services Departments, ensuring co-operation across all areas at the LRI. This is particularly important when setting up new research groups; this year five laboratories have been refurbished for incoming group leaders. We help with the running of laboratories, provide advice about equipment repairs and maintenance as well as other technical support. We also support the Purchase to Pay (P2P) system and are a first point of contact for questions Scientific Officers (SOs) may have regarding the placing of orders. In addition, this year saw the involvement of many SOs with the Crick Quadrant Working Groups, an exercise undertaken to help plan the new laboratory space at the Crick with our colleagues from the NIMR. In order to prepare for sharing more communal space at the Crick, the team has also instigated an institute-wide laboratory clear out to try and dispose of any unwanted reagents, equipment and paper before the move. IT Surinder Dio Claire Brewer Marion Edwards* Andy Foster Jacki Goldman Simon Grierson Ellen Gyapong Mat Hillyard Andrew Jordan Chris Manser Santosh Nittala Wing Poon Phil Spratt Mark Tomlinson* Lab Aides Susan Hill Ian Morris Chris Coomber Kim Crane Mark Dalton Gareth Dineen Carol Du Preez Brenda Foran Annette Pereira Lucia Scalco Jill Sheehan Patricia Smith-Carington Susan Smith-Carington Michelle Wood Laboratory Management Team Nigel Peat Mark Johnson Hans Nicolai Fiona Johnson Elizabeth Li Kathryn Snelgrove Reception, Clare Hall Susan Hughes Janet Almond Research Administrators Jessica Adams* Katherine Ames Aleksandra Banasiak Helen Batley Nicola Howes Sophie Kidane* Sophie Kontakkis Jackie Martinez Aileen Nelson Mary Nicolaou Anastasia Photiou Stores David Hawkes Phillip O’Brien Andy Bendon Anthony Crane Lee Goldstein Ronald Main Gary Martin Paul Riley Michael Wilkins Washroom Jayson Webb Wayne Bushell Yaw Sarpong Andy Perry Paul Chambers *= Part year. Health and Safety The Health and Safety team at the LRI provides advice, training and support in all aspects of welfare and safety throughout the institute, whether for scientific, administrative or maintenance work. This year the team have been working with Property Services to implement changes to the way waste is recycled across the institute and in the processing of laboratory waste. This has led to a saving of £30,000 over the year. The Health and Safety team have also been working hard in planning for the safe decommissioning of 44 LIF and Clare Hall prior to the move to the Crick. IT The LRI IT team have been involved with: • An expansion of the archive solution to allow for a multi-tiering storage solution, that automatically migrates files older than a specified date to be migrated to slower storage • The implementation of a soft quota’s system to allow for monitoring and reporting of storage both at user and group level • Continued expansion of the network • Increased activity in Crick-related work • Piloting of the EduRoam wifi service • Improvements on the security of network services with the deployment of a new system for hosting externally facing web sites • Improvements to the overall service delivery of IT within the LRI • Improved disaster recovery for the virtual infrastructure with replication across LIF and CH of essential services Outreach 2014 has been a busy year for the LRI’s outreach programme, which aims to engage the public, especially school children, in our research. We encourage all staff to contribute to these activities. • Code Breaking: Reading the Genetic Code with Raspberry Pi What is the genome, how can we ‘read’ it, and why do ‘mistakes’ in the genome cause cancer? This autumn we explored these questions with 12-15 year-old students from the Maria Fidelis School in Camden to highlight the role of computers in cancer research. In the third iteration of this programme, LRI scientists helped students write programs to ‘decode’ DNA in search of potential cancer-causing mutations. By providing course materials adapted to inexpensive technologies, such as the Raspberry Pi computer and open source software, we are doing our part to help schools bring programming into the classroom. • ‘Exploring the Microworld’ Workshop In this CREST Accredited project, LRI scientists led short activities for 12-17 year-old students aimed at demystifying the microscope. Students built £10 microscopes, learned about using inexpensive microscopes for field diagnosis, and got the chance to use various microscopes to examine fruit flies, zebrafish and nematode worms to learn how these model organisms help us answer big questions in cancer biology. The workshop was featured at the Royal College of Pathologists, Guildhall and a local Camden school. • Science Museum Lates LRI scientists played a big part at this Crick-led event, which attracted approximately 7000 visitors. Groups from the LRI discussed the power of electron microscopy, the role of the immune system in cancer and designer drugs. • Work Experience Finally, we continued the LRI Schools Work Experience Scheme at both Clare Hall and Lincoln’s Inn Fields, allowing students to spend time in both Core Technology Facilities and research laboratories. Additionally two Nuffield Science Bursary places were offered to 6th form students in the Macromolecular Structure and Function Laboratory and Light Microscopy Facility. Crick Transition During 2013-14 we have continued our work to prepare for the LRI’s transition to the Francis Crick Institute. As the transfer approaches we have undertaken detailed planning activity to ensure all elements of the transfer are understood and the required work to ensure a smooth transition is underway. The LRI Transition Core Team, made up of the Workstream Leads and the Transition Project team, meet regularly to review progress and discuss key issues. The Core Team reports monthly to the LRI Transition Steering Group. LRI transition is governed by the CRUK Crick Programme Board chaired by CRUK’s Chief Executive, Harpal Kumar. The priority for the next year will be to execute the legal transfer of the LRI to the Francis Crick Institute and to continue working on the preparation for the physical move and building closedown activity of 44LIF and Clare Hall. Successful transition will require strong partnership working between the LRI, CRUK and the Crick. ADMINISTRATION 189 ACADEMIC PROGRAMME At any one time the LRI has approximately 100 graduate students and nearly 180 postdoctoral fellows carrying out their research and participating in scientific training programmes designed to develop their skills and lay the groundwork for their future careers. The students and postdocs form the core of the institute’s scientific community, carrying out high quality research, as evidenced in the research highlights and publications sections of this report. Graduate Students Our graduate students are recruited via a highly competitive annual selection process designed to identify outstanding students who are passionate about carrying out research leading to a PhD. In 2014 we recruited the first cohort of students that will follow the new Crick PhD programme. 27 Crick-LRI students were selected to join the programme in September 2014 alongside 20 Crick-NIMR PhD students. Nine of these students are jointly funded by the Crick and either Imperial College London (Imperial) or King’s College London (King’s). These joint students, with an institute supervisor and Imperial or King’s supervisor, will carry out collaborative and interdisciplinary research projects spanning both supervisor’s research groups. The students contribute to the international flavour of our institute, with 30% of the 2014 intake coming from the rest of Europe and 20% from outside the EU. A key feature of both the LRI PhD programme and the new Crick PhD programme is the cohesion of the student community, which provides a strong peer-to-peer support network and can lead to highly fruitful scientific collaborations. This network is fostered from the very beginning of the PhD programme when new students attend a 4-day induction. The September 2014 Crick PhD student induction programme included sessions to introduce students to the Crick and its founding institutes and university partners, to prepare them for undertaking their PhD, and to help them to get to know each other. Topics covered via talks and interactive sessions included organising yourself and your research, keeping on top of the literature, effective experiment design, and introductions to the Core Technology Facilities. The week ended with a student-organised symposium named after Francis Crick’s book on scientific discovery, ‘What Mad Pursuit’. Two students from each institute, in each year of their PhD, gave short presentations about their research project and provided some advice for new students embarking on their PhDs. 190 Scientific report 2014 LONDON RESEARCH INSTITUTE The new Crick-LRI students are registered for their PhDs with one of the Crick partner universities, Imperial, King’s or University College London (UCL). The universities provide an important source of additional training, facilities and support, which complements those provided within the institute. In addition to guidance from their primary supervisor and other research group members, students receive scientific advice from their thesis committee, which they meet with at key ‘progression points’ during their PhD. Thesis committees are made up of three senior researchers including one faculty member from the university that the student has registered with for their PhD. An exciting new activity in 2014 was the development and introduction of the new ‘Crick Grad Log’ – an online system to record and monitor students’ progress and training. Students submit their research reports, upload talks, log training and receive feedback from their supervisors and thesis committee via the Log. Further support for PhD students throughout their time at the institute is provided by the LRI student admin team: Sally Leevers, LRI Academic Director, Andrew Brown, LRI Research Manager for Graduate Studies and Sabina Ebbols, LRI Academic HEI Liaison Manager, and by their NIMR and Crick colleagues. While the 2014 PhD students are following the new Crick PhD Programme, the LRI PhD Programme continues for the institute’s second, third and fourth year students. For example, the second year students participated in CRUK’s course on communicating science to scientific and non-scientific audiences prior to giving their second year seminars, while the third year students attended the National Cancer Research Institute annual conference, where many of them gave poster presentations on their research. Other student activities included attending the International PhD Student Cancer Conference at the DKFZ in Heidelberg, the student-organised summer event and Christmas lecture as well as Intake of Crick 2014 PhD Students contributing to Cancer Research UK fundraising by giving talks to supporters and other activities. Postdoctoral Fellows The LRI postdocs are a vibrant and diverse community – about 20% are British, 60% are from other European countries, and 20% from further afield. Approximately half are funded by Cancer Research UK postdoctoral fellowships, some are supported by external grants awarded to their group leaders, and many of them secure their own personal fellowships from external agencies such as the European Molecular Biology Organisation, the Human Frontiers Science Programme and Marie Skłodowska-Curie Actions. LRI Postdocs can access a training programme that was developed collaboratively by postdocs, group leaders and the Academic Director. The programme starts with an induction to orient postdocs and make them aware of the facilities and activities available, and to introduce them to the Core Technology Facilities that will facilitate their research. Soon after starting, postdocs submit a project proposal and throughout their programme, they have annual career development reviews with their group leaders. These reviews focus on the postdocs’ science, taking a broader perspective than normal day-to-day conversations, and provide structure and focus for postdocs’ scientific and career development to aid them in their future career. Postdocs also present seminars about 18 months and 3 years after starting at the LRI. Their second seminar is followed by a Postdoc Career Development Discussion. Postdocs select and invite several group leaders to join them for these discussions about their scientific progress, publication strategy and future career plans. About quarter of the postdocs leaving the LRI take up independent positions to establish their own research groups, about half go to a second postdoctoral position, and others move into areas such as pharma, biotech, science communication, publishing, and clinical trails. Postdocs are invited to attend Postdoc Consultative Meetings (PDCMs), which provide an important opportunity for postdocs to discuss issues that affect them within the LRI and to communicate with LRI Administration and Management. In addition, postdoc representatives attend the LRI Scientific Staff Meeting and the Fellowships Committee to feedback to the PDCM what was discussed at these meetings. The 2014 Crick Postdoc Retreat, organised by a committee of postdocs from the LRI and NIMR, was held in June at the British Library, next to the Francis Crick Institute site (see the Seminars and Conferences section of this report). More than 150 postdocs from LRI, NIMR and the Crick’s university partners attended the stimulating and interactive day with a theme of ‘Inspiring Science’. Speakers from academia and pharma gave presentations on their science and careers, and there was a discussion session on open access publishing. Other highlights included a postdoc networking nexus, and an orienteering trail from the British Library to the evening venue, which provided an opportunity to explore the area around the Crick. Joint Crick 2014 Post Doc retreat at The British Library ACADEMIC PROGRAMME 191 SEMINARS AND CONFERENCES The London Research Institute hosts a Special Seminar Series to invite external speakers from around the world to present their work, covering a broad spectrum of cutting-edge topics within the areas of genome integrity, signal transduction, structural biology, developmental biology and immunology. There are also a number of Special Interest Groups within the different areas of interest within the institute, which are open to external visitors to attend, providing a unique networking opportunity to encourage collaboration within London and the surrounding area. A selection from this year’s programme is listed below: Special Seminars Judith Campisi, The Buck Institute, USA Cancer and aging: Rival demons? Aaron Straight, Stanford University, USA Turning on the genome: mechanisms of zygotic genome activation Advanced Bioimaging Kishan Dholakia, St Andrews University, UK Shaping the future of imaging for biomedicine Rainer Heintzmann, University of Jena, Germany Structured Illumination and the Analysis of Single Molecules in Cells Computational and Mathematical Biology Franca Fraternali, Kings College London, UK A Multiscale view of Protein-Protein Interactions Trevor Graham, Barts Cancer Institute, UK Quantifying the evolution of human intestinal stem cells Developmental Biology Immunology Shane Crotty, La Jolla Institute, USA T follicular helper cell (Tfh) differentiation and genetics Antal Rot, University of Birmingham, UK New pathophysiological roles of atypical chemokine receptors. Molecular Medicines & Therapeutics Sandra Misale, Istituto di Candiolo, Turin, Italy Vertical suppression of the EGFR pathway to overcome acquired resistance to anti-EGFR therapy in colorectal cancer Andy West, GlaxoSmithKline, UK Imaging drug distribution using mass spectrometry – from tissues to cells Signalling Katsuhiko Shirahige, University of Tokyo, Japan Transcriptional regulation by Cohesin loader NIPBL-Mau2 complex Oriol Casanovas, Catalan Institute of Oncology, Barcelona, Spain Anti-angiogenic Therapies: Learning from their Limitations Alison Woollard, Biochemistry, University of Oxford, UK C. elegans development: getting the seams right Victoria Sanz Moreno, King’s College London, UK Signalling pathways controlling amoeboid tumour dissemination Genes to Cells Kazuhiro Maeshima, National Institute of Genetics, Japan Chromatin fluctuation in live mammalian cells 192 Roop Mallik, Tata Institute of Fundamental Research, Mumbai, India Biophysics of Motor protein driven transport in Phagosome maturation Scientific report 2014 LONDON RESEARCH INSTITUTE Conferences 31st March - 1 April LRI Retreat The last LRI Retreat took place at the University of Oxford from 31st March - 1 April. 360 delegates from Lincoln’s Inn Fields and Clare Hall enjoyed a packed couple of days in the city of Oxford. The Retreat began with talks from newer group leaders who have not attended the LRI Retreats before: Nate Goehring, Paola Scaffidi and Alessandro Costa, along with Borbala Mifsud who spoke on behalf of Nick Luscombe. There was also a slot dedicated to the Create the Change Campaign, CRUK’s campaign to raise money for the Francis Crick Institute. These talks were followed by a busy and successful poster session. The next day saw a busy schedule of talks from more group leaders and Core Technology Facilities heads, (Dinis Calado, Peter Cherepanov, Hasan Yardimci, Francois Lassailly, Bram Snijders) and a few postdocs who had been nominated to speak - including Marco Sapanaro from the Mechanisms of Gene Transcription Group, and Sophie Acton from the Immunobiology Group. Barbara Schraml (Immunobiology Group), winner of the 2013 Hardiman-Redon Prize also gave a talk. Poster session at the last LRI retreat, Oxford, April 2014 23rd June Postdoc Retreat The 3rd Francis Crick Institute Postdoc Retreat was held on Monday 23rd June 2014 in association with the British Library at their conference centre. The theme of the event was ‘Inspiring Science’ and consisted of speakers from academia and industry offering a diverse range of talks. The Retreat was opened by the Francis Crick Institute Research Directors Richard Treisman (LRI Director) and Jim Smith (NIMR Director and MRC Deputy Chief Executive and Director of Strategy), and Roly Keating, Chief Executive of the British Library. The speakers were at various stages in their career with inspirational stories to tell about how they got there. Speakers included: Professor Mark Lythgoe, Founder and Director of the UCL Centre for Advanced Biomedical Imaging (CABI); Dr Claire Spottiswoode, Research Fellow at the Department of Zoology, University of Cambridge; Dr Nessa Carey, Senior Director in External R&D Innovation at Pfizer; Professor Julian Parkhill, Head of Pathogen Genomics at the Wellcome Trust Sanger Institute and Professor Olivier Voinnet leading expert in the field of small RNAs. All the speakers were very well received and their talks stimulated lively discussions. More tan 150 Postdocs from the Francis Crick Institute’s partner organisations attended and were given the opportunity to interact in a ‘Postdoc nexus’ networking session, and at the evening social event. More than 90% of attendees rated the retreat as ‘very good’ or ‘excellent’ and indicated that they would attend future Francis Crick Institute Postdoc Retreats. The Retreat was organised by a committee consisting of postdocs from the LRI and NIMR. 11th - 13th June International Graduate Student Conference This year’s International PhD Student Cancer Conference, was hosted and organised by students from the German Cancer Research Center (DKFZ) in Heidelberg. This annual two and a half day conference is held with other Cancer Research UK funded institutes (Beatson, Paterson, LRI and the Oxford Institute for Radiation Oncology) as well as the Netherlands Cancer Institute (NKI), the DKFZ, the European School of Molecular Medicine in Milan (SEMM: IFOM-IEO), the Vita-Salute San Raffaele University also in Milan (DIBT), and the Spanish National Cancer Centre (CNIO) in Madrid. All attendees presented their research - which covered many topics related to cancer, from basic biology to clinical aspects of the disease. The scientific talks and poster sessions were mixed with social activities. Lutz Gissman, Professor of Genome Modifications and Carcinogenesis at DKFZ, and Gottfried Schatz, Emeritus Professor of Biochemistry at the University of Basel gave highly engaging plenary lectures, and faced numerous excellent questions from the audience. SEMINARS AND CONFERENCES 193 EXTERNAL FUNDING Awards and Grants Association for International Cancer Research Axel Behrens – Mammalian Genetics Julian Downward – Signal Transduction Astellas Sharon Tooze – Secretory Pathways Medical Research Council Lucy Collinson – Electron Microscopy Dinis Calado – Immunity and Cancer Breast Cancer Campaign Mark Petronczki – Cell Division & Aneuploidy Erik Sahai – Tumour Cell Biology National Institute of Health Peter Cherepanov – Chromatin Structure and Mobile DNA British Council Holger Gerhardt – Vascular Biology Pfizer Charles Swanton – Translational Cancer Therapeutics Cephalon Peter Parker – Protein Phosphorylation Cancer Research UK Travel Award Holger Gerhardt – Vascular Biology Erik Sahai – Tumour Cell Biology European Molecular Biology Organisation (EMBO Young Investigator Prize) Mark Petronczki – Cell Division & Aneuploidy Barry Thompson – Epithelial Biology European Commission Axel Behrens – Mammalian Genetics Simon Boulton – DNA Damage Response Peter Cherepanov – Chromatin Structure and Mobile DNA John Diffley – Chromosome Replication Julian Downward – Signal Transduction Caroline Hill – Developmental Signalling Caetano Reis e Sousa – Immunobiology Jesper Svejstrup – Mechanisms of Gene Transcription Thomas Surrey – Microtubule Cytoskeleton Charles Swanton – Translational Cancer Therapeutics Richard Treisman – Signalling and Transcription Frank Uhlmann – Chromosome Segregation Stephen West – Genetic Recombination Genetech Dominique Bonnet – Haematopoietic Stem Cell Facundo Batista – Lymphocyte Interaction Leducq Holger Gerhardt – Vascular Biology Leukaemia and Lymphoma Research Dominique Bonnet – Haematopoietic Stem Cell 194 Louis Jeantet Paul Nurse – Cell Cycle Richard Triesman – Signalling and Transcription Steve West – Genetic Recombination Scientific report 2014 LONDON RESEARCH INSTITUTE Rosetrees Trust Charles Swanton – Translational Cancer Therapeutics Cancer Research Technology Reward to Inventors Peter Parker – Protein Phosphorylation Neil McDonald – Structural Biology The Scripps Research Institute Facundo Batista – Lymphocyte Interaction Unity through Knowledge Fund Adrian Hayday – Immuno Surveillance Weizmann Institute Giampietro Schiavo – Molecular Neuropathobiology Wellcome Trust Paul Nurse/Jacqueline Hayles – Cell Cycle Barry Thompson – Epithelial Biology Fellowships Canadian Institute of Health Research Jasmine Abella – Cell Motility Laurent L’Epicier-Sansregret – Cell Division and Aneuploidy/Translational Cancer Therapeutics Cancer Research UK Samra Turajlic – Translational Cancer Therapeutics European Commission – Marie Skłodowska-Curie actions Corella Casas Delucci – Chromosome Replication Ieva Gailite – Apoptosis and Proliferation Control Eishu Hirata – Tumour Cell Biology Jatta Huotari – Immunobiology Christoph Kurat – Chromosome Replication Nuria Martinez – Lymphocyte Interaction Marco Montagner – Tumour Cell Biology Maria Jose Martin Pereira – Genetic Recombination Claire Sheridan – Signal Transduction Joanna Soroka – Genetic Recombination Martina Wirth – Secretory Pathways FEBS Return-to-Europe-Fellowship Joseph Yeels – Chromosome Replication Fondazione Veronesi Italy Davide Zecchin – Signal Transduction German Academy of Sciences Katharina Deiss – Protein Phosphorylation Human Frontier Science Project Madhu Kumar – Signal Transduction Japan Society for the Promotion of Science Yasutaka Kakui – Chromosome Segregation Takayuki Koyano – Cell Regulation Sir Henry Wellcome Fellowship Sophie Acton – Immunobiology Esther Arwert – Tumour Cell Biology Hanna Mischo – Mechanisms of Gene Transcription Johanna Roostalu – Microtubule Cytoskeleton Patrycja Kozik – Immunobiology Frances Willenbrock – Protein Phosphorylation Swedish Research Council Omar Khan – Mammalian Genetics Swiss National Science Foundation Pierre Maillard – Immunobiology Kanagaraj Radhakrishnan – Genetic Recombination Student Fellowships Agency for Science, Technology and Research Minghao Chia – Cell Fate and Gene Regulation Boehringer Ingelheim Fund Agnieszka Janska – Chromosome Replication James Patterson – Cell Cycle Janneke Van Blijswijk – Immunobiology EC Marie Curie Initial Training Network Tom Watkins – Translational Cancer Therapeutics Fundação para a Ciência e a Tecnologia Filipa Neto – Vascular Biology Overseas Research Scholarship - UCL Djamil Damry – Immunity and Cancer Melisa Guven – Mammalian DNA Repair Marina Ivanova – Structural Biology Carlson Tsui – Lymphocyte Interaction Medical Research Council Rafael Di Marco Barros – Immuno Surveillance Medical Research Council Marco Gerlinger – Translational Cancer Therapeutics Ministerio de Ecomomia y Competitividad Ander Abarrategi – Haematopoietic Stem Cell Netherlands Organisation for Scientific Research Annemarthe van der Veen – Immunobiology EXTERNAL FUNDING 195 Institute management London Research Institute Management Committee Richard Treisman PhD FRS (Chair) Julian Downward PhD FRS John Diffley PhD FRS Erik Sahai PhD Michael Way PhD Stephen West PhD FRS Ava Yeo PhD London Research Institute Faculty Committee Richard Treisman PhD FRS (Chair) Michael Way PhD (Co-Chair) Simon Boulton PhD Holger Gerhardt PhD Caroline Hill PhD Neil McDonald PhD Erik Sahai PhD Jesper Svejstrup PHD FRS Frank Uhlmann PhD Ava Yeo PhD (in attendance) Julian Downward PhD FRS (ex officio) John Diffley PhD FRS (ex officio) London Research Institute Fellowships Committee John Diffley PhD FRS (Chair) Nic Tapon PhD (Deputy Chair) Simon Boulton PhD Adrian Hayday PhD Ilaria Malanchi PhD Mark Petronczki PhD Neil McDonald PhD Peter Parker PhD FRS Sharon Tooze PhD Takashi Toda PhD Sally Leevers PhD (ex officio) Ava Yeo PhD (in attendance) 196 Scientific report 2014 LONDON RESEARCH INSTITUTE London Research Institute Graduate Students Advisors Committee Sally Leevers PhD (Chair) Peter Cherepanov PhD Caroline Hill PhD Thomas Surrey PhD Jesper Svejstrup PhD Charles Swanton PhD Nic Tapon PhD Kaila Srai PhD (UCL) Ava Yeo PhD (in attendance) Andrew Brown (in attendance) Sabina Ebbols (in attendance) CONTACT DETAILS London Research Institute Scientific Report 2014 Kings Cross St Pancras Lincoln’s Inn Fields Laboratories 44 Lincoln’s Inn Fields London WC2A 3LY ON ST EU AD RO GR Euston NR S IN AY Clare Hall Laboratories Blanche Lane South Mimms Potters Bar Hertfordshire EN6 3LD D OA Russell Square U SO ON PT AM TH RO W Holborn Cancer Research UK Registered charity in England & Wales (1089464), Scotland (SC041666) and the Isle of Man (1103). A company limited by guarantee. Registered as a company in England and Wales (4325234) and the Isle of Man (5713F). Registered address: Angel Building, 407 St John Street, London EC1V 4AD. No. 44 RE G AC AL N LO Lincoln’s Inn Fields YCH DW Temple Covent Garden Leicester Square D AN OO RL TR BR ES TH N TE WA E IDG Tel 44 (0) 20 7242 0200 www.cancerresearchuk.org LBORN O HIGH H 500 metres Charing Cross An electronic version of this report can be found at: http://crick.ac.uk/news/publications/ Waterloo ISSN 1479-0378 Copyright © 2014 Cancer Research UK GR EYH OU ND E AN EL EA LA NE CH RL SL N LA B AN E Printed by Howard Hunt Group BIGNELLS CORNER 5 M2 NS R LBA ST A OAD Clare Hall Laboratories JUNCTION 29 E NCH BLA BIGNELLS CORNER E LAN A1081 A1 FR OM LO ND ON N London Research Institute Cancer Research UK London Research Institute 44 Lincoln’s Inn Fields London WC2A 3LY United Kingdom