2012 report - Cambridge Institute for Medical Research
Transcription
2012 report - Cambridge Institute for Medical Research
School of Clinical Medicine Cambridge Institute for Medical Research Research Report 2012 Front cover illustration: Crystal structure of the lysosomal enzyme galactocerebrosidase which, when defective, causes widespread demyelination resulting in the fatal neurodegenerative disorder Krabbe disease. The structure illustrates how the substrate-mimic galactose binds in the active site. Figure modified from Deane et al. (2011) Proc Natl Acad Sci USA. Cambridge Institute for Medical Research University of Cambridge Wellcome Trust/MRC Building Hills Road Cambridge CB2 0XY www.cimr.cam.ac.uk Photography by Media Studio, Cambridge University Hospitals NHS Foundation Trust Print management by H2 Associates (Cambridge) School of Clinical Medicine Research Report 2012 Contents 4 Foreword by Paul Luzio 40 MRC Mitochondrial Biology Unit 6 CIMR Principal Investigators 7 Science in the Institute 7 Folma Buss 8 Janet Deane 9 Andres Floto 10 Bertie Göttgens 11 Tony Green 12 Fiona Gribble 13 Gillian Griffiths 14 James Huntington 15 Brian Huntly 16 Fiona Karet 17 Paul Lehner 18 David Lomas 19 Paul Luzio 20 Stefan Marciniak 21 Katrin Ottersbach 22 David Owen 23 Lucy Raymond 24 Randy Read 25 Evan Reid 26 Frank Reimann 27 Margaret Robinson 28 David Rubinsztein 29 Christopher Rudd 30 Peter St George-Hyslop 31 Matthew Seaman 32 Symeon Siniossoglou 33 Ken Smith 34 John Todd 35 The Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory 36 John Trowsdale 37 Linda Wicker 38 Geoff Woods 41 CIMR Affiliated Principal Investigators 41 Jenefer Blackwell 41 Damian Crowther 42 Sadaf Farooqi 42 Sergey Nejentsev 43 Stephen O’Rahilly 43 David Ron 44 Principal Investigators who will be moving on from CIMR shortly 44 David Clayton 44 Andrew Peden 46 Postgraduate Opportunities in the Institute 47 CIMR Research Retreat 2011 48 Administration, IT and Technical Support 50 Funding of CIMR 52 NIHR Cambridge Biomedical Research Centre core facilities supporting CIMR PIs 53 Other Information 57 Publications for Scientists in CIMR since 2010 79 Regulations for CIMR 3 Foreword: Cambridge Institute for Medical Research Paul Luzio CIMR Director Mark Dawson Rhys Roberts Wellcome-Beit Prize/ Wellcome Trust Intermediate Clinical Fellows 4 The Cambridge Institute for Medical Research (CIMR), which is housed in the Wellcome Trust/ MRC Building, is a cross-departmental institute of the University of Cambridge Clinical School. It provides a unique interface between basic and clinical science, houses over 250 scientists and has as its major goal, the determination and understanding of the molecular mechanisms of disease. Over 40% of our Principal Investigators (PIs) are medically qualified and clinically active, a percentage that has remained steady since the CIMR opened in 1998. Our annual research grant expenditure is now the third highest in the University behind the Departments of Engineering and Physics (Cavendish Laboratory). In the University’s financial year 2010–11 the expenditure exceeded £21M, with about 60% funded by the Wellcome Trust and 15% by the MRC (see p 50). Research in the CIMR is led by individual Principal Investigators (PIs) who determine their own research priorities and follow their own instincts. It is focussed on four research themes that transcend individual research groups, namely misfolded proteins and disease, intracellular membrane traffic in health and disease, autoimmune disease and haematopoietic stem cells and their disorders. In each of these themes, there have been many recent discoveries and advances that are referenced in this Research Report on the pages of individual PIs. Examples of activities that are further down the translational path to patient benefit include (i) the introduction by Tony Green of the diagnostic use of genetic mutations for frontline investigation of myeloproliferative disorders that are now a central part of national and international guidelines; (ii) Ken Smith’s identification and use of novel transcription signatures to predict prognosis in autoimmune disease; (iii) the clinical safety trial of an autophagy-inducing drug for Huntington’s disease following David Rubinsztein’s pioneering studies of the activation of autophagy to clear mutant huntingtin in cell and animal models of Huntington’s disease; (iv) David Lomas’ high through-put screens to identify small molecules to block serpin aggregation and so treat patients with serpinopathies and (v) the interactions of Fiona Gribble and Frank Reimann with pharmaceutical companies to develop new therapies for type II diabetes. Taken together our four research themes constitute a comprehensive approach to molecular and cellular mechanisms of disease in which there is clear ‘added value’ from the extensive interactions between basic and clinical scientists and across different themes. A question that is increasingly asked about research activities funded by major grant giving bodies in Universities is how they fit with the University’s overall research strategy. In a recent submission to the Wellcome Trust, the Heads of the Schools of Biological Sciences and Clinical Medicine have pointed out that the ability to attract outstanding biomedical scientists working on important problems increasingly depends upon the establishment of ‘critical mass’ and the provision of optimal research infrastructure. Together, these have required a change in strategic approach exemplified by the establishment and development within the University of cross-departmental, interdisciplinary institutes including the CIMR. There are ten strategic research themes within the basic biological and biomedical sciences that cross the two Schools and the CIMR plays a major role in two of these, Structural and Cell Biology and its Application to Medicine, and Genomics and its Application to Medicine. PIs in the CIMR also make a significant contribution to four other strategic themes, Infection and Immunity, Neurosciences and Mental Health, Stem Cell Biology and Medicine, and Systems Medicine. The research activities in the CIMR are complementary to those in other research institutes and departments of the University and NHS as well as major research institutes in the Cambridge area, with many of whom we have excellent interactions. Through the activities and collaborations of individual PIs and often through the placing of research students from PhD programmes administered through the CIMR, we have active interactions with the Wellcome Trust Sanger Institute, the MRC-Laboratory of Molecular Biology, the Babraham Institute, the CRUK-Cambridge Research Institute, the MRC Cancer Cell Biology Unit, the Wellcome Trust/ CRUK Gurdon Institute and the Wellcome Trust Centre for Stem Cell Research. Within the Clinical School, three of our PIs are currently Heads of Department, namely Ken Smith (Medicine), Tony Green (Haematology) and John Todd (Medical Genetics). We have recently strengthened our relationship with the Institute of Metabolic Science through the appointment of one of their PIs, David Ron to our Institute Management Committee. CIMR PIs also contributed significantly to the recent successful renewal of the NIHR Cambridge Biomedical Research Centre (BRC). John Todd, David Rubinsztein, Ken Smith, Tony Green and David Lomas are all theme leads in the BRC and Peter St George-Hyslop is the lead PI for the new NIHR Cambridge Dementia Biomedical Research Unit. No research strategy or focus can be meaningful without the appointment and nurturing of outstanding individual scientists. A remarkably high proportion of our PIs have part or all of their salaries funded through very competitive fellowships awarded by biomedical funding agencies. The CIMR presently has the highest number of Wellcome Trust Principal Research Fellows (8, although David Clayton will retire in March 2012), in any institute or department in the country. Amongst these are David Owen and David Rubinsztein who won their awards in the past two years. We are also proud of our record in training and developing young scientists including clinician scientists. In the past two years, two young CIMR clinician scientists, Mark Dawson and Rhys Roberts were awarded WellcomeBeit Prize Fellowships. It was especially pleasing that Rhys Roberts and also Mike Weekes gained their intermediate fellowships from the Wellcome Trust following periods funded as CIMR Next Generation Fellows to help them back into research from full time clinical posts. We hope that in time all of our intermediate clinical research fellows will do well enough to be competitive for senior fellowships, following in the footsteps of other clinician scientists such as Evan Reid and Stefan Marciniak who became CIMR PIs in recent years. We also take seriously the development and promotion of women scientists. It is a pleasure to note that Katrin Ottersbach has taken up a Bennett Senior Fellowship from Leukaemia and Lymphoma Research and that Janet Deane has been appointed as one of our new PIs following the recent award of a Royal Society University Research Fellow- ship. In the CIMR we are aware that our track record of consistently having around 30% of our PIs who are women compares favourably with other institutes. However, this proportion must be at least maintained and preferably increased. Therefore, we will fully support Fiona Karet, one of our PIs, in her work leading the Clinical School’s efforts to develop an application for an Athena Swan Silver Award. We must aim to improve our recruitment, retention and promotion of women in the CIMR as well as elsewhere in the Clinical School. I have referred briefly above to the imminent retirement of David Clayton who has contributed so much to the work of the Diabetes and Inflammation Laboratory in the CIMR. I thank David for his work in the institute and wish him well in the future. There are two other people who will reach retirement in 2012 that I also wish to thank for their support of the CIMR. The first is Sir John Walker FRS, the Director of the MRC Mitochondrial Biology Unit with whom we share our building. I hope that we are able to build up equally good relations with his successor. The second is the Regius Professor of Physic, Sir Patrick Sissons who has been a stalwart supporter of the CIMR both as Regius and in his previous role as Head of the Department of Medicine. It has been a great pleasure to work with Patrick who has led the Clinical School with great wisdom and care to a point where it is poised for very exciting developments in the future. In the CIMR the major challenges are both to continue to improve our science and to be a flagship in the UK for interdisciplinary research at the interface between basic and clinical science. We remain very grateful for the Wellcome Trust Strategic Award, currently extended until late 2012. That award greatly enhances our efforts as a translational institute and specifically supports our core scientific facilities, funds PhD studentships and also provides funds to attract young clinicians back into research. Our high proportion of clinically active PIs allows the rapid translation of scientific breakthroughs into clinical practice. In the coming years such translation is likely to include the identification of new pathways of disease, the identification of new small molecule and cell based therapies to treat protein misfolding diseases and new molecular diagnostics for haematological malignancies. However, there is no simple mechanical route from scientific research to clinical practice and it behoves us all to keep in mind the words of Marie Curie, who in an address to Vassar College in Poughkeepsie, New York on May 14th, 1921 said, “But we must not forget that when radium was discovered no one knew that it would prove useful in hospitals. The work was one of pure science. And this is a proof that scientific work must not be considered from the point of view of the direct usefulness of it. It must be done for itself, for the beauty of science, and then there is always the chance that a scientific discovery may become like the radium a benefit for humanity.” Paul Luzio January 2012 Current Membership of the CIMR Strategy Committee: Patrick Sissons (Chairman, Regius Professor of Physic), Alastair Compston (Clinical Neurosciences), John Danesh (IPH, Faculty Board), Tony Green (Haematology), David Lomas (Deputy Director, CIMR), Paul Luzio (Director, CIMR), Stephen O’Rahilly (Clinical Biochemistry), Bruce Ponder (CRUK CRI, Faculty Board), Geoffrey Smith (Pathology), Ken Smith (Medicine), John Todd (Diabetes and Inflammation Laboratory/Medical Genetics). Current Membership of the International Scientific Advisory Board: Nick Hastie, Chairman (University of Edinburgh), Dennis Ausiello (Harvard University), Peter Cresswell (Yale University), John Dick (University of Toronto), Louise Johnson (University of Oxford), Sandra Schmid (Scripps Research Institute), Patrick Vallance (GlaxoSmithKline plc) 5 CIMR Principal Investigators 6 Principal Investigator Investigator Status Home Department Folma Buss Wellcome Trust University Award Clinical Biochemistry Janet Deane Royal Society University Research Fellow Haematology Andres Floto Wellcome Trust Senior Research Fellow in Clinical Sciences Medicine Bertie Göttgens Personal Chair Haematology Tony Green University Chair Haematology Fiona Gribble Wellcome Trust Senior Research Fellow in Clinical Sciences Clinical Biochemistry Gillian Griffiths Wellcome Trust Principal Research Fellow and Personal Chair Medicine James Huntington Personal Chair Haematology Brian Huntly Senior Clinical Lecturer Haematology Fiona Karet Personal Chair Medical Genetics Paul Lehner Wellcome Trust Senior Research Fellow in Clinical Sciences and Personal Chair Medicine David Lomas University Chair and Deputy Director CIMR Medicine Paul Luzio Personal Chair and Director CIMR Clinical Biochemistry Stefan Marciniak MRC Senior Clinical Research Fellow Medicine Katrin Ottersbach Leukaemia & Lymphoma Research Bennett Senior Fellow Haematology David Owen Wellcome Trust Principal Research Fellow and Personal Chair Clinical Biochemistry Lucy Raymond University Reader Medical Genetics Randy Read Wellcome Trust Principal Research Fellow and Personal Chair Haematology Evan Reid Wellcome Trust Senior Research Fellow in Clinical Sciences Medical Genetics Frank Reimann Wellcome Trust Senior Research Fellow in Basic Biomedical Science Clinical Biochemistry Margaret Robinson Wellcome Trust Principal Research Fellow and Personal Chair Clinical Biochemistry David Rubinsztein Wellcome Trust Principal Research Fellow and Personal Chair Medical Genetics Christopher Rudd Wellcome Trust Principal Research Fellow and Personal Chair Pathology Peter St George-Hyslop Wellcome Trust Principal Research Fellow and Personal Chair Clinical Neurosciences Matthew Seaman MRC Senior Research Fellow Clinical Biochemistry Symeon Siniossoglou MRC Senior Research Fellow Clinical Biochemistry Ken Smith University Chair Medicine John Todd University Chair and Director DIL Medical Genetics John Trowsdale University Chair Pathology Linda Wicker Personal Chair and Co-Director DIL Medical Genetics Geoff Woods Personal Chair Medical Genetics Myosin Motor Proteins in Health and Disease We want to understand how molecular motor proteins function in intracellular transport processes and how defects in these molecular machines are linked to human diseases. Our primary interests are the actin-based myosin motors and their roles in cargo transport, cell signalling and membrane dynamics. The main focus of our current research is myosin VI, which is unique because it moves in the opposite direction along actin filaments to all the other myosins. Myosin VI is an extremely versatile motor that functions in membrane trafficking pathways associated with secretion and endocytosis and is thereby linked to cell migration and cytokinesis. We have shown that these diverse roles of myosin VI are mediated by its interaction with a wide range of distinct binding partners (adaptors) that connect it to different cargoes. Overexpression or mutations or the absence of myosin VI have been linked to such diverse pathological processes as deafness, cardiomyopathy, neurodegeneration and cancer. We are currently unravelling the novel function(s) of the myosins and their cargo adaptors in neurodegenerative diseases such as Alzheimer’s and motor neuron disease. In addition we are investigating the molecular role of myosin VI in cardiomyocytes and heart disease. The human genome contains 40 distinct myosins and therefore we are expanding our studies to investigate the complementary roles of these different myosins in membrane trafficking pathways. For example we are currently focusing on the role of class I myosins in endocytic membrane recycling. Our long-term goal is to establish how the different actin- and microtubule-based motor proteins interact together and how their actions are coordinated and regulated to maintain the health and functions of cells. Cargo attachment of myosin VI is mediated by binding partners identified in Mammals (yellow) and in Drosophila (blue) (from Buss & Kendrick-Jones (2011) PNAS 108, 5927–5928). Myosin VI and its adaptor GIPC colocalise on early endosomes. Folma Buss Sue Arden Lisa Bond Hemma Brandstätter David Tumbarello Bennett Waxse Funding: Wellcome Trust National Institutes of Health (USA) Puri, C., Chibalina, M. V., Arden, S. D., Kruppa, A. J., KendrickJones, J. and Buss, F. (2009). Overexpression of myosin VI in prostate cancer cells enhances PSA and VEGF secretion, but has no effect on endocytosis. Oncogene 29, 188–200. Chibalina, M. V., Poliakov, A., Kendrick-Jones, J. and Buss, F. (2010). Myosin VI and optineurin are required for polarised EGFR delivery and directed migration. Traffic 11, 1290–1303. Bond, L. M., Peden, A. A., Kendrick-Jones, J., Sellers, J. R. and Buss, F. (2011). Myosin VI and its binding partner optineurin are novel players in secretory vesicle fusion at the plasma membrane. Mol Biol Cell 22, 54–66. 7 Janet Deane Chris Hill Funding: The Royal Society Schneider, M. C., Prosser, B. E., Caesar, J. J., Kugelberg, E., Li, S., Zhang, Q., Quoraishi, S., Lovett, J. E., Deane, J. E., Sim, R. B., Roversi, P., Johnson, S., Tang, C. M. and Lea, S. M. (2009). Neisseria meningitidis recruits factor H using protein mimicry of host carbohydrates. Nature 458, 890–893. Deane, J. E., Abrusci, P. Johnson, S. and Lea, S. M. (2010). Timing is everything: the regulation of type III secretion. Cell Mol Life Sci 67, 1065– 1075. Host-pathogen Interactions During Shigella Infection Diseases caused by bacteria, viruses and other parasites are major causes of death, disability, and social and economic disruption for millions of people. Our ability to effectively treat infectious diseases is being progressively depleted due to increasing resistance of microbes to antimicrobial drugs. A top priority today is the development of new low-cost drugs that can replace those that are becoming ineffective. An essential part of developing new therapeutics is a clear molecular understanding of how pathogens evade our immune defences and hijack our cellular components. Our research focuses on the bacterial pathogen Shigella flexneri. This pathogen invades cells of our digestive tract and hijacks components of the actin polymerization pathway in order to aid its movement within our cells and allowing it to spread directly from cell to cell. In this way Shigella can limit its detection by our Deane, J. E., Graham, S. C., Kim, N. N., Stein, P. E., McNair, R., Cachón-González, M. B., Cox, T. M. and Read, R. J. (2011). Insights into Krabbe disease from structures of Galactocerebrosidase. Proc Natl Acad Sci USA 108, 15169– 15173. Structure of the lysosomal enzyme galactocerebrosidase illustrating clinicallyidentified mutations which cause the fatal neurodegenerative disorder Krabbe disease. Figure taken from Deane et al. (2011) PNAS. 8 immune system. As an extra defence S. flexneri produces proteins that specifically disarm the intracellular immune response to pathogens. We study the structure, interactions and regulation of Shigella virulence proteins and the host proteins they target. In collaboration with the laboratories of Prof Read and Prof Luzio in the CIMR and Prof Cox in the Department of Medicine we are also studying how clinically-identified mutations of the enzyme galactocerebrosidase (GALC) result in Krabbe disease, a devastating neurodegenerative disorder. Our recent structure of GALC has provided new insight into the molecular role of pathogenic mutations and allowed us to identify patients that might be responsive to a new form of therapy known as pharmacological chaperone therapy. Antigen Processing by Macrophages and Dendritic Cells We are interested in understanding how antigen processing is controlled by macrophages and dendritic cells and how it may be dysregulated in autoimmune disease and subverted by pathogenic bacteria. Our research is focused on two interrelated areas: 1. Control of phagosomal function Particulate antigen and bacteria are internalised into a membrane compartment known as the phagosome that successively fuses with early and late endosomes and finally lysosomes. The behaviour of the phagosome regulates cargo degradation and bacterial killing, cytosolic export of antigen for crosspresentation and inflammasome activation. Using a variety of live cell imaging, cellular and biochemical techniques, we are examining how i) signalling from Fc! receptors (Fc!Rs), Tolllike receptors (TLRs) and scavenger receptors interact to control phagosomal function in human macrophages and dendritic cells; ii) how ion channels and transporters modulate phagosomal pH and peri-phagosomal calcium Andres Floto levels to control cargo degradation and lysosomal fusion; and iii) how macro-autophagy pathways interact with phagosomes to determine functional outcomes. 2. Mycobacterial infection We are studying how M. tuberculosis and non-tuberculous mycobacteria (NTM) interact with macrophages and dendritic cells particularly i) defining novel host factors controlling bacterial detection, intracellular killing and antigen presentation (through functional screening of genes identified from genetic studies); ii) examining whether therapeutic enhancement of autophagy can be used to improve intracellular killing and how inadvertent pharmacological block of autophagy might predispose to NTM infection; iii) identifying new genetic determinants controlling the pathogenesis of the NTM species M. abscessus through the combination of whole genome sequencing (in collaboration with Julian Parkhill, WTSI), in vitro functional phenotyping and clinical disease characteristics. Karen Brown Dorothy Grogono Krisztina Hegyi Lucy Hepburn Tracey Pollard Elspeth Potton Catherine Schaffner Mark Schiebler Funding: Wellcome Trust Medical Research Council NIHR Cambridge Biomedical Research Centre Papworth Hospital Health Enterprise East SBRI Floto, R. A., MacAry, P. A., Boname, J. M., Mien, T. S., Houben, E. N. G., Pieters, J., Day, C., Oehlmann, W., Singh, M., Smith, K. G. C. and Lehner, P. J. (2006). Dendritic cell stimulation by mycobacterial HSP70 is mediated through the HIV coreceptor CCR5. Science 314, 454–458. Williams, A., Sarkar, S., Cuddon, P., Ttofi, E. K., Saiki, S., Siddiqi, F. H., Jahreiss, L., Fleming, A., Pask, D., Goldsmith, P., O’Kane, C. J., Floto, R. A. and Rubinsztein, D. C. (2008). Novel targets for Huntington’s disease in an mTOR-independent autophagy pathway. Nat Chem Biol 4, 295–305. Calcium oscillations surrounding phagosomes containing IgG-coated beads (1,2) are detected in primary macrophages using the membrane-targeted calcium reporter Lck-GCaMP3 and are only partially mirrored by plasma membrane calcium signalling (3). Renna, M., Schaffner, C., Brown, K., Shang, S., Henao Tamayo, M., Hegyi, K., Grimsey, N. J., Cusens, D., Coulter, S., Cooper, J., Bowden, A. R., Newton, S. M., Kampmann, B., Helm, J., Jones, A., Haworth, C. S., Basaraba, R. J., DeGroote, M. A., Ordway, D. J., Rubinsztein, D. C. and Floto, R. A. (2011). Azithromycin blocks autophagy and may predispose cystic fibrosis patients to mycobacterial infection. J Clin Invest 121, 3554–3563. 9 Bertie Göttgens Fernando Calero-Nieto Yi-Han Cheng Evangelia Diamanti Debbie Goode Rebecca Hannah Anagha Joshi Sarah Kinston Vicki Moignard Felicia Ng Judith Schuette Jonathan Sive Aileen Smith Marloes Tijssen Adam Wilkinson Nicola Wilson Transcriptional Control of Normal and Leukaemic Blood Stem / Progenitor Cells Haematopoiesis has long served as a model system for studying the molecular processes that control cell fate decisions within complex differentiation cascades. Underpinning the molecular control of haematopoiesis are core circuits of regulatory networks, that when unified define the gene regulatory state of a particular cell. These regulatory networks are composed of both the transcription factors and the cis-regulatory elements they are bound to. Regulatory network reconstruction therefore requires the identification of cis-regulatory elements as well as the upstream factors which bind them. The long-term research goal of the Göttgens group is to decipher the molecular hierarchy of regulatory networks responsible for blood stem cell and endothelial development. To this end, the group uses complementary state-of-the-art approaches including embryonic stem cell and transgenic assays, bioinformatics, high throughput sequencing and mathematical modelling. 10 The importance of transcriptional control in both normal and leukaemic cells is underlined by the large number of transcription factor genes that cause leukaemia when disrupted or mutated. Future work will address how transcriptional programmes are perturbed in specific subtypes of leukaemia and may thus open up new avenues for the development of targeted therapies. A Regulatory Network Model for Blood Stem Cells. Shown are experimentally validated regulatory interactions for a core set of transcriptional regulators in blood stem/progenitor cells. Funding: Leukaemia & Lymphoma Research BBSRC Medical Research Council Cancer Research UK Leukemia & Lymphoma Foundation (USA) EU/Marie Curie EMBO NIHR Cambridge Biomedical Research Centre NC3Rs Wilson, N. K. et al. (2010). Combinatorial transcriptional control in blood stem/progenitor cells: Genome-wide analysis of 10 major transcriptional regulators. Cell Stem Cell 7, 532–544. The cumulative output of more than 40 research papers over the last 4 years has been the development of the most comprehensive network model for any adult stem cell type with over 40 transcription factors and more than 100 in vivo validated direct functional interactions. This integrated approach has resulted in the discovery of previously unrecognised combinatorial interactions between key regulators of blood stem cells with important implications for the transcriptional control of stem cell development and differentiation. Griffiths, D. S. et al. (2011). LIF independent JAK signalling to chromatin in embryonic stem cells uncovered from an adult stem cell disease. Nat Cell Biol 13, 13–21. Tijssen, M. R. et al. (2011). Genome-wide analysis of simultaneous GATA1/2, RUNX1, FLI1, and SCL binding in megakaryocytes identifies hematopoietic regulators. Dev Cell 20, 597–609. Haematopoietic Stem Cells and Haematological Malignancies The JAK/STAT pathway plays a key role in stem cell biology and is implicated in many human malignancies. A single acquired gain-of-function V617F mutation in JAK2 is present in most patients with a myeloproliferative neoplasm (MPN), a spectrum of clonal haematological malignancies which arise in the haematopoietic stem cell compartment. These disorders are experimentally tractable, permit clonal analysis and provide a window on the earliest stages of tumorigenesis. The Green lab is focussing on the pathogenesis of the MPNs and particularly on the molecular and cellular consequences of JAK2 mutations. Recent highlights of general relevance for stem cell and cancer biology include unexpected insights into chromatin biology, clonal evolution and haematopoietic stem cell function. non-CML MPNs. Our subsequent results suggest that V617F-positive ET and PV form a phenotypic continuum and that V617F homozygosity contributes to the PV phenotype. We made the surprising observation that leukaemic transformation is associated with loss of the JAK2V617F mutation and identified a cluster of new JAK2 mutations which define a previouslyunrecognised MPN variant. In addition we have demonstrated that inhibition of Bcl-xL deamidation represents a novel mechanism underlying DNA damage accumulation. Most recently we have described a novel nuclear function for JAK2 in the phosphorylation of histone H3, have demonstrated that JAK2V617F unexpectedly impairs haematopoietic stem cell function and have identified an unexpected role for STAT1 signalling in MPN pathogenesis. Human MPNs represent a spectrum of clonal haematological malignancies, with four main members: polycythaemia vera (PV), essential thrombocythaemia (ET), primary myelofibrosis (PMF) and chronic myeloid leukaemia (CML). A single acquired gain-of-function JAK2V617F mutation is present in most patients with These data are laying the foundation for new approaches to the diagnosis, classification and therapy of the myeloproliferative disorders – JAK2 mutation status is already embedded in clinical guidelines and JAK2 inhibitors are in clinical trials. Tony Green Jong-Sook Ahn Maria Ahn Athar Aziz Edwin Chen Jyoti Evans Anna Godfrey Tina Hamilton David Kent Ana Leal Cervantes Juan Li Charlie Massie June Park Dean Pask Rachel Pestridge Yvonne Silber Funding: Medical Research Council Leukaemia & Lymphoma Research Leukemia & Lymphoma Society (USA) Cancer Research UK Kay Kendall Leukaemia Fund Scott, L. M. et al. (2007). JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med 356, 459–468. Dawson, M. A.†, Bannister, A. J.†, Göttgens, B., Foster, S. D., Bartke, T., Green, A. R.* and Kouzarides, T.* (2009). JAK2 phosphorylates histone H3Y41 and excludes HP1" from chromatin. Nature 461, 819–822. * joint senior authors; † joint first author Analysis of individual haematopoietic colonies revealed an unexpected role for STAT1 signalling in the pathogenesis of distinct myeloproliferative disorders. Colonies carrying an identical JAK2V617F mutation from patients can exhibit differential activation of the STAT1 signalling pathway which provides an explanation for the distinct clinical phenotypes associated with these disorders (Chen et al. Cancer Cell 2010). ET, essential thrombocythaemia; PV, polycythaemia vera. Chen, E. et al. (2010). Distinct clinical phenotypes associated with JAK2V617F reflect differential STAT1 signaling. Cancer Cell 18, 524–535. 11 Fiona Gribble Alice Adriaenssens Rune Kuhre Claire Meek Helen Parker Paul Richards Gwen Tolhurst Funding: Wellcome Trust European Foundation for the Study of Diabetes Lister Institute of Preventive Medicine BBSRC/AstraZeneca European Union FP7 Full4Health Stimulus-secretion Coupling Mechanisms in Intestinal Enteroendocrine Cells Hormones from the gastrointestinal tract play key roles in the control of appetite and insulin release. Drugs based around one of these hormones, glucagon like peptide-1 (GLP-1), have recently proved to be highly successful for treating type 2 diabetes. This has led to the current focus on developing strategies to enhance the release of endogenous GLP-1 and other gut peptides, in the hope that this may identify novel treatments for diabetes and obesity and mimic the beneficial physiological effects of gastric bypass surgery. Understanding the mechanisms underlying secretion from gut endocrine cells is central to this approach, and forms the focus of our research. In collaboration with the group of Dr Frank Reimann, we employ a variety of optical and electrophysiological recording techniques to monitor stimulus detection and vesicle release from primary and immortalised GLP-1 secreting L cells. As the identification of living gut endocrine cells was a previous major impediment to Greenfield, F. R., Farooqi, I. S., Keogh, J., Henning, E., Habib, A. M., Blackwood, A., Reimann, F., Holst, J. J. and Gribble, F. M. (2009). Oral glutamine increases circulating GLP-1, glucagon and insulin levels in lean, obese and type 2 diabetic subjects. Am J Clin Nutr 89, 106–113. Tolhurst, G., Zheng, Y., Parker, H. E., Habib, A. M., Reimann, F. and Gribble, F. M. (2011). Glutamine triggers and potentiates glucagon-like peptide-1 secretion by raising cytosolic Ca2+ and cAMP. Endocrinol 152, 405–413. Parker, H. E., Wallis, K., le Roux, C. W., Wong, K. Y., Reimann, F. and Gribble, F. M. (2011). Molecular mechanisms underlying bile acid stimulated glucagon-like peptide-1 secretion. Br J Pharmacol (in press). A. Intracellular [glucose] in GLP-1 secreting cells monitored by a FRET-based glucose sensor, showing that blocking GLUT transporters abolishes glucose uptake. B. Model of L-cell glucose-sensing. 12 this field of research, we have generated transgenic mouse models in which GLP-1-expressing cells, or other target cells of interest, are labelled by cell-specific fluorescent markers. These are amenable to purification by flow cytometry and identification in primary culture for single cell recordings. The combination of single cell recordings with transcriptional analysis of FACS-purified cell populations has generated new models for nutrient sensing by L cells. Detection of nutrients in the gut lumen appears to involve electrogenic ion-coupled transporters located on the apical L cell membrane, generating small electrical signals that are subsequently amplified by voltage gated Ca2+ entry and other signalling pathways such as cAMP elevation and stored Ca2+ release. Comparisons between different enteroendocrine cell types suggest that the pathways identified in L cells are likely to reflect more general stimulus detection mechanisms employed by a wider population of gut endocrine cells. Control of Secretion at the Immunological Synapse Cells of the immune system need to communicate in order to co-ordinate an effective immune response. The immunological synapse formed between effector cells and their antigen-presenting cells provides a mechanism for directed communication, either via cell surface receptors or secreted proteins. Secretion is focused within the synapse: in the case of cytotoxic T lymphocytes (CTL) this is essential so that only the infected target is destroyed. This lab studies the mechanisms that control secretion within the immunological synapse using a range of functional, biochemical and imaging techniques. We have close clinical collaborations that allow us to study CTL from patients with genetic diseases (eg Haemophagocytic Lymphohistiocytosis) that disrupt secretion at the immunological synapse, and thereby identify the machinery required for granule delivery and fusion at the synapse as well as providing a better understanding of the genetic disease. We have discovered that CTL and NK cells use a novel secretory mechanism, with the centrosome polarizing to the precise site of secretion within the immunological synapse. This mechanism, that requires the centrosome to dock at the plasma membrane, bears striking similarities to cilia formation, with endocytosis and exocytosis focused at the point of centrosome docking in both. We are currently exploring the molecular similarities between cilia and synapse formation using genetic, morphological and functional studies. CTL provide an excellent model for understanding the mechanisms that control centrosome polarization, and we have exploited genetic models in which components of the T cell receptor signaling pathway can be turned off. A key aspect to all of these studies is the use of live imaging to follow synapse formation and secretion in real time, and dissect out the delivery of the secretory machinery to the immunological synapse. A cytotoxic T cell, with the membrane labelled with CD8 (purple), and microtubules (green) polarised towards a target cell. The secretory lysosomes of the CTL (red) move towards the microtubule organising centre and deliver their contents at the immunological synapse, leading to the destruction of the target. Nuclei of both CTL and target are stained blue. Gillian Griffiths Karen Angus Yukako Asano Maike de la Roche Sam Grieve Yvonne Hackmann Jeansun Lee Alex Ritter Jane Stinchcombe Roseanne Zhao Funding: Wellcome Trust Griffiths, G. M., Tsun, A. and Stinchcombe, J. C. (2010). The immunological synapse: a focal point for endocytosis and exocytosis. J Cell Biol 189, 399–406. Daniele, T., Hackmann, Y., Ritter, A. T., Wenham, M., Booth, S., Bossi, G., Schintler, M., AuerGrumbach, M. and Griffiths, G. M. (2011). A role for rab7 in the movement of secretory granules in cytotoxic T lymphocytes. Traffic 12, 902–911. Tsun, A., Qureshi, I., Stinchcombe, J. C., Jenkins, M. R., de la Roche, M., Kleczkowska, J., Zamoyska, R. and Griffiths, G. M. (2011). Centrosome docking at the immunological synapse is controlled by Lck signaling. J Cell Biol 192, 663–674. 13 James Huntington Ty Adams Daniel Johnson Bernhard Lechtenberg Giles Lewis Henriet Meems Thomas Murray-Rust Stéphanie Polderdijk Anja Pomowski Timothy Sendall Funding: Medical Research Council Wellcome Trust British Heart Foundation Molecular Recognition in Haemostasis Haemostasis (blood coagulation) is a complex process under tight regulatory control. Dysregulation leads to bleeding if the response is insufficiently robust, and to thrombosis if coagulation is not limited. Understanding these regulatory mechanisms is crucial for the prevention and treatment of diseases such as haemophilia, deep vein thrombosis, pulmonary embolism, heart attack and stroke. My lab studies the structures of individual coagulation factors and of the multi-protein complexes they form, using the techniques of X-ray crystallography and NMR, in order to understand how coagulation is controlled. One project focuses on the final protease generated by the blood coagulation cascade, thrombin. Other projects focus on the large multiprotein complexes responsible for factor Xa and thrombin formation, the intrinsic tenase (factors VIIIa and IXa) and prothrombinase (factors Va and Xa). Finally, all of the proteases generated during the haemostatic response must eventually be inhibited to avoid thrombosis. We have a long-standing interest in how serpins antithrombin, heparin cofactor II, protein C inhibitor and protease nexin-1 are regulated by cofactors such as heparin. We have solved several crystal structures of relevant complexes, including all of the antithrombin-heparin-protease complexes, and now have a detailed and complete description of how heparin regulates serpin function. The importance of serpins is highlighted by the association of deficiency with disease, often caused by missense mutations leading to the accumulation of ‘polymers’ within the endoplasmic reticulum of secretory cells. We recently solved two crystal structures of polymers that showed distinct large-scale domain swaps, and have demonstrated that the intermolecular swapping of the 30 C-terminal residues is the molecular basis of serpin polymerization within living cells. Johnson, D. J. D., Langdown, J. and Huntington, J. A. (2010). Molecular basis of factor IXa recognition by heparin-activated antithrombin revealed by a 1.7Å structure of the ternary complex. Proc Natl Acad Sci USA 107, 645–650. Lechtenberg, B., Johnson, D. J. D., Freund, S. and Huntington, J. A. (2010). NMR resonance assignments of thrombin reveal the conformational and dynamic effects of ligation. Proc Natl Acad Sci USA 107, 14087– 14092. Yamasaki, M., Sendall, T. J., Pearce, M. C., Whisstock, J. C. and Huntington, J. A. (2011). Molecular basis of "1-antitrypsin deficiency revealed by the structure of a domain-swapped trimer. EMBO Rep 12, 1011–1017. Crystal structure of an "1-antitrypsin trimer formed by the ‘swapping’ of the C-terminal portion from one molecule into another. 14 Mechanisms of Myeloid Leukaemogenesis and Leukaemia Stem Cell Biology The aims of our group are broadly to study mechanisms of leukaemogenesis. In particular we aim to characterise leukaemia stem cells (LSC) at the molecular and cellular level in myeloid haematological malignancies. Leukaemias and many other cancers have recently been demonstrated to be wholly dependent upon a population of so-called cancer stem cells for their continued growth and propagation. These cells represent the most critical targets for treatment of leukaemia and a greater understanding of their biology and its interface with normal haematopoietic stem cell (HSC) function is fundamental to improving treatment outcomes. We utilise a combination of functional and genomic assays in complementary mouse models and human primary cells to study normal HSC biology and how this is subverted in LSC and leukaemogenesis. An evolving paradigm in myeloid malignancies is transcriptional dysregulation and many of the mutations involved in myeloid leukaemogenesis involve transcriptional and epigenetic regulators. Ongoing projects within the group look at aberrant epigenetic regulation at chromatin via the bromodomain and extra terminal (BET) proteins in acute myeloid leukaemia (AML), at transcriptional dysregulation downstream of the AML-associated fusion genes MOZ-TIF2, NUP98-HOXA9 and MLL fusions, at the HMG-box gene SOX4, a critical AML target gene and at functions of epigenetic regulators such as CBP in HSC and LSC function. In addition, we are interested in the induction and evolution of myeloid preleukaemic disorders such as chronic myeloid leukaemia (CML) and myeloproliferative neoplasms (MPN) and are generating mouse models and studies in human cells to outline disease mechanisms and identify therapeutic targets. Our long-term aim is to improve biological understanding of leukaemogenesis, to identify rational targets for therapeutic intervention and to design novel therapeutics to improve the outcomes in myeloid malignancies. Brian Huntly Shubha Anand Polly Chan Mark Dawson George Giotopoulos Emma Gudgin Sarah Horton Sarah Putwain Frances Stedham Funding: Kay Kendall Leukaemia Fund Cancer Research UK Leukaemia & Lymphoma Research Wellcome Trust Leukemia & Lymphoma Society (USA) Anand, S., Stedham, F., Gudgin, E., Cambell, P., Beer, P., Green, A. R. and Huntly, B. J. P. (2011). Increased basal intracellular signaling patterns do not correlate with JAK2 genotype in human myeloproliferative neoplasms. Blood 118, 1610–1621. Kvinlaug, B. T. et al. (2011). Common and overlapping pathways contribute to the evolution of acute myeloid leukaemias. Cancer Res 71, 4117–4129. Protein-protein interaction inhibitors of BET bromodomain proteins (I-BET), prevent aberrant chromatin recruitment of transcriptional complexes and switch off expression programmes that drive LSC in MLL leukaemias. Dawson, M. A., Prinjha, R. K., Dittman, A., Giotopoulos, G., Bantscheff, M., Chan, W. I., Robson, S. C., Chung, C., Hopf, C., Savitski, M. M., Huthmacher, C., Gudgin, E., Lugo, D., Beinke, S., Soden, P. E., Döhner, K., Delwel, R., Burnett, A. K., Auger, K. R., Mirguet, O., Jeffrey, P., Drewes, G., Lee, K., Huntly, B. J. P.* and Kouzarides, T.* (2011). Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478, 529–533. *joint senior authors 15 Fiona Karet Molecular Physiology of Renal Tubular Homeostasis in Health and Disease Our aims are twofold: to characterize molecular mechanisms governing human renal tubular homeostasis (particularly acid-base balance) and to dissect functions of urinary proteins. Katy Blake-Palmer Zoe Golder Tannia Gracia Liz Norgett Caroline Robinson Graham Smith Ya Su Vivian Yiu Funding: Wellcome Trust NIHR Cambridge Biomedical Research Centre Kidney Research UK Intact distal nephron "-intercalated functions (secretion of protons in to the urine coupled to bicarbonate reclamation) are necessary for appropriate excretion of the acid load of a normal diet, and for generation of adequate amounts of bicarbonate for buffering. The transporters responsible, and their regulatory pathways, are complex and incompletely understood. We have used rare single-gene disorders where "-IC function is inadequate, imparting severely detrimental effects on the kidney’s ability to maintain normal body fluid pH. The clinical consequences are metabolic acidosis, rickets and calcium deposition in the kidney. Recessively inherited syndromes present with very severe changes in early childhood, and sensorineural hearing loss is often associated. Su, Y., Blake-Palmer, K. G., Best, A., Brown, A. C. N., Horita, S., Fry, A. C., Zhou, A., Smith, A. N., Toye, A. M. and Karet, F. E. (2011). Glyceraldehyde 3-phosphate dehydrogenase is required for band 3 (anion exchanger 1) membrane residency in the mammalian kidney. Am J Physiol 300, F157–166. Hiemstra, T. F., Charles, P. D., Hester, S. S., Karet, F. E.* and Lilley, K. S.* (2011). Uromodulin exclusion list improves urinary exosomal protein identification. J Biomol Techniques (in press). *joint authors Smith, G. D., Robinson, C., Stewart, A. P., Edwards, E., Karet, H. I., Norden, A. G. W., Sandford, R. N. and Karet Frankl, F. E. (2011). Characterization of a recurrent in-frame UMOD indel mutation causing late-onset autosomal dominant end-stage renal failure. Clin J Am Soc Nephrol (in press). Human urinary exosomes induce lysis of E. Coli. (A) proportion lysed; (B) lysed and (C) intact phenotypes. 16 Having described mutations in three genes: SLC4A1 (encoding the basolateral anion exchanger AE1), ATP6V1B1 and ATP6V0A4 (encoding kidney-specific B1 and a4 subunits of the "-IC surface proton pump), we initially showed that the latter two also participate in normal inner ear function, and discovered four novel subunit isoforms (C2, G3, d2, e2). We have gone on to functional characterization, and shown that essential membrane targeting motifs for AE1 reside in its C-terminal tail; that AE1 is regulated by GAPDH and interacts with the sodium pump. Conversely, we showed that binding of the glycolytic enzyme PFK-1 is essential for normal proton pump function, as is phosphorylation by PKA. We have catalogued the human urinary exosomal proteome and demonstrated that these urinary microvesicles are important innate immune effectors, maintaining urinary tract sterility. The most abundant urinary protein, uromodulin, is mutated in another dominant renal disease, FJHN. We have molecularly and clinically characterized a novel UMOD mutant, and continue with clinical and laboratory studies of other tubulopathies. Viral Immune Evasion We are interested in viral evasion of the immune system. As the ultimate intracellular parasites, viruses target the host cellular machinery to enable their replication and avoid elimination. Using functional genetic and proteomic technologies we identify novel cellular receptors manipulated by viruses, understand why these receptors are targeted and elucidate the mechanisms used by viruses to manipulate the cellular immunoreceptors. Why is the study of viral evasion mechanisms of interest? Understanding the mechanisms used by viruses to manipulate cellular processes provides unique insights into fundamental cell biological pathways, teaches us about viral pathogenesis and offers novel therapeutic approaches, for instance by targeting the newly identified receptor. Viral regulation of cell surface receptors – new approaches: Cell surface receptors are modulated by all intracellular pathogens. Our work on viral evasion of the MHC class I antigen presentation pathway fuelled an interest in how ubiquitin regulates immunoreceptor turnover. Paul J Lehner We identified the K3 and K5 viral ubiquitin E3 ligases which ubiquitinate and degrade cell surface MHC I via the endolysosomal pathway. We recently developed non-biased methods to identify cell surface receptors up- or downregulated by viruses. Using SILAC-based proteomics we can compare the relative abundance of >600 plasma membrane proteins, and identify those receptors whose expression is altered by viruses. We have identified novel amino acid transporters regulated by HIV, and drug transporters regulated by HCMV. Defining the mechanism of action of these novel receptors: Using genetic, biochemical and cell biological approaches we then determine how newly identified cell surface proteins are compromised by intracellular pathogens. For example, our siRNA screens identified novel E3 ligases, such as TRC8, involved in receptor regulation as well as their physiological substrates. We use traditional biochemical as well as mutagenesis-based genetic screens to identify the critical pathway components required for immunoreceptor regulation. Jessica Boname Marian Burr Helen Bye Florencia Cano Lidia Duncan Nick Matheson Radu Rapiteanu Suzanne Talbot Shireen Tan Richard Timms Dick van den Boomen Michael Weekes Funding: Wellcome Trust Medical Research Council Addenbrooke’s Charitable Trust Randow, F. and Lehner, P. J. (2009). Viral avoidance and exploitation of the ubiquitin system. Nat Cell Biol 11, 527–534. Stagg, H. R., Thomas, M., van den Boomen, D., Wiertz, E. H., Drabkin, H. A., Gemmill, R. M. and Lehner, P. J. (2009). The TRC8 E3 Ligase ubiquitinates MHC class I molecules before dislocation from the ER. J Cell Biol 186, 685–692. Burr, M. L., Cano, F., Svobodova, S., Boyle, L. H., Boname, J. M. and Lehner, P. J. (2011). HRD1 and UBE2J1 target misfolded MHC class I heavy chains for endoplasmic reticulum-associated degradation. Proc Natl Acad Sci USA 108, 2034–2039. By downregulating critical cell surface receptors, CMV prevents infected dendritic cells from trafficking to draining lymph nodes. 17 David Lomas Jenny Dickens Annelyse Duvoix Sarah Faull Imran Haq James Irving Tim Newton Adriana Ordonez Benoit Roussel Lu Tan Alison Warrington Funding: Medical Research Council EPSRC Wellcome Trust Alzheimer’s Research Trust Diabetes UK GlaxoSmithKline BBSRC British Lung Foundation NIHR Cambridge Biomedical Research Centre Children’s Liver Disease Foundation Protein Misfolding, "1-Antitrypsin Deficiency and the Serpinopathies One in twenty-five of the Northern European population carry the Z allele of "1-antitrypsin (342Glu Lys). Homozygotes for this mutation retain "1-antitrypsin within hepatocytes as inclusion bodies that are associated with neonatal hepatitis, juvenile cirrhosis and hepatocellular carcinoma. We have shown that Z "1-antitrypsin is retained within hepatocytes by the formation of ordered polymers in which the reactive centre loop of one molecule inserts into #-sheet A of another. Alpha-1-antitrypsin is a member of the serine protease inhibitor or serpin superfamily of proteins. Mutants of other serpins, antithrombin, C1-inhibitor and "1-antichymotrypsin also form polymers in association with plasma deficiency that causes thrombosis, angioedema and emphysema respectively. Perhaps most striking is our description of the same process in a neurone specific serpin, neuroserpin in association with an inclusion body dementia that we have called familial encephalopathy with neuroserpin inclusion bodies (FENIB). In view of their common Miranda, E. et al. (2010). A novel monoclonal antibody to characterize pathogenic polymers in liver disease associated with "1-antitrypsin deficiency. Hepatol 52, 1078–1088. Ekeowa, U. I. et al. (2010). Defining the mechanism of polymerization in the serpinopathies. Proc Natl Acad Sci USA 107, 17146–17151. Yusa, K. et al. (2011). Targeted gene correction of "1-antitrypsin deficiency in induced pluripotent stem cells. Nature 478, 391–394. 18 Immunostaining of polymeric "1-antitrypsin (green) or all forms of "1-antitrypsin (red) in human iPS cell–derived hepatocytes from individuals with "1antitrypsin deficiency and controls (from Rashid et al, J Clin Invest 2010). mechanism we have grouped these conditions into a new class of disease that we have called the serpinopathies. The structure and significance of the pathological loop-sheet polymers has been defined using biochemical, biophysical, crystallographic, and cell biology studies and with monoclonal antibodies and animal models of disease. We are now using this information to undertake small molecule screens to identify compounds that can bind to, and prevent the polymerisation of, mutant "1-antitrypsin in vitro and in vivo. More recently, we have used human induced pluripotential stem cells derived from skin fibroblasts to develop hepatocyte-like cells that recapitulate the intracellular polymers that characterise "1-antitrypsin deficiency. We then used zinc finger nuclease technology to correct the genetic defect and so produce hepatocytelike cells that secrete wild-type "1-antitrypsin. This takes us closer to a cell based therapy for "1-antitrypsin deficiency and the serpinopathies. Molecular Cell Biology of Post-Golgi Membrane Traffic Pathways Lysosomes are small membrane bound organelles ~0.5$m diameter, which are full of proteases and other hydrolytic enzymes as well as internal membranes. They function late in the endocytic pathway that takes up macromolecules from the cell surface, by fusing with endosomes, but also play a key role in phagocytosis, autophagocytosis and probably cell surface membrane repair, the latter by fusing with the plasma membrane. The late endosomes that fuse with lysosomes are observed as MVBs (multivesicular bodies) in the electron microscope and sorting of membrane proteins into the lumenal vesicles of these MVBs is mediated by ESCRT (endosomal sorting complex required for transport) proteins. Our main focus is on how cells achieve sorting and delivery of endocytosed macromolecules to lysosomes. A key protein in fusion events involving late endocytic organelles is the membrane protein VAMP7. This is targeted to the late endocytic pathway through interactions with the clathrin adaptor AP-3 and also by binding to the clathrin adaptor Hrb, that ensures its retrieval from the plasma membrane. The function of VAMP7 is regulated by interaction with Varp (Vps9 and ankyrin repeat containing protein). We are also studying the role of different protein machineries including the ESCRT proteins and HOPS (homotypic fusion and vacuole protein sorting) proteins in preparing endosomes for fusion with lysosomes. Our structural studies are in collaboration with David Owen and we also collaborate with Paul Lehner on the intracellular sorting of Class I MHC molecules following down-regulation from the cell surface by viruses. Our group maintains an active interest in diseases of membrane traffic, including the work of Rhys Roberts on Charcot-Marie-Tooth Disease. Paul Luzio Nicholas Bright Sally Gray Geoff Hesketh Rhys Roberts Jennifer Warren Lena Wartosch Funding: Medical Research Council Wellcome Trust Canadian Institutes of Health Research EMBO Pryor, P. R., Parker, L., Gray, S. R., Thompson, A., Sanderson, C. M., Evans, P. R., Owen, D. J.* and Luzio, J. P.* (2008). Molecular basis for sorting of the SNARE VAMP7 into endocytic clathrincoated vesicles by the ArfGAP Hrb. Cell 134, 817–827. Roberts, R. C., Peden, A. A., Buss, F., Bright, N. A., Latouche, M., Reilly, M. M., KendrickJones, J. and Luzio. J.P. (2010). Mistargeting of SH3TC2 away from the recycling endosome causes Charcot-Marie-Tooth Disease Type 4C. Hum Mol Genet 19, 1009–1018. Lawrence, S. P., Bright, N. A., Luzio, J. P.* and Bowers, K.* (2010). The sodium/proton exchanger NHE8 regulates late endosomal morphology and function. Mol Biol Cell 21, 3540–3551. *equal last authors Electron microscopic tomography of a high pressure frozen HeLa cell showing fusion pores between late endocytic organelles. A, a micrograph from the reconstructed volume data set showing a plane of section containing a fusion pore. B, the reconstructed 3D volume showing a second fusion pore. C, a rendered surface model of the 3D data set revealing the fusion pores ‘en face’. 19 Stefan Marciniak Hanna Clarke Lucy Dalton Jenny Dickens Elke Malzer Sally Thomas Funding: Medical Research Council Diabetes UK British Lung Foundation June Hancock Mesothelioma Research Fund The Role of Endoplasmic Reticulum Stress in Disease Proteins destined for secretion or for insertion into the cell membrane are first folded within the endoplasmic reticulum. The process of protein folding can become defective in many pathological states such as hypoxia, malignancy and some forms of diabetes. When the level of misfolded proteins within the endoplasmic reticulum increases, the cell is said to experience “endoplasmic reticulum stress”. We wish to understand the cellular consequences of endoplasmic reticulum stress, in particular its effects on tissue growth and cell survival. In doing so we hope to identify targets for the development of novel therapies. During endoplasmic reticulum stress, protein biosynthesis is initially inhibited by phosphorylation of the translation initiation factor eIF2" by the stress-sensing kinase PERK. Subsequent dephosphorylation of eIF2" by the phosphatase GADD34 restores protein translation thereafter. We discovered that this recovery of translation could itself contribute to the toxic effects of endoplasmic reticulum stress. This raises the exciting possibility that modulation of eIF2" phosphorylation may provide a useful target for the development of novel drugs to protect tissues from cell death. Cellular stresses frequently impair cell cycle progression, which can impair tissue growth. Using mammalian cell biology and Drosophila genetics we recently described a novel G2 cell cycle check point initiated by translation attenuation during endoplasmic reticulum stress. This too provides potential targets for the development of new therapies. Malzer, E., Daly, M. L., Moloney, A., Sendall, T. J., Thomas, S. L., Ryder, E., Ryoo, H. D., Crowther, D. C., Lomas, D. A. and Marciniak, S. J. (2010). Impaired tissue growth is mediated by checkpoint kinase 1 (CHK1) in the integrated stress response. J Cell Sci 123, 2892–2900. Marciniak, S. J. and Ron, D. (2010). The unfolded protein response in lung disease. Proc Am Thor Soc 7, 356–362. Yusa, K.*, Rashid, S. T.*, StrickMarchand, H., Varela, I., Liu, P.-Q., Paschon, D. E., Miranda, E., Ordóñez, A., Hannan, N., Rohani, F., Alexander, A., Marciniak, S. J., Holmes, M. C., Di Santo, J. P., Lomas, D. A.§, Bradley, A.§ and Vallier, L.§ (2011). Targeted gene correction of "1-antitrypsin deficiency in induced pluripotent stem cells. Nature 478, 391–394. joint first* and senior§ authors 20 Drosophila eye imaginal discs expressing dPERK-KR (upper panel) or dPERK-WT (lower) in mosaic clones (GFP absent). PERK is stained in blue, phosphohistone H3 in red. The Regulation of Haematopoiesis during Development and Disease Haematopoietic stem cells (HSCs) have been intensely studied for many decades as a model system for stem cell biology. Our work focuses on the emergence and regulation of the first haematopoietic stem and progenitor cells in the mouse embryo in order to identify the basic mechanisms that control their generation from precursors and their initial expansion and dissemination to the different haematopoietic organs. Knowledge of these early regulatory pathways has proven to be invaluable for understanding how adult HSCs can be manipulated for clinical purposes and how interference with these processes may result in blood-related disorders. We have recently obtained funding to specifically study how embryonic blood cells can be the target for transformation in a particularly severe type of infant leukaemia. Our research therefore complements that of several groups within the university which also work on various aspects of normal and leukaemic stem cell biology. We have recently further defined the region where HSCs are first detected and have incorporated this information into the design of a number of microarray experiments with the aim of identifying novel regulators of HSC generation in the AGM. Functional validation experiments have revealed a role for the cell cycle regulator p57Kip2 and the growth factor Igf2 in the generation, maintenance and/or migration of the first pool of HSCs. The role of other cytokines in these processes is also being investigated. Furthermore, we have also found evidence that the regulation of HSC generation is influenced by the developing sympathetic nervous system, thus providing a functional link between these two systems in the mouse embryo. We are now in the process of adding an additional level to our expression studies by identifying miRNAs that are essential for developmental haematopoiesis. Katrin Ottersbach Marie-Laure Aknin Simon Fitch Maria Mascarenhas Funding: Leukaemia & Lymphoma Research Kay Kendall Leukaemia Fund British Society for Haematology Mascarenhas, M. I., Parker, A., Dzierzak, E. and Ottersbach, K. (2009). Identification of novel regulators of hematopoietic stem cell development through refinement of stem cell localization and expression profiling. Blood 114, 4645– 4653. Ottersbach, K., Smith, A., Wood, A. and Göttgens, B. (2010). Ontogeny of Haematopoiesis: Recent Advances and Open Questions. Br J Haematol 148, 343–355. Haematopoietic stem cells are located in the middle region of the midgestation dorsal aorta. Robin, C.*, Ottersbach, K.*, Boisset, J.-C., Oziemlak, A. and Dzierzak, E. (2011). CD41 is developmentally regulated and differentially expressed on mouse hematopoietic stem cells. Blood 117, 5088–5091. *equal contribution 21 David Owen Stephen Graham† Bernard Kelly Lauren Jackson Sharon Miller Funding: Wellcome Trust Royal Commission of 1851† Jackson, L. P., Kelly, B. T., McCoy, A. J., Gaffry, T., James, L. C., Collins, B. M., Höning, S., Evans, P. R. and Owen, D. J. (2010). A large-scale conformational change couples membrane recruitment to cargo binding in the AP2 clathrin adaptor complex. Cell 141, 1220–1229. Kelly, B. T. and Owen, D. J. (2011). Endocytic sorting of transmembrane protein cargo. Curr Opin Cell Biol 23, 404–412. Structural Biology of Transport Vesicle and Organelle Biogenesis Transmembrane proteins are moved between organelles in transport vesicles. Once cargo has been sorted into a forming vesicle, the vesicle buds from the donor membrane and is then transported to and fuses with the target membrane. The protein coats that surround transport vesicle possess both a self-assembly function, which results in polymeric lattice formation and a cargo recognition function. The latter is mediated by the direct binding of coat components to determinants in the cytosolic portions of trans-membrane cargo, most commonly used being short linear motifs. In clathrin-coated vesicles (CCVs) the most commonly used motifs are recognised by clathrin adaptors : Yxx% and ExxxLL recognised by AP complexes, FxNPxY recognised by PTB containing proteins and poly-ubiquitination modifications recognised by epsins and GGAs. SNAREs are membraneembedded proteins, which provide specificity and energy to transport vesicle:organelle fusion events. Appropriate SNAREs must also be actively sorted into transport vesicles to allow the vesicles to fuse with their desired target organelle and also to return SNAREs that are required for subsequent vesicle transport events Miller, S. E., Sahlender, D., Graham, S. C., Hoening, S., Robinson, M. S., Peden, A. A.* and Owen, D. J.* (2011). The molecular basis for the endocytosis of small R-SNAREs by the clathrin adaptor CALM. Cell 147, 1118–1131. * joint corresponding authors The clathrin adaptor CALM selects endocytic SNAREs e.g. VAMP8 for packaging into endocytic CCVs to ensure that they can fuse with their target endosomes. 22 to their correct location. These recognition events, which occur in parallel with standard cargo selection, are mainly mediated by direct and highly specific recognition of the folded regions of SNAREs. In CCV-mediated transport we have elucidated the molecular mechanisms by which a variety of SNAREs (Vti1b, VAMP7, VAMPs3&8) are transported by the clathrin adaptors (epsinR, Hrb and CALM) respectively. Vesicle coat components as well as proteins that control transport vesicle:organelle and organelle:organelle fusion through SNARE regulation and membrane tethering are recruited to appropriate membranes through their binding to membrane-inserted small G-proteins and membrane phospholipids as well as by their interactions with various transmembrane proteins. In collaboration with various groups both within and outside the CIMR we are taking an integrated structural/functional approach to studying such interactions from a number of transport vesicle and organelle biogenesis events. Familial Intellectual Disability: From Genes to Disease We aim to understand how genetic abnormalities cause severe intellectual impairment. We have recruited >1000 families with moderate to severe intellectual disability in the iGOLD Genetic of Learning Disability Study and have identified many genes on the X chromosome that cause disease using an exome sequencing and detailed array comparative genomic hybridization approach (aCGH). We are now taking a whole exome approach and also testing the contribution of non-coding variants to disease. We are continuing to quantify the genetic contribution to disease by testing whether genetic abnormalities make a significant impact on the poor intellectual outcome of babies born less than 24 weeks gestation, or who are stillborn or result in neonatal death. Current estimates are that defects in >1500 genes are sufficient to cause severe cognitive impairment and the future challenge is to Lucy Raymond understand the unifying pathways and processes that are disrupted that cause disease. We are working on defining in detail the clinical consequences of mutations in patients and to understand the phenotypic effects of loss of function mutations in neuronal cell models of disease. In collaboration with Professor Paul Fletcher, Wellcome Trust Senior Clinical Fellow, we are interested in using functional MRI to define subtle abnormalities of brain morphology and in collaboration with Dr Roger Barker, Department of Neuroscience, we are working on human fetal brain tissue to understand the role of disease genes within human primary neurons. Our ultimate aim is to be able to translate these findings into better diagnostic tools for families with intellectual disability and to consider unifying cellular phenotypes that can be used in developing therapeutic agents that will ameliorate the disease. Elaine Baker Kate Baker Peng Gong Aoife Murray Funding: Medical Research Council Action Medical Research Baily Thomas Foundation NIHR Cambridge Biomedical Research Centre Addenbrooke’s Charitable Trust Academy of Medical Sciences Tarpey, P. S. et al. (2009). A systematic, large-scale resequencing screen of X-chromosome coding exons in mental retardation. Nat Genet 41, 535–543. Noor, A. et al. (2010). Disruption at the PTCHD1 Locus on Xp22.11 in Autism spectrum disorder and intellectual disability. Sci Transl Med 2, 49–68. A 6kb deletion upstream of the CUL4B transcripts segregated with disease and resulted in loss of CUL4B expression in affected individuals. Whibley, A. C., Plagnol, V., Tarpey, P. S., Abidi, F., Fullston, T., Choma, M. K., Boucher, C. A., Shepherd, L., Willatt, L., Parkin, G., Smith, R., Futreal, P. A., Shaw, M., Boyle, J., Licata, A., Skinner, C., Stevenson, R. E., Turner, G., Field, M., Hackett, A., Schwartz, C. E., Gecz, J., Stratton, M. R. and Raymond, F. L. (2010). Finescale survey of X chromosome copy number variants and indels underlying intellectual disability. Am J Hum Genet 87, 173–88. 23 Randy Read Gábor Bunkóczi Wee Lee Chan Mykhaylo Demydchuk Airlie McCoy Robert Oeffner Yahui Yan Aiwu Zhou Funding: Wellcome Trust National Institutes of Health (USA) Zhou, A., Carrell, R. W., Murphy, M. P., Wei, Z., Yan, Y., Stanley, P. L. D., Stein, P. E., Broughton Pipkin, F. and Read, R. J. (2010). A redox switch in angiotensinogen modulates angiotensin release. Nature 468, 108–111. Protein Crystallography and Pathogenesis Research in my group is in the field of protein crystallography. Crystallography is the primary method for determining the three-dimensional structure of a protein, which provides an essential framework for a detailed understanding of its biochemistry. We work both on extending the scope and power of the methods used in protein crystallography, and on applying those methods to determine new protein structures. In choosing what to study we focus on proteins involved in pathogenesis and disease, the structures of which can be exploited in the development of new therapies. One focus of recent structural work has been on members of the serpin family, most of which undergo an extraordinary conformational change on cleavage by proteases. The structures of two hormone-binding globulins show how they harness this conformational change to deliver thyroxine and cortisol to their sites of action. Our work on angiotensinogen, the source of the hormone angiotensin, has shed new light on how blood pressure is modulated. We also DiMaio, F., Terwilliger, T. C., Read, R. J., Wlodawer, A., Oberdorfer, G., Wagner, U., Valkov, E., Alon, A., Fass, D., Axelrod, H. L., Das, D., Vorobiev, S. M., Iwaï, H., Pokkuluri, P. R. and Baker, D. (2011). Improving molecular replacement by densityand energy-guided protein structure optimization. Nature 473, 540–543. Deane, J. E., Graham, S. C., Kim, N. N., Stein, P. E., McNair, R., Cachón-González, M. B., Cox, T. M. and Read, R. J. (2011). Insights into Krabbe disease from structures of galactocerebrosidase. Proc Natl Acad Sci USA 108, 15169– 15173. Schematic illustration of the methods (including our program Phaser) used to solve the crystal structure of angiotensinogen, by averaging electron density over multiple crystal forms. 24 have an interest in enzymes mutated in inherited metabolic diseases. The structure of galactocerebrosidase is helping to understand how mutations in this enzyme lead to Krabbe disease. In crystallographic theory, we focus on the understanding of probability distributions relating the structure factors that arise from the diffraction experiment. A detailed understanding of these probability distributions underlies new developments in maximum likelihood methods, which we are implementing in our program Phaser. The current version of Phaser can solve structures by molecular replacement (i.e. using the known structures of related proteins), by using the information from single-wavelength anomalous diffraction (SAD), and by a combination of the two. In a collaboration with the developers of the modelling program Rosetta, we have been excited to find that the combination of molecular replacement and advanced modelling can solve structures that elude previous methods. Molecular Cellular Pathology of Axonal Degeneration Our research is focused on the hereditary spastic paraplegias (HSPs), genetic conditions in which there is a distal degeneration of the longest axons of the motor pathway in the spinal cord. These conditions provide an excellent opportunity to understand molecular mechanisms crucial for axonal function, potentially of relevance to more common neurological diseases. We want to understand the normal functions of HSP proteins and how these functions are disrupted to cause axonopathy. Many of the disease proteins function in membrane traffic processes, especially at the endoplasmic reticulum and at endosomes. Our work concentrates on understanding the functions of this membrane traffic subgroup of HSP proteins and is based on several related themes: 1. Understanding the functions of spastin. This is crucial to understanding the pathogenesis of HSP, since mutations in the spastin gene are the most frequent cause of the disease, and since the spastin protein is at the hub of an interacting network of HSP proteins. Spastin is a microtubule severing protein and our work has shown that it is recruited to membrane traffic sites, where it couples microtubule regulation to membrane modelling processes. Spastin functions at the early secretory pathway, where it is involved in endoplasmic reticulum morphogenesis. It also functions at endosomes. We are unravelling the role of spastin at these locations, using a variety of cell biological and in vivo approaches. 2. Understanding the role of HSP proteins in BMP signalling. We are especially interested to determine whether a group of HSP proteins cause HSP by disrupting Bone Morphogenetic Protein (BMP) signalling in the axon, by affecting membrane traffic of BMP receptors. If so, targeting this pathway could open the possibility of new therapeutic approaches for the HSPs. Evan Reid Rachel Allison Virginia Clowes James Connell Caroline Freeman James Gingell Funding: Wellcome Trust Medical Research Council Tsang, H. T. H., Edwards, T. L., Wang, X., Connell, J. W., Davies, R. J., Durrington, H. J., O’Kane, C. J., Luzio, J. P. and Reid, E. (2009). The hereditary spastic paraplegia proteins NIPA1, spastin and spartin are inhibitors of mammalian BMP signaling. Hum Mol Genet 18, 3805–3821. Blackstone, C., O’Kane, C. J. and Reid, E. (2011). Hereditary Spastic Paraplegias: membrane traffic and the motor pathway. Nat Rev Neurosci 12, 31–42. Montenegro, G., Rebelo, A., Connell, J., Allison, R., Babalini, C., Schle, R., Ishiura, H., D’Aloia, M., Montieri, P., Price, J., Strickland, A., Gonzalez, M., Baumbach, L., Deconinck, T., Huang, J., Bernardi, G., Vance, J., Rogers, M. T., Tsuji, S., de Jonghe, P., Pericak-Vance, M., Schöls, L., Orlacchio, A., Reid, E.*, Züchner, S. (2012). Mutations in the ER-shaping protein Reticulon2 cause the axonal degenerative disorder Hereditary Spastic Paraplegia type 12. J Clin Invest (in press). * joint corresponding and senior author The microtubule severing enzyme spastin regulates endosomal tubulation. Image shows increased tubulation of an endosomal compartment (labelled with SNX1) in a HeLa cell depleted of spastin by siRNA knockdown. 25 Frank Reimann Eleftheria Diakogiannaki Catherine Moss Funding: Wellcome Trust European Foundation for the Study of Diabetes BBSRC/AstraZeneca European Union FP7 Full4Health Reimann, F., Cox, J. J., Belfer, I., Diatchenko, L., Zaykin, D. V., McHale, D. P., Drenth, J. P. H., Dai, F., Wheeler, J., Sanders, F., Wood, L., Wu, T. X., Karppinen, J., Nikolajsen, L., Männikkö, M., Mitchell, B. M., Kiselycznyk, C., Poddar, M., te Morsche, R. H. M., Smith, S., Gibson, D., Maixner, W., Gribble, F. M. and Woods, C. G. (2010). Pain perception in common painful conditions is altered by a nucleotide polymorphism in SCN9A. Proc Natl Acad Sci USA 107, 5148–5153. Stimulus Secretion Coupling in Enteroendocrine Cells After a meal a range of hormones is released from specialised intestinal cells, the enteroendocrine cells, which coordinate the body’s response to the availability of nutrients. As enteroendocrine cells lie scattered throughout the intestinal epithelium we have made a number of transgenic mice expressing marker proteins under the control of specific hormone promoters to aid their identification and manipulation. Tagging with fluorescent markers allows the purification of specific enteroendocrine subpopulations for expression analysis. Surprisingly, we have found cells expressing a number of hormones concomitantly, challenging the traditional classification of enteroendocrine cells based on cell morphology and limited immunohistochemical characterisation. Fluorescent markers also allow the identification of enteroendocrine cells in mixed epithelial cultures, enabling the use of single-cell techniques, like patch-clamping and live cell fluorescent imaging, to study stimulussecretion coupling mechanisms. Whilst we have, in collaboration with Dr Fiona Gribble’s lab, identified electrogenic sodium-coupled glucose uptake via SGLT-1 as a major contributor to Friedlander, R. S., Moss, C. E., Mace, J., Parker, H. E., Tolhurst, G., Habib, A. M., Wachten, S., Cooper, D. M., Gribble, F. M. and Reimann, F. (2011). Role of phosphodiesterase and adenylate cyclase isozymes in murine colonic glucagon like peptide 1 secreting cells. Br J Pharmacol 163, 261–271. Rogers, G. J., Tolhurst, G., Ramzan, A., Habib, A. M., Parker, H. E., Gribble, F. M. and Reimann, F. (2011). Electrical activity-triggered Glucagon-Like Peptide-1 secretion from primary murine L-cells. J Physiol 589, 1081–1093. L-cells expressing YFP are immuno-stained against different gut hormones (red fluorescence) and analysed by FACS. Overlap between cell types is shown by proportions of YFPpositive cells (dark) that are immunopositive (light). 26 carbohydrate sensing, we are less sure about the lipid sensing machinery of enteroendocrine cells. Ingested lipids are, however, at least as good a stimulus as glucose for the release of glucagon-like peptide-1 (GLP-1) and glucosedependent insulinotropic polypeptide (GIP), two hormones which between them are believed to account for the stimulation of >50% of insulin released after a meal. One current focus of the lab is thus the investigation of potential lipid sensing mechanisms of GLP-1 and GIP releasing cells with the aim to identify potential targets to manipulate hormone release in the treatment of type 2 diabetes and obesity. A second interest of our group is in the understanding of electrophysiological mechanisms underlying pain perception. In collaboration with the group of Professor Geoff Woods, we characterise mutations affecting channel genes, as exemplified by the voltage-gated sodium channel gene SCN9A, mutations of which can give rise to hypersensitivity or complete obliviousness to painful stimuli. Margaret Robinson Coated Vesicle Adaptors Proteins are transported between the various organelles of the cell by vesicles, which bud from one membrane and fuse with another. The formation of these vesicles and the selection of the right sort of cargo are dependent on coat proteins. Several types of coated vesicles have been described, the best characterised of which are the clathrin-coated vesicles (CCVs). The coats on CCVs consist of two major components, clathrin and adaptor protein (AP) complexes, as well as other less abundant proteins. To look for novel adaptors and other components of the CCV machinery, we are using two approaches. One is a proteomics analysis of CCVs, both to establish the protein composition of CCVs and to gain insights into the functional relationships between the various components by carrying out “CCV profiling”. The second approach is to screen a human genome-wide library for siRNAs that disrupt clathrin-mediated trafficking. Top hits include several subunits of the vacuolar ATPase, and we are investigating the role of V-ATPase in CCV formation. We are also making use of a novel system that we developed, called a knocksideways, to investigate the consequences of rapidly inactivating CCV machinery in cultured cells, and we are in the process of generating a knocksideways mouse model. Although for over 10 years it has been assumed that there are four AP complexes, we recently discovered a fifth AP complex. We are currently characterizing the AP-5 pathway, and how mutations affecting this pathway might lead to hereditary spastic paraplegia. In addition, we are investigating how the HIV-1-encoded protein Nef hijacks adaptor-mediated trafficking to down regulate the cell surface proteins MHC Class I and CD4. Georg Borner Maja Choma Jennifer Hirst Nicola Hodson Patrycja Kozik Daniela Sahlender Funding: Wellcome Trust Medical Research Council Hirst, J., Sahlender, D. A., Choma, M., Sinka, R., Harbour, M. E., Parkinson, M. and Robinson, M. S. (2009). Spatial and functional relationship of GGAs and AP-1 in Drosophila and HeLa cells. Traffic 10, 1696–1710. Robinson, M. S.*, Sahlender, D. A. and Foster, S. D. (2010). Rapid inactivation of proteins by rapamycin-induced rerouting to mitochondria. Dev Cell 18, 324–331. *corresponding author Hirst, J., Francisco, G. C., Sahlender, D. A., Seaman, M. N. J., Dacks, J. B. and Robinson, M. S. (2011). The fifth adaptor protein complex. PLoS Biol 9, e1001170. The “knocksideways” system reroutes much of the AP-2 adaptor complex (red, left) to mitochondria (green) just 6 seconds after adding a small molecule (red, middle). 27 David Rubinsztein Carla Bento Jenny Carmichael Angeleen Fleming Moises Garcia-Arencibia Zeyn Green-Thompson Warren Hochfeld Maike Lichtenberg Shouqing Luo Dunecan Massey Fiona Menzies Kevin Moreau Claudia Puri Marie Quick Maurizio Renna Oana Sadiq Maria Sanchez Farah Siddiqi Frances Thomson Pearl Toh Eszter Zavodszky Aggregate-prone Intracellular Proteins and Disease – from Biology to Therapeutic Strategies Intracellular protein aggregation is a feature of many late-onset neurodegenerative diseases, including Huntington’s disease (HD) and the muscle disease oculopharyngeal muscular dystrophy (OPMD). Most of these mutant proteins cause disease via toxic gain-of-function mechanisms. Therefore, the factors regulating their clearance are crucial for understanding disease pathogenesis and for developing rational therapeutic strategies. The two major intracellular protein degradation pathways are the ubiquitin-proteasome system and (macro)autophagy. David Rubinsztein’s laboratory studies the relevance of autophagy in neurodegenerative disease, and the basic cell biology of this important catabolic process. He aims to further the understanding of the pathogenesis of Huntington’s disease and oculopharyngeal muscular dystrophy, and to develop rational therapeutic strategies for these conditions. He has identified FDA-approved drugs that may have benefit in both diseases, since he has demonstrated their efficacies and protective mechanisms in a range of cell and animal models. Over the last two years, the highlights of his work include: 1. Characterising early steps in autophagosome biogenesis, and showing that the plasma membrane contributes to autophagosome precursors. 2. Showing that lysosomal positioning affects both autophagosome biogenesis and autophagosome-lysosome fusion, and that this is a strategy that can influence toxicity of aggregate prone proteins that cause diseases like Huntington’s disease. 3. Demonstrating that mutations that cause forms of Parkinson’s disease and the degenerative epilepsy, Lafora disease, inhibit autophagy. 4. Showing that nitric oxide accumulation, which occurs in many neurodegenerative diseases, impairs autophagy, and that inhibition of nitric oxide formation alleviates the aggregation and toxicity of mutant huntingtin in cell culture and in vivo. 5. Demonstrating that cystamine alleviates the toxicity of the mutation causing OPMD in cells and in mice. Funding: Wellcome Trust Medical Research Council NIHR Cambridge Biomedical Research Centre European Union FP7 BBSRC/Lilly CASE Studentship Tau Consortium Korolchuk, V. I., Saiki, S., Lichtenberg, M., Siddiqi, F. H., Roberts, E. A., Imarisio, S., Jahreiss, L., Sarkar, S., Futter, M., Menzies, F. M., O’Kane, C. J., Deretic, V. and Rubinsztein, D. C. (2011). Lysosomal positioning coordinates cellular nutrient responses. Nat Cell Biol 13, 453–460. 28 Sarkar, S., Korolchuk, V. I., Renna, M., Imarisio, S., Fleming, A., Williams, A., Garcia-Arencibia, M., Rose, C., Luo, S., Underwood, B. R., Kroemer, G., O’Kane, C. J. and Rubinsztein, D. C. (2011). Complex inhibitory effects of nitric oxide on autophagy. Mol Cell 43, 19–32. Moreau, K., Ravikumar, B., Renna, M., Puri, C. and Rubinsztein, D. C. (2011). Autophagosome precursor maturation requires homotypic fusion. Cell 146, 303–317. Clathrin-coated pit labelled against Atg16L1, a protein associated with autophagosome precursors (15 nm gold), and clathrin (10 nm gold). See Nat Cell Biol (2010) 12, 747–757. T-cell signalling and immune function Professor Rudd’s research focuses on deciphering the signal transduction pathways in T-cells, and their connection to immune responses and various immune disorders. My laboratory was the first to discover the CD4 and CD8-p56lck (lck) complexes, initiators of the phosphorylation and activation cascade in T-cells. We are presently interested in the identity of downstream substrates, in particular, complexes responsible for the ‘inside-out’ pathway for adhesion and the movement of T-cells in lymph nodes. These events are needed for T-cell migration to sites of inflammation and interaction with antigen presenting cells. Two mediators, adhesion- and degranulation-promoting adapter protein (ADAP) and src kinase-associated phosphoprotein 1 (SKAP1) control LFA-1 activation, while SKAP1 binding to RapL controls the ‘slowing’ of T-cells in lymph nodes for interactions with dendritic cells. ADAP and SKAP1 also regulate the NF&b and ERK pathways as well as infection by the human immunodeficiency virus (HIV-1). We are interested in the targeting of these pathways in Christopher Rudd the development of new drugs for the treatment of infection and cancer. We are also interested in the signalling events responsible for the function of co-receptors such as CD28, ICOS, CTLA-4 and PD-1. These co-receptors qualitatively change immune responses, and have been successfully targeted in the treatment of auto-immune disorders and cancer. We identified CD28 binding to the adaptor Grb2 in the activation of the transcription factor NF&B, while the CD28 binding to PI 3K activates glycogen synthase kinase (GSK3) for optimal cytolytic T-cell (CTL) function. We have also proposed a new model ‘reverse stop signal model’ to explain CTLA-4 dampening of immune responses and protection against the development of autoimmunity. CTLA-4 activates T-cell motility and limits T-cell-dendritic cell interactions. Our laboratory uses a variety of approaches from basic biochemistry to cellular immunology and the generation of models of immunodeficiency and infection. Kittiphat Chanthong Karsten Koehler Christian Ku Daina Lim Hebin Liu Yuning Lu Asha Recino Pavel Riha Helga Schneider Xin Smith Alison Taylor Raj Thaker Funding: Wellcome Trust Rudd, C. E. (2008). The reverse stopsignal model for CTLA4 function. Nat Rev Immunol 8, 153–160. Raab, M., Smith, X., Wang, H., Lu, Y., Wu, Y., Strebhardt, K., Ladbury, J.E. and Rudd, C.E. (2010). T cell receptor “Inside-Out” pathway via signaling module SKAP1RapL regulates T cell motility and interactions in lymph nodes. Immunity 32, 541–556. Purbhoo, M. A, Liu, H., Oddos, S., Owen, D. M., Neil, M. A. A., Pageon, S., French, P. M., Rudd, C. E. and Davis, D. M. (2010). Dynamics of sub-synaptic vesicles and surface microclusters at the immunological synapse. Science (Signal) 3, ra36. Upper panel: T-cell receptor signalling pathway for activation of LFA-1 on T-cells. Lower panel: motifs within the cytoplasmic tail of CD28 bind various signalling proteins. 29 Peter St George-Hyslop Katie Cox Roger Dodd Yi Li Stephen Lu Beth McDonald William Meadows Seema Qamar Molecular Mechanisms of Neurodegeneration My laboratory focuses on understanding the molecular mechanisms underlying neurodegenerative diseases, using molecular genetics, animal modelling and biophysical methods. The presenilin complex performs the intra-membranous cleavage of the amyloid precursor protein to generate the neurotoxic, aggregation-prone Abeta peptide. Using a series of biophysical tools including negative stain electromicroscopy, mass spectrometry, FLIM-FRET and analytical ultracentrification, we have generated a model of the presenilin complex and demonstrated how the binding of small molecule inhibitors and substrates induces a conformational change in the complex. Ongoing work now seeks to improve the resolution of this model. In a separate series of experiments we have also investigated how mutations in the FUS gene cause motor neuron disease, we have generated a model in C.elegans. In this model, mutations cause paralysis, the severity of which directly mimics the severity of the human disease associated with that mutation. We have shown that this arises because of aggregation of the mutant FUS protein, and preliminary evidence suggests that the disease phenotype can be reversed by small molecule inhibitors of aggregation. Funding: Wellcome Trust Medical Research Council National Institute for Health Research Howard Hughes Medical Institute Ontario Research Fund Naj, A. C. et al. (2011). Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer's disease. Nat Genet 43, 436–441. Höglinger, G. U. et al. (2011). Identification of common variants influencing risk of the tauopathy progressive supranuclear palsy. Nat Genet 43, 699–705. Murakami, T. et al. (2011). ALS mutations in FUS causes neuronal dysfunction and death in C. elegans by a dominant gainof-function mechanism. Hum Mol Genet (epub). View from extracellular surface of 3D reconstruction of the presenilin complex in the absence of ligand/substrate (left) and in the presence of ligand/substrate (right). 30 The Molecular Mechanisms of Endosome-toGolgi Retrieval Protein localization within eukaryotic cells is essential for normal cellular physiology. The mechanisms that govern the localization of membrane proteins within the post-Golgi endocytic system control diverse processes such as; nutrient uptake, signaling receptor downregulation, synaptic transmission, the response to infection and generation of immunity. There is a growing awareness and appreciation of the importance of correct endosomal protein sorting in various neurodegenerative disorders. In my lab we study the functioning of the endosome-to-Golgi retrieval pathway and have extensively characterized the role of the retromer complex in that pathway. Retromer is a conserved protein complex that localizes to endosomes to sort membrane proteins into nascent tubules. The retromer complex can be functionally dissected into two subcomplexes; a cargo-binding complex comprising a heterotrimer of VPS35VPS29-VPS26 proteins and the sorting nexin dimer of Snx1 or Snx2 with Snx5 and Snx6. We have recently demonstrated the role that the small GTPase, Rab7a, plays in regulating the membrane association of the cargo-selective retromer complex. In addition to Rab7a, the TBC1D5 protein also interacts with the cargoselective retromer complex by direct binding to VPS29. As a member of the Rab GTPase activating protein (GAP) family, TBC1D5 operates antagonistically to Rab7a. Therefore, Rab7a and TBC1D5 ensure that the cargoselective retromer complex mediates the sorting of membrane proteins through a dynamic cycle of association and dissociation of the complex with the membrane. Attempts to identify proteins that interact with the retromer complex and contribute to the efficiency of endosomal protein sorting are on-going and comprise a number of complementary techniques including; native immunprecipitation to identify proteins associated with retromer and functional genomics using high-throughput siRNA screening technologies to identify candidate genes required for endosome-to-Golgi retrieval. As a result of these efforts, we have recently reported the role of the retromer complex in mediating the recruitment of the endosomal WASH complex. One of the components of the WASH complex is a protein called Strumpellin that is mutated in Hereditary Spastic Paraplegia (HSP) – a neuronal degenerative disorder. Loss of Strumpellin function results in dysregulation of endosomal tubules suggesting that defective regulation of endosomal membrane dynamics may contribute to the pathology of HSP. Matthew Seaman Michael Harbour Sophia Breusegem Funding: Medical Research Council Seaman, M. N. J., Harbour, M. E., Tattersall, D., Read, E. and Bright, N. (2009). Membrane recruitment of the cargoselective retromer subcomplex is catalysed by the small GTPase Rab7 and inhibited by the RabGAP TBC1D5. J Cell Sci 122, 2371–2382. Kozik, P., Francis, R. W., Seaman, M. N. J.* and Robinson, M. S.* (2010). A screen for endocytic motifs. Traffic 11, 843–855. *co-corresponding authors Harbour, M. E., Breusegem, S. Y. A., Antrobus, R., Freeman, C., Reid, E. and Seaman, M. N. J. (2010). The cargo-selective retromer complex is a recruiting hub for protein complexes that regulate endosomal tubule dynamics. J Cell Sci 123, 3703–3717. A. Schematic diagram of the retromer complex. B. Cells treated with siRNA to abolish expression of the Strumpellin protein exhibit markedly more endosomal tubules (highlighted with arrows and arrow heads). Bar = 20$m. 31 Symeon Siniossoglou Hiroshi Sembongi Funding: Medical Research Council Siniossoglou, S. (2009). Lipins, lipids and nuclear envelope structure. Traffic 10, 1181–1187. Karanasios, E., Han, G.-S., Xu, Z., Carman, G. M. and Siniossoglou S. (2010). A phosphorylationregulated amphipathic helix controls the membrane translocation and function of the yeast phosphatidate phosphatase. Proc Natl Acad Sci USA 107, 17539–17544. Linking Phospholipid Metabolism to Membrane and Organelle Function Lipids are building blocks for membranes and their regulated production during development often underlies striking morphological changes in a variety of specialized cell types. In addition, lipids act as signals by which organelles and cells communicate with each other and as energy storage molecules. Defects in lipid metabolism or signalling can lead to a large number of disorders such as metabolic syndrome or cancer. The aim of our laboratory is to understand how cells maintain lipid and membrane homeostasis during growth and development. Our current studies focus on a fundamental step in lipid metabolism, the dephosphorylation of phosphatidic acid (PA) to diacylglycerol (DAG) that is catalyzed by a conserved family of lipid phosphatases, lipins. DAG generated by lipins is used for the biosynthesis of (a) membrane phospholipids and (b) triglyceride stored in lipid droplets. Consistent with these essential Choi, H. S., Su, W. M., Morgan, J. M., Han, G.-S., Xu, Z., Karanasios, E., Siniossoglou, S. and Carman G. M. (2011). Phosphorylation of phosphatidate phosphatase regulates its membrane association and physiological functions in Saccharomyces cerevisiae. J Biol Chem 286, 1486–1498. Lipins catalyze the dephosphorylation of phosphatidic acid to diacylglycerol (left panel). Loss of lipin 1 inhibits lipid accumulation in adipocytes (upper panels, red:lipid, blue:DNA, bar: 5$m). Loss of the lipin Pah1 causes nuclear/ER membrane expansion in yeast (lower panel, arrows point to the expanding nucleus in green, bar: 1$m). 32 metabolic functions, lipins are important regulators of both organelle biogenesis and fat storage in many eukaryotic cells. Recent work from our group led to the identification of key regulators of the yeast lipin Pah1p and a model on how these cooperate to recruit Pah1p onto membranes for DAG production. We are currently expanding these studies to both mouse and human fat cells (adipocytes) that express lipin 1, 2 and 3 and investigate their roles both during adipocyte differentiation and lipogenesis. Unexpectedly, lipins also translocate into the nucleus where they regulate gene expression. We are using a combination of genetics and gene expression profiling approaches to identify the nuclear targets of lipins and uncover the function of these lipid phosphatases into the nucleus. Immune Regulation, Autoimmune Disease and Infection We aim to discover how genetically determined variation in immune regulation balances the risks of autoimmune disease and infection. Regulation of the B cell immune response is a major interest. We have investigated the function of inhibitory receptors such as FcyRIIb, which act as “brakes” on the immune system. We have shown that even subtle changes in expression of FcyRIIb on B cells can prevent or induce SLE in mice. Natural polymorphisms in FcyRIIb are associated with immune dysregulation (see Figure) and autoimmune diseases such as systemic lupus erythematosus (SLE) in both mouse and man. Some reduce function and are common in Asia and Africa, where malaria is endemic. We have used mouse models of malaria and human genetics (in collaboration with the KEMRI/ Wellcome Trust Unit in Kilifi, Kenya), and have shown that FcyRIIb deficiency can protect against severe malaria, perhaps contributing to the evolution of predisposition to SLE in some ethnic groups. We have recently discovered a regulatory T cell which specifically regulates the germinal centre and may have autoimmune implications. Other lab members are defining how abnormal control of plasma cell function and migration contributes to autoimmunity, and are studying specific micro-RNAs we have shown to control B cell activation, lymphoid development and oncogenesis. We have established a programme in human autoimmunity, working with patients with vasculitis, SLE, inflammatory bowel disease and renal transplants (and led with Paul Lyons). By performing detailed transcriptomic studies of purified white blood cells we have discovered novel biomarkers now being assessed for their ability to guide therapy to improve efficacy and reduce treatment toxicity. We are also using genetics to identify novel pathways for investigation. Examples include our initiation of the European Vasculitis Genetics Consortium which has just completed the first genome-wide association study in ANCA-associated vasculitis, and a novel analysis of the inflammatory bowel disease GWAS which has identified genetic variants associated with clinical course rather than diagnosis, and defined their functional implications in humans using the Cambridge BioResource, and in mouse models. These programmes together allow us to interrogate mouse and human biology in an integrated fashion, increasing our capacity to probe immunity and disease and to translate basic biological findings to the clinic. Naturally occurring polymorphisms in the promoter of the inhibitory Fcgr2b “knocked-in” to the control C57BL6 background (KI) lead to increased germinal centre formation compared to controls (WT). Immunohistology 8 days after immunisation. Blue: B220 (= B cell follicle), green: CD8 (= T cell zone), red: KI67 (= germinal centre). McKinney, E. F., Lyons, P. A., Carr, E. J., Hollis, J. L., Jayne, D. R., Willcocks, L. C., Koukoulaki, M., Brazma, A., Jovanovic, V., Kemeny, D. M. et al. (2010). A CD8+ T cell transcription signature predicts prognosis in autoimmune disease. Nat Med 16, 586–591. Ken Smith Daniele Biasci Susanne Bökers Menna Clatworthy Devina Divekar Marion Espeli Shaun Flint Mariana Fonseca Claire Hales Alex Hatton Lorna Jarvis Elaine Jolly James Lee Michelle Linterman Paul Lyons Rebeccah Matthews Eoin McKinney James Peters Huzefa Ratlamwala Tim Rayner Arianne Richard John Sowerby David Thomas Liz Wallin Tim Wilson Matt Zilbauer Funding: Wellcome Trust Medical Research Council British Heart Foundation NIHR Cambridge Biomedical Research Centre Lister Institute of Preventive Medicine European Union Lee, J. C., Lyons, P. A., McKinney, E. F., Sowerby, J. M., Carr, E. J., Bredin, F., Rickman, H. M., Ratlamwala, H., Hatton, A., Rayner, T. F., Parkes, M., Smith, K. G. C. (2011). Gene expression profiling of CD8+ T cells predicts prognosis in patients with Crohn disease and ulcerative colitis. J Clin Invest 121, 4170–4179. Linterman, M. A., Pierson, W., Lee, S. K., Kallies, A., Kawamoto, S., Rayner, T. F., Srivastava, M., Divekar, D. P., Beaton, L., Hogan, J. J. et al. (2011). Foxp3(+) follicular regulatory T cells control the germinal center response. Nat Med 17, 975–982. Paul Lyons 33 John Todd Xaquin Castro Dopico Jason Cooper Tony Cutler Ricardo Ferreira Joanna Howson Marcin Pekalski Corina Shtir Austin Swafford (Co-supervisor – Mike Lenardo, NIH, Bethesda, USA) Funding: Wellcome Trust Juvenile Diabetes Research Foundation International National Institutes of Health (USA) NIHR Cambridge Biomedical Research Centre Medical Research Council European Union FP7 Nejentsev, S., Walker, N., Riches, D., Egholm, M. and Todd, J. A. (2009). Rare variants of IFIH1, a gene implicated in antiviral responses, protect against type 1 diabetes. Science 324, 387–389. Dendrou, C. A., Plagnol, V., Fung, E., Yang, J. H. M., Downes, K., Cooper, J. D., Nutland, S., Coleman, G., Himsworth, M., Hardy, M., Burren, O., Healy, B., Walker, N. M., Koch, K., Ouwehand, W. H., Bradley, J. R., Wareham, N. J., Todd, J. A. and Wicker, L. S. (2009). Cell-specific protein phenotypes for the autoimmune locus IL2RA using a genotype-selectable human bioresource. Nat Genet 41, 1011–1015. Heinig, M. et al. (2010). A trans-acting locus regulates an anti-viral expression network and type 1 diabetes risk. Nature 467, 460–464. 34 Identification of Molecular and Cellular Mechanisms in Autoimmune Disease Our aim is to further characterise the molecular basis for the autoimmune inflammatory disease type 1 diabetes. We use an integrated combination of genetics, in large collections of type 1 diabetic families and case/control, statistics, genome informatics and data mining, and gene expression and immunological studies. Our major effort now is to correlate susceptibility genotypes with biomarkers and phenotypes e.g. we have correlated flow cytometric phenotypes of T lymphocytes with disease-predisposing genotypes of the IL-2RA (CD25) gene. This is a first step towards identifying disease precursors that could be used in future therapeutic studies to stratify patients. To achieve this we have helped build a local biobank of volunteers and patients in whom we can study the effects of disease-associated genotypes and environmental factors (The Cambridge BioResource: www.cambridgebioresource.org.uk). Our research efforts are part of the Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory (DIL), which includes the laboratory of Linda Wicker, as well as collaborations with the Department of Haematology (Willem Ouwehand), the Department of Paediatrics (David Dunger), and the Wellcome Trust Sanger Institute. The methylation/ demethylation of the FOXP3 gene, which determines T regulatory cell number in humans, is associated with the genotype of the autoimmunity, immune signalling gene PTPN22 The Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory The Juvenile Diabetes Research Foundation/ Wellcome Trust Diabetes and Inflammation Laboratory (JDRF/WT DIL) has two Principal Investigators and is composed of four integrated research efforts: John Todd (human genetics and immunology), Linda Wicker (mouse modelling, human immunology), Chris Wallace (statistics) and Frank Waldron-Lynch (clinical). In the last ten years under programmatic funding we have discovered and defined several mouse and human susceptibility loci for type 1 diabetes. In the last five years we have been correlating the presence of the identified susceptibility alleles with their functions to determine which molecules and pathways are underlying the pathogenesis of type 1 diabetes. Our recent results have shown how important the interleukin-2 pathway is in autoimmunity and type 1 diabetes, and we are exploring further these mechanisms and their possible implications for new, mechanism-based therapeutic strategies. A key approach is the collection of local healthy volunteers and patients (The Cambridge BioResource) who are willing to donate blood samples, with which we can study immune cell populations and activities in relation to genotypes associated with susceptibility and resistance to type 1 diabetes and autoimmune disease. The JDRF/WT DIL is centrally involved in the activities of two major international consortia, the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)-and JDRF-funded Type 1 Diabetes Genetics Consortium (T1DGC) and the Wellcome Trust Case Control Consortium (WTCCC). Immunological and gene-phenotype studies have been strengthened in the establishment of a new JDRF Centre: Diabetes Genes, Autoimmunity and Prevention (D-GAP) which supports collaborations with Mark Peakman, Tim Tree, David Dunger and Polly Bingley. Investigations of therapeutic strategies in type 1 diabetes are supported by a collaborative project funded by the European Union’s 7th Framework Programme (FP7), Natural immunomodulators as novel immunotherapies for type 1 diabetes (NAIMIT). Systems Management Vin Everett (Head) Wojciech Giel Sundeep Singh Nanuwa Sample Processing Helen Stevens (Head) Pamela Clarke Gillian Coleman Sarah Dawson Jennifer Denesha Simon Duley Simon Knights Meeta Maisuria-Armer Trupti Mistry Interaction of a human T regulatory cell and dendritic cell involving the type 1 diabetes susceptibility molecule, CTLA-4. Photograph reproduced with kind permission from Paul MacAry and Laura Esposito. Affiliates Cambridge BioResource Sarah Nutland (Co-ordinator) Kelly Beer Simon Hacking Alison Hayday Jamie Rice Rachel Simpkins Hannah Stark Clinical Research Frank Waldron-Lynch (Head) Louise Bell (Co-ordinator) Pippa Tagart Maureen Wiesner DIL Laboratory Management and Administration Judy Brown (Head) Jennifer Newton Anna Simpson Niall Taylor John Todd, Director Linda Wicker, Co-director Research Group (Todd) Xaquin Castro Dopico Jason Cooper Tony Cutler Ricardo Ferreira Joanna Howson Marcin Pekalski Corina Shtir Austin Swafford (Co-supervisor – Mike Lenardo, NIH, Bethesda, USA) Research Group (Wicker) Jan Clark Laura Esposito Sarah Howlett Dan Rainbow Kara Rainbow Research Group (Wallace) Chris Wallace Hui Guo Nikolas Pontikos Xin Yang Genotyping and Sequencing Debbie Smyth (Head) Genome Informatics Oliver Burren (Head) Premanand Achuthan Ellen Adlem Mikkel Christensen Richard Coulson Sample Informatics Matt Woodburn (Head) James Macfarlane Krishna Mohan Jakub Pawlinski Data Services Neil Walker (Head) Nigel Ovington Helen Schuilenburg Statistics Chris Wallace (Head) Jason Cooper Hui Guo Joanna Howson Nikolas Pontikos Corina Shtir Xin Yang 35 John Trowsdale Jyothi Jayaraman Wei Jiang Maki Ohashi David Rhodes James Traherne Funding: Wellcome Trust Medical Research Council Louise Boyle Genetic and Functional Relationships between Polymorphic Immune Receptors We study proteins encoded in a key set of human genes, the human Major Histocompatibility Complex (MHC). This dense cluster of polymorphic loci influences susceptibility to a large number of human diseases. These include most, if not all, autoimmune conditions, many infections and other diverse conditions, from Narcolepsy to Schizophrenia. The key products of the MHC are Class I and Class II molecules. These play a pivotal role in alerting the immune system to infection by presentation of antigenic peptides to receptors on T cells. A major part of our approach concerns the regulation and functions of these molecules. Our early work involved discovery of antigen processing molecules encoded in the MHC, such as the TAP peptide transporter and immunoproteasome subunits (J Immunol 2008 Pillars of Immunology: antigen presentation: discovery of the peptide TAP). Following on from this, we found a novel molecule, TAPBPR, which is related to the MHC-encoded TAPASIN molecule. TAPBPR appears to modulate MHC class I expression and may have a profound affect on immune recognition. Louise Boyle is leading a project to elucidate the function of TAPBPR in order to understand the role of this new player in MHC class I processing and presentation. Further information on the state of health of a cell is provided by interaction of MHC class I molecules with other receptors on Natural Killer (NK) cells. Like some MHC genes, NK receptors form an extensive, polymorphic gene family. We are particularly interested in understanding the interplay between NK receptors and MHC class I molecules, in relation to disease. Clemens Hermann Keith Porter Traherne, J. A. et al. (2010). Mechanisms of copy number variation and hybrid gene formation in the KIR immune gene complex. Hum Mol Genet 19, 737–751. Rhodes, D. A., Boyle, L. H., Boname, J. M., Lehner, P. J. and Trowsdale, J. (2010). Ubiquitination of lysine-331 by Kaposi's sarcoma-associated herpesvirus protein K5 targets HFE for lysosomal degradation. Proc Natl Acad Sci USA 107, 16240–16245. Barrow, A. D. et al. (2011). OSCAR is a collagen receptor that costimulates osteoclastogenesis in DAP12deficient humans and mice. J Clin Invest 121, 3505–3516. 36 Frequencies of unique KIR haplotypes with variable gene copy number. Inset charts: frequencies of centromeric and telomeric haplotype structures (Figure: J Traherne). Identification of Molecular and Cellular Mechanisms in Autoimmune Disease Our group, as part of the Juvenile Diabetes Research Foundation/Wellcome Trust Diabetes and Inflammation Laboratory (DIL), is focused on understanding the molecular and cellular mechanisms mediating autoimmune syndromes such as type 1 diabetes (T1D) by identifying and characterising the function of genes that contribute to disease susceptibility in both humans and nonobese diabetic (NOD) mice. Some T1D-associated genes and molecular pathways are shared by humans and mice as exemplified by the CTLA-4 and IL-2 pathways. Both CTLA-4 and IL-2 are required for the function of FOXP3+CD25+CD4+ regulatory T cells, a T cell subset that dampens the immune response and is less functional in T1D patients and NOD mice. FOXP3+CD25+CD4+ regulatory T cells are a therapeutic target in T1D and the subject of intense investigation by many laboratories, including the DIL. To link the gene variants that cause autoimmune disease with their biological effects within the immune system, we study the peripheral blood of T1D patients and healthy volunteers accessed through the Cambridge BioResource. Genotypes at T1D-causal genes are correlated with phenotypes ranging from protein expression of the disease-causing gene to parameters of immune cell activation and differentiation. One phenotype for which we have discovered a genotype correlation is the expression of CD25 on naïve CD4+ T cells (see Figure). The variant of the gene encoding CD25 that increases the likelihood of developing T1D and multiple sclerosis is correlated with an increased frequency of CD25+ naïve CD4+ T cells. In the susceptible genotype, the frequency of CD25+ naïve CD4+ T cells is influenced by both age and sex. We are currently investigating this poorly characterised subpopulation of naïve CD4+ T cells and testing the hypothesis that CD25 expression decreases the activation threshold thereby making it more likely that naïve CD4+ T cells with affinity to self antigens will differentiate into pathogenic effector cells. Linda Wicker Jan Clark Laura Esposito Sarah Howlett Dan Rainbow Kara Rainbow Funding: Wellcome Trust Juvenile Diabetes Research Foundation International National Institutes of Health (USA) Dendrou, C. A., Plagnol, V., Fung, E., Yang, J. H. M., Downes, K., Cooper, J. D., Nutland, S., Coleman, G., Himsworth, M., Hardy, M., Burren, O., Healy, B., Walker, N. M., Koch, K., Ouwehand, W. H., Bradley, J. R., Wareham, H. J., Todd, J. A. and Wicker, L. S. (2009). Cell-specific protein phenotypes for the autoimmune locus IL2RA using a genotypeselectable human bioresource. Nat Genet 41, 1011–1015. Fraser, H. I., Dendrou, C. A., Healy, B., Rainbow, D. B., Howlett, S., Smink, L. J., Gregory, S., Steward, C., Todd, J. A., Peterson, L. B. and Wicker, L. S. (2010). NOD congenic strain analysis of autoimmune diabetes reveals genetic complexity of the Idd18 locus and identifies Vav3 as a candidate gene. J Immunol 184, 5075–5084. Using multi-colour flow cytometry and blood samples from Cambridge BioResource donors, we have correlated an increased frequency of CD25+ naïve CD4+ T cells (outlined in red on the figure) with susceptibility to type 1 diabetes and multiple sclerosis. Rainbow, D. B., Moule, C., Fraser, H. I., Clark, J., Howlett, S. K., Burren, O., Christensen, M., Moody, V., Steward, C. A., Mohammed, J. P., Fusakio, M. E., Masteller, E. L., Finger, E. B., Houchins, J. P., Naf, D., Koentgen, F., Ridgway, W. M., Todd, J. A., Bluestone, J. A., Peterson, L. B., Mattner, J. and Wicker, L. S. (2011). Evidence that Cd101 is an autoimmune diabetes gene in NOD mice. J Immunol 187, 325–336. 37 Geoff Woods Ofelia Carvalho Maryam Khurshid Adeline Nicholas Funding: Wellcome Trust Pfizer, UK Birth Defects Foundation Nicholas, A. K., Khurshid, M., Désir, J., Carvalho, O. P., Cox, J. J., Thornton, G., Kausar, R., Ansar, M., Ahmad, W., Verloes, A. et al. (2010). Wdr62 is associated with the spindle pole and is mutated in human microcephaly. Nat Genet 42, 1010–1014. Bakircioglu, M., Carvalho, O. P., Khurshid, M., Cox, J. J., Tuysuz, B., Barak, T., Yilmaz, S., Caglayan, O., Dincer, A., Nicholas, A. K. et al. (2011). The essential role of centrosomal NDE1 in human cerebral cortex neurogenesis. Am J Hum Genet 88, 523–535. Sir, J.-H., Barr, A. R., Nicholas, A. K., Carvalho, O. P., Khurshid, M., Sossick, A., Reichelt, S., D'Santos, C., Woods, C. G.*, Gergely, F.* (2011). A primary microcephaly protein complex forms a ring around parental centrioles. Nat Genet 43, 1147–1153. 38 The Study of Mendelian Disorders of Neurogenesis and Pain We have two main areas of research: first, to understand early human neurogenesis by the study of families with microcephaly; and second, to find the genes causing Mendelian disorders of pain perception. humans should manifest only as a brain of reduced size is now our “big question”. Another strand of our work is the evolution of the human brain and whether the MCPH genes have contributed to the recent threefold increase in size. We have shown that “automosoal recessive primary microcephaly” (MCPH for short) is a disorder of neurogenic mitosis I the embryonic and foetal developing brain. After birth the MCPH brain is small, but architecturally normal, and that the phenotype is mental retardation – but not physical retardation. The MCPH proteins are highly expressed in the neuro-epithelium lining the interior of the brain, and from which the majority of neurones arise in foetal life. Our initial focus was to find the MCPH genes, now we are trying to find what these genes do and how perturbation of this process leads to a small human brain. As all MCPH genes to date are centrosomal components we are studying their effects on mitosis, cytokinesis, cell polarity and the centriole and centrosome cycle. We have recently found that most of the MCPH genes form a complex. Completely unexpectedly this complex is involved in daughter centriole attachment and maturation on the mother centriole – an apparently fundamental process in animal cells. Why defects of centriole maturation in We recently discovered that non-sense mutations in SCN9A, encoding a voltage gated membrane sodium channel, lead to an inability to feel any pain. Subsequently we have begun the study of a number of Mendelian disorders where too much or too little pain is felt, or analgesics are ineffective. For each we have found/are finding the causative gene and then determining its function in normal nociception. This work has the clear potential to generate new analgesic targets and is part funded by Pfizer. Using similar methodologies we study a number of other Mendelian neurodevelopmental disorders, e.g. complete sex reversal, novel recessive neurodevelopmental disorders. Whilst these conditions are rare, each will yield essential insights into normal neurodevelopment after we identify the causative gene and determination of its function(s). Magnified image of mouse E13 cerebral cortex neuroepithelium showing Wdr62 (red) in apical mitotic precursors (nuclei in blue/DAPI). Strong Wdr62 expression is seen in apical neural precursor cells undergoing mitosis with a pair of centrosomes (green) either side of a metaphase plate. Core Scientific Facilities at CIMR Mark Bowen (Confocal/Imaging) Matthew Gratian (Confocal/Imaging) Kamburapola Jayawardina (Jay) (Proteomics) Nikol Simecek (Bioinformatics/Systems) Anna Petrunkina Harrison (Flow Cytometry) (Also Associate Professor, Biomedical Engineering & Reproductive Biology, University of Veterinary Medicine, Hanover and Associate Editor in Reproduction, Fertility, Development.) The above are all core staff funded as part of CIMR’s Wellcome Trust Strategic Award 39 MRC Mitochondrial Biology Unit John Walker The MRC Mitochondrial Biology Unit evolved from the former MRC Dunn Human Nutrition Unit. Its formation by the MRC recognises growing evidence for the involvement of mitochondria and their dysfunction in an ever-increasing range of human diseases most notably in neuromuscular and neurodegenerative diseases, and even, perhaps, in the process of ageing. The Unit has three major scientific aims: s To understand the fundamental processes taking place in mitochondria. s To understand the involvement of these processes in human diseases. s To exploit knowledge of these fundamental processes for the development of new therapies to treat human diseases. Today, the Unit has ten independent research groups, studying different aspects of the mitochondrion ranging from structural and functional studies of central respiratory enzymes (John Walker, Judy Hirst and Leo Sazanov) and transport proteins (Edmund Kunji), to studies of mtDNA replication, mitochondrial genetics and biogenesis of mitochondria in relation to mitochondrial disease (Ian Holt, Michal Minczuk and Antonella Spinazzola), and to aspects of apoptosis and generation of reactive oxygen species in relation to ageing (Michael Murphy). A bioinformatics group (Alan Robinson) has developed modelling an in silico model of the mitochondrion. The goal is to develop models for the dynamics and control of the metabolic and bioenergetic pathways, and to use them to analyse various human pathologies. A proteomics group (John Walker and Ian Fearnley) is using modern mass spectrometry to characterise proteins involved in respiration, in signalling in the mitochondrion, in mtDNA replication and organelle biogenesis. Their activities are focussed on understanding the fundamental biochemical and biological processes which occur in mitochondria. Via collaborations with clinical colleagues in several countries, the Unit is building on its fundamental knowledge to try to understand how mitochondrial dysfunction leads to human disease. In addition, Unit members are engaged in collaborations with pharmaceutical companies, so as to exploit our fundamental knowledge to generate new therapies. Currently, the MRC Mitochondrial Biology Unit has a staff of 105 including 49 PhD students. Martin Montgomery prepares to carry out an X-ray diffraction experiment on a protein crystal. 40 CIMR Affiliated Principal Investigators Genetics, Genomics and Vaccine Studies in Leishmaniasis The major highlight of the research based at CIMR during 2009–2011 has been a genome-wide association study (GWAS) of visceral leishmaniasis undertaken as part of the Wellcome Trust Case Control Consortium Phase Two (WTCCC2). This involved the analysis of discovery and replication case-control samples from India, and families from north-eastern Brazil (with Shyam Sundar, India; Selma Jeronimo, Brazil; Mary Wilson, USA). The HLA-DRB1-DQA1 locus was the only region to show strong evidence of association (P=1.47x10-9 at rs9271252; 2.14x10-9 at rs9271255) in the Indian discovery dataset. Replication occurred within India (P=5.85x10-5 at rs9271255) and Brazil (P=7.41x10-6 at rs9271252); Pcombined=5.55x10-16 across three studies. The protective allele at SNP rs9271252 (and at rs9271255; r2=1) tagged three DRB1 allele groups at the two digit level: *15, *16 and *01. These findings are of relevance to our further analysis of immune responses to 12 novel vaccine candidates for leishmaniasis that we have analysed in exposed donors (recovered cases, skin-test positive asympotomatics) from Brazil and India. Congenic mice bearing the HLA DRB1 risk and protective alleles are under construction with genOway to help us further understand the mechanisms by which these HLA class II molecules control disease pathogenesis and response to vaccination. Head, Division of Genetics and Health Telethon Institute for Child Health Research 100 Roberts Road, Subiaco Western Australia, 6008 (GPO Box 855, West Perth, WA 6872) Tel: +61 (08) 9489 7910 Email: [email protected] www.ichr.uwa.edu.au/research/divisions/gh The Molecular Pathogenesis of Alzheimer’s and Related Neurodegenerative Diseases My laboratory uses Drosophila melanogaster, Caenorhabditis elegans and prokaryotic systems to study specific protein aggregates that have been implicated in proteinopathies such as Alzheimer’s disease, tauopathies, Parkinson’s disease and Familial Encephalopathy with Neuroserpin Inclusion bodies. We use invertebrate model organisms to link the biophysical properties of particular proteins, and their aggregates, with whole-organism phenotypes such as reduced longevity and locomotor abnormalities. One of the strengths of these systems is the power of the genetic toolkits that are available for studying the biological pathways that underpin disease phenotypes. Alzheimer’s disease appears to be initiated by the aggregation of a peptide called A#. We have created a fly model of Alzheimer’s disease by expressing this toxic peptide in the insects’ brains. We have sought to modify the consequent longevity and behavioural phenotypes by a range of genetic manipulations and have discovered modifiers of A# toxicity in the fly. Similarly in the worm we can compare the network of genes that mediate the toxicity of A# with the orthologous network of genes implicated by human genome wide association studies in Alzheimer’s disease. Analogous experiments have been performed on proteins involved in a range of neurodegenerative disorders. Alzheimer’s Research Trust Senior Research Fellow Department of Genetics University of Cambridge Downing Street, Cambridge CB2 3EH Tel: +44 (0) 1223 760346 Email: [email protected] www.neuroscience.cam.ac.uk/directory/profile. php?damiancrowther Jenefer Blackwell Michaela Fakiola E Nancy Miller Fakiola, M. et al. (2010). Genome wide linkage study for visceral leishmaniasis in India. PLoS One 5, e15807. Fakiola, M., et al. (2011). Genetic and functional evidence implicating DLL1 as the gene that influences susceptibility to visceral leishmaniasis at chromosome 6q27. J Infect Dis 204, 467–477. Damian Crowther Jahn, T. R. et al. (2011). Detection of early locomotor abnormalities in a Drosophila model of Alzheimer’s disease. J Neurosci Methods 197, 186–189. Liu, B. et al. (2011). Iron promotes the toxicity of amyloid beta peptide by impeding its ordered aggregation. J Biol Chem 286, 4248–4256. 41 Sadaf Farooqi Greenfield, J. R. et al. (2009). Modulation of blood pressure by central melanocortinergic pathways. N Engl J Med 360, 44–52. Bochukova, E. et al. (2010). Large, rare chromosomal deletions associated with severe early-onset obesity. Nature 463, 666–670. Sergey Nejentsev Nejentsev, S. et al. (2009). Rare variants of IFIH1, a gene implicated in antiviral responses, protect against Type 1 Diabetes. Science 324, 387–389. Curtis, J. et al. (2011). Association analysis of the LTA4H gene polymorphisms and pulmonary tuberculosis in 9,115 subjects. Tuberculosis 91, 22–25. 42 Genetic, Molecular and Physiological Basis of Human Obesity We use a number of complementary genetic strategies including whole exome sequencing to study a cohort of over 4000 patients with severe early onset obesity in collaboration with the Wellcome Trust Sanger Institute. Through these approaches we are discovering mutations in novel obesity genes whose function is studied using a number of molecular and cellular approaches. As many of these proteins modulate hypothalamic neural circuits involved in the regulation of appetite, we are developing the use of patient specific neural cell lines derived from inducible pluripotent stem cells (obtained from fibroblasts), as a model system for investigating molecular mechanisms and for drug discovery. As part of our parallel programme of translational research, we undertake physiological studies in patients with monogenic obesity syndromes to examine the role of the relevant molecules in eating behaviour, energy expenditure and peripheral metabolism. Our overall aim is to make a major contribution to the design of pharmacological, nutritional and behavioural interventions to benefit patients with severe obesity. Wellcome Trust Senior Clinical Fellow and Professor of Metabolism and Medicine University of Cambridge Metabolic Research Laboratories Level 4, Institute of Metabolic Science Box 289, Addenbrooke’s Hospital Hills Road, Cambridge CB2 0QQ www.mrl.ims.cam.ac.uk/staff/PI/Farooqi/ Genetic and Functional Mechanisms of Susceptibility to Infection Genetic variants in the human genome may confer susceptibility to or resistance from infectious diseases. To discover such variants and to understand underlying functional mechanisms we use methods of human genetics, including genome-wide association studies (GWAS) and exome sequencing, cellular models of infection as well as methods of molecular biology. We study patients with primary immunodeficiencies caused by Mendelian mutations that suffer from severe and/or recurrent infections, in particular from mycobacterial and viral infections. We also study patients that suffer from pulmonary tuberculosis, a common infectious disease and a global medical problem. Our objective is to understand biological pathways involved in protection from infection and, eventually, to find new diagnostic approaches and novel targets for therapeutic intervention. Wellcome Trust Senior Research Fellow Department of Medicine University of Cambridge Level 5, Addenbrooke's Hospital Hills Road, Cambridge CB2 0QQ Tel: +44 (0) 1223 762 605 Fax: +44 (0) 1223 336 846 http://tb.med.cam.ac.uk/ Molecular and Pathophysiological Mechanisms in Human Obesity and Insulin Resistance I have a long-standing interest in the aetiology and pathophysiology of human metabolic and endocrine disease and how such information might be used to improve diagnosis, prognostication, therapy and prevention. My two principle areas of research are obesity and insulin resistance. We use a wide range of human genetic and physiological approaches, accompanied by studies in cellular and animal disease models to better understand the biological causes of extreme forms of obesity, insulin resistance and lipodystrophy and also the mechanisms whereby more widespread genetic variants predispose to common forms of disease. Our long-term aim is that better understanding of the pathways that control human energy balance, adipocyte biology and insulin sensitivity will, ultimately, lead to clinical benefits. Our research is supported by the MRC Centre for Obesity and Related Metabolic Diseases (MRC CORD), the Wellcome Trust, the NIHR Cambridge Biomedical Research Centre and the EU. Director, University of Cambridge Metabolic Research Laboratories Co-Director, Institute of Metabolic Science Scientific Director, NIHR Cambridge Biomedical Research Centre Level 4, Institute of Metabolic Science Box 289, Addenbrooke's Hospital Hills Road, Cambridge CB2 0QQ www.mrl.ims.cam.ac.uk Bochukova, E. G. et al. (2010). Large, rare chromosomal deletions associated with severe early-onset obesity. Nature 463, 666–670. Gandotra, S. et al. (2011). Perilpin deficiency and autosomal dominant partial lipodystrophy. N Eng J Med 364, 740–748. Protein Misfolding and Metabolic Disorders Proteins that fail to attain or maintain their structure reduce fitness in part through toxic gain of function mechanisms referred to as "proteotoxicity". The latter conspicuously affects poorly-renewable tissues of long-lived organisms in which the threat of protein misfolding can exert its deleterious consequences over extended periods of time. Protein misfolding is compartment-specific and its extent is influenced by the burden of newly-synthesized unfolded proteins presented to a given compartment (cytosol, endoplasmic reticulum, mitochondria) and by the protein folding environment in that compartment. The latter is influenced by structural elements operating within and on the compartment and by its metabolic state. Both parameters are regulated by complex homeostatic pathways, constituting what has been termed heuristically a proteostasis network, in which compartment-specific unfolded protein responses (UPR) are important. Stephen O’Rahilly and proteotoxic features of rare mutant proteins have revealed the importance of proteostasis to the function of tissues such as the endocrine pancreas, liver and fat that figure heavily in metabolic control. Less well understood, but of potentially considerable importance, are the emerging links between intermediary metabolism and the protein folding environment in the various compartments of the eukaryotic cell. Our research aims to uncover the molecular basis of the aforementioned reciprocal links. Wellcome Trust Principal Research Fellow University of Cambridge Metabolic Research Laboratories Level 4, Institute of Metabolic Science Box 289, Addenbrooke's Hospital Hills Road, Cambridge CB2 0QQ Tel: +44 (0) 1223 768 940 Email: [email protected] http://ron.medschl.cam.ac.uk David Ron Wiseman, R. L. et al. (2010). Flavonol activation defines an unanticipated ligand binding site in the kinase-rnase domain of ire1. Mol Cell 38, 291–304. Zito, E. et al. (2010). Oxidative protein folding by an endoplasmic reticulum localized peroxiredoxin. Mol Cell 40, 787–797. Interesting reciprocal links have been uncovered between protein folding homeostasis and metabolism: Defects in handling unfolded protein load 43 Principal Investigators who will be moving on from CIMR shortly David Clayton Wallace, C., Smyth, D. J., Maisuria-Armer, M., Walker, N. M., Todd, J. A. and Clayton, D. G. (2010). The imprinted DLK1-MEG3 gene region on chromosome 14q32.2 encodes susceptibility to type 1 diabetes. Nat Genet 42, 68–71. Clayton, D. (2010). On inferring presence of an individual in a mixture: a Bayesian approach. Biostatistics 11, 661–673. Andrew Peden Gordon, D. E. et al. (2010). A targeted siRNA screen to identify SNAREs required for constitutive secretion in mammalian cells. Traffic 11, 1191–1204. Miller, S. E. et al. (2011). The molecular basis for the endocytosis of small R-SNAREs by the clathrin adaptor CALM. Cell 147, 1118–1131. 44 Biostatistical Methods in Genetics Our aim is to develop and apply statistical methods in genetic epidemiology and, to a lesser extent, in other aspects of biostatistics. The main focus of this work is provided by the Diabetes and Inflammation Laboratory (DIL). Implementation and dissemination of analytical methods is an important part of our work. The snpMatrix package developed in my group became part of the Bioconductor international project for bioinformatic software in the R language. In addition to its uses in the analysis of genome-wide association studies, it became integral to the tools developed for “genetic genomics”. Largely to support and extend these uses, the package has been further developed and has been re-badged as “snpStats”. The group continues to provide biostatistical support to the DIL in the rapidly changing research environment which is evolving in the wake of the great successes of genomewide association studies. This has involved us addressing new problems presented by new source of data from, for example, DNA and RNA sequencing, expression microarrays, and flow cytometry. Our existing work on methods in genetic association studies has continued, in collaboration with Matt Hurles and Jeff Barrett at the Wellcome Trust Sanger Institute. Particular focus has been on, respectively, copy number variation and multivariate and multinomial phenotypes. Retiring at the end of March 2012, however will leave the continuing research programme in biostatistics in CIMR in the very capable hands of Chris Wallace and other members of the statistics team. Elucidating the Pathways and Molecular Machinery of Constitutive Secretion Constitutive secretion is a fundamental, conserved process that delivers newly synthesised proteins to the extracellular environment and is key to many important physiological processes such as antibody secretion. Hypersecretion of antibodies is associated with many immune-derived diseases such as Amyloidosis, Macroglobulinemia and Monoclonal Gammopathy of undetermined significance and may also have a role in auto-immune diseases such as Systemic Lupus Erythematosus. Elevated levels of circulating antibodies can cause severe pathology and lead to conditions such as Cryoglobulinemia and hyperviscosity syndrome. Thus, having the ability to modulate the levels of circulating antibodies has significant therapeutic potential. We hope that by studying the secretory pathway and determining the machinery required for antibody secretion we will not only provide new insights into a fundamental cellular process, but also identify new druggable targets for the treatment of these diseases. My research has three main aims: 1. To understand the role of SNAREs in constitutive secretion. 2. To identify and characterise novel machinery required for post-Golgi trafficking and antibody secretion. 3. To develop novel inhibitors of antibody secretion. Moving to the Department of Biomedical Science at the University of Sheffield to take up the post of Lecturer. 45 Postgraduate Opportunities in the Institute Introduction The CIMR offers an exciting and vibrant environment for graduate students. Approximately 30% of the 270 scientists within the CIMR are postgraduate students carrying out research towards a PhD. There are a variety of schemes that we offer to allow both clinical and non-clinically qualified individuals to pursue a PhD within the Institute. Some of these are for three years whilst others are four year programmes funded by the Wellcome Trust and Medical Research Council (MRC). Wellcome Trust/MRC Four Year PhD Programmes at the CIMR The CIMR houses 2 four year PhD programmes for non-clinical scientists. The Wellcome Trust four year PhD programme in Infection and Immunity (Director: Professor Paul Lehner) offers 6 studentships per annum. Approximately 50% of the faculty for this programme are members of the CIMR with other principal investigators being within the Department of Pathology, the Veterinary School and the School of Clinical Medicine. The CIMR also runs its own four year programme that is funded by the Wellcome Trust and the MRC. All principal investigators and affiliated principal investigators of the CIMR serve as faculty for this programme. The four year programmes allow individuals to undertake 3 ten week mini projects in the first year before deciding on the project that they wish to pursue for their three year PhD. These programmes are advertised in November with interviews in January/February. The successful applicants start the programme in October. principal investigators. They are advertised separately from the four year programmes run within the CIMR. The 3 year PhD students have access to the same seminars, equipment and mentoring as four year PhD students. Clinical Research Training Fellowships Clinical training fellowships are available as competitive awards through the Medical Research Council, the Wellcome Trust and the Association of Medical Charities. We also host the University of Cambridge/Sanger Institute Wellcome Trust PhD Programme for Clinicians that funds 7 clinical fellows per annum for 5 years. This scheme is administered from the CIMR although the faculty is drawn from the whole of the School of Clinical Medicine and the wider University of Cambridge. The Director of the four year non-clinical PhD programmes and the Wellcome Trust clinical PhD programme is Professor David Lomas, the Director for Graduate Studies within the Institute is Dr Folma Buss and the Programme Administrator is Sonia Lyne. Prof. Sheena Radford University of Leeds External examiner for CIMR 1+3 MRes/PhD Programme 2009–11 Non-Clinical Three Year PhD Programmes The CIMR offers standard three year PhD programmes. These are usually awarded by the research charities to individual Sonia Lyne CIMR 4 Yr PhD Students, 2011 intake 46 CIMR Research Retreat 2011 21–22 March 2011 Wellcome Trust Conference Centre, Hinxton, Cambridge The annual CIMR Research Retreat was held on 21–22 March 2011. This year it was the turn of the postdoctoral fellows and graduate students to present to members of the Institute. The presentations reflected the diverse nature of ongoing research within the building. We also had excellent presentations on antibody engineering from Sir Greg Winter and on chromosomal instability by Ashok Venkitaraman. We continued our tradition of inviting a member of the MRC Mitochondrial Biology Unit to talk at the Retreat and were delighted to hear from Ian Holt about his work on mitochondrial myopathies. The presentations from members of the Institute were of a very high standard and the judges awarded the prize for the best presentation from a postdoctoral fellow to Dr Janet Deane (Prof Randy Read’s group) now a PI herself, with the runner-up being Dr Jenny Lumb (Dr Evan Reid’s group). The prize for the best presentation from a graduate student went to Miss Hemma Brandstätter (Dr Folma Buss’s group) with the runner-up being Mr David Gordon (Dr Andrew Peden’s group). The poster prizes were given to Dr Xin Smith (postdoctoral fellow in Prof Chris Rudd’s group) and jointly to Miss Sally Thomas and Miss Elke Malzer (graduate students with Dr Stefan Marciniak). David Lomas Deputy Director Janet Deane Jenny Lumb Hemma Brandstätter David Gordon Sally Thomas/Elke Malzer Xin Smith 47 Administration, IT and Technical Support Contacts Administrator Miss Sarah Smith Tel: 01223 336457 E-Mail: [email protected] Laboratory/Facilities Manager Mr Dave Cheesman Tel: 01223 762314 E-Mail: [email protected] It is a privilege for me to introduce this section of the Report to acknowledge the work of all the support staff in what has been another busy and productive year. All staff endeavour to provide a support service to our scientists in an efficient and effective and customer focussed manner so that the scientific community remains satisfied with the value of the services provided. very happy to welcome Neil Kent to the team in August 2011. He leads the Accounts and Purchasing Unit and replacing Susan Reeder who retired having completed 12 years service. Neil has joined us from Cambridge Assessment, a department of the University, where he worked for nine years and qualified as a CIMA Management Accountant. As the Institute’s Administrator I have responsibility for overseeing the Administration’s individual units and for providing advice and guidance on a wide variety of administrative issues to the Director, Institute committees, the scientists, and staff. In this period of reduced funding opportunities, of particular importance is ensuring that the grant application process is followed appropriately with particular emphasis on costings to ensure that sufficient funds will be available to meet the needs of the research, whilst remaining within the limits set by the grant awarding bodies. I also have direct operational management responsibility for the Human Resources Unit. The service includes providing advice on all aspects of employment issues including work permits, adhering to HR best practice, and initiating appointments/extensions/termination of employment for employees. The team supports scientists by undertaking daily accounts work including the processing of purchase orders and expense claims, dealing with invoice queries, managing accounts receivable and providing information and advice regarding the financial position of grants. Training and development is key to offering a good service and most of the team are currently studying to aid their professional development. Since the last Report we have experienced the lowest turnover of administrative staff since the Institute was opened. However, there was one significant managerial change. We were Jonathan Wilson, continues to lead the IT Unit, has responsibility for the Institute's IT facilities, including the provision of a support service to both the scientific and administrative staff. His team manages the data network and audiovisual facilities within the Institute, and maintains the Intranet and Web site. Over the next 5 years the IT Unit will invest £300k into renewing the Institute's internal network, data centre and communal computing facilities. In 2010 the IT Unit modernised the facilities in the Sackler lecture theatre to provide state of the art digital presentation. The generation of scientific data within CIMR is now doubling annually and to Building Services Supervisor Finance Officer Computer Officer Mr Ray Woodstock Mr Neil Kent Mr Jonathan Wilson Tel: 01223 762666 E-mail: [email protected] Tel: 01223 762139 E-mail: [email protected] Tel: 01223 763194 E-mail: [email protected] 48 meet the challenge of data storage and back-up, the IT Unit is developing an off-site data replication system. The website presence has been revamped but there is always more we can do to communicate our services and facilities. The Laboratory and Facilities team remains under the direction of Dave Cheesman. As with the Accounts and Purchasing Unit, various members of the team have undertaken courses leading to professional accreditation. This team is responsible for the provision of building-wide services such as cleaning, catering, security, media preparation and glass-washing, and for the CIMR communal laboratories. The shared facilities provide essential equipment for use by the Research Staff and include dark room services, centrifugation, phosphor imaging, and shared areas such as Tissue Culture Labs and Containment Level 2 suites. Due to the nature of the service provided by this unit, there is a great deal of overlap with the Building Services Unit. Ray Woodstock manages the Building Services Unit which has recently expanded to include the Custodian function. The Unit provides the dayto-day maintenance of the building, including numerous small projects such as minor building modifications for new equipment and redecorating of the public areas. The Unit is also involved with major projects which have recently included cladding and external insulation improvements, lift refurbishment and the replacing of the building’s heating and cooling control system. Planned projects for the next year include replacing of the floor covering in all the public areas, and installing new cycle sheds. Sarah Smith CIMR Administrator CIMR Support Organisational Chart CIMR Support Organisational Chart Director CIMR Deputy Director CIMR PA to the Director PhD Administrator Laboratory/Facilities Manager Building Services Supervisor Deputy Lab Manager Snr. Building Services Tech. 2x Media Technicians Building Services Tech. Senior Level Co-ordinator 2x Custodians Assistant Level Co-ordinator 2x Glasswash Assistants Other Support Staff Administrator Secretary Office Manager Senior HR Clerk Finance Officer 4x Receptionists 2x HR Clerks Accounts & Purchasing Supervisor Purchasing Clerk (Snr. Custodian) 2x Accounts Clerks University Funded Wellcome Trust Infrastructure Funded Joint University / MRC Funded Various Sources of Funds Computer Officer Proteomics Technician Computing Technician Microscopy Facility Manager Bioinformatics / Systems Support Deputy Microscopy Facility Manager Head of Flow Cytometry Flow Cytometry Technician Flow Cytometry Technician University Funded / Cost Recovery Level Co-ordinator MRC Funded / Cost Recovery 49 Funding of CIMR Total expenditure on grants has continued to increase over the last two years, from £20.0m in 2008/9 to £21.4m in 2010/11, a rise of 7%. Overall Wellcome Trust support is approximately 60%, as it has been for the last 5 years. One-third of non-Wellcome Trust grants are funded by the MRC. CIMR Grant Expenditure Total Value of Grants Held in CIMR 50 Recurrent Costs for the CIMR In respect of building management and maintenance for the Wellcome Trust/MRC Building and the Cambridge Institute for Medical Research annual recurrent costs remain split between the University and MRC at a ratio directly proportional to the space occupied in the building. ecurrent Costs for the CIMR Recurrent Recurrent Costs for Costs CIMR August 2009–July 2010 August 2010–July 2011 Recurrent Costs for the Wellcome Trust/MRC Buil Recurrent Costs for the Wellcome Trust/MRC Build for the Wellcome Trust/MRC & CIMR Recurrent Costs Building for the Wellcome Trust/MRC Building & CIMR August 2009–July 2010 August 2010–July 2011 51 NIHR Cambridge Biomedical Research Centre core facilities supporting CIMR Principal Investigators Over the last four years the Clinical School with National Institute for Health Research (NIHR) Biomedical Research Centre (BRC) funding has been able to establish several critical core facilities that enable translational medical research across the main research themes on campus (see BRC website, http:// cambridge-brc.org.uk/), including many researchers in the Cambridge Institute for Medical Research. 1 The Cambridge BioResource (http://www.cambridgebioresource.org.uk/) allows researchers to recall volunteers for their studies based on disease-associated genotypes and phenotypes. This resource, with the renewal of the Cambridge Biomedical Research Centre to 2017, will now include patients from several clinics in the hospital. Moreover, we have just been awarded additional funding by NIHR to roll out the BioResource nationally for patients and populations. 2 The Cell Phenotyping Hub. This is a centralised flow cytometry and microscopy facility for phenotypic and functional analyses of human blood and tissue samples. The facility has state of the art equipment and is closely aligned to the Cambridge BioResource, for example in gene-to-phenotype analyses of immune-mediated diseases, such as lupus and type 1 diabetes. 3 The Eastern Sequence and Informatics Hub (EASIH) provides facility and expertise in next-generation sequencing and bioinformatics. Major projects include exome and human microbial pathogen sequencing, as well as the translation of high throughput sequencing into the Clinical Regional Genetics Laboratory, for example, in HLA tissue typing. Also housed in Clinical Biochemistry are facilities for core biomedical assays (Core Biochemical Assay Laboratory: http://www.cuh.org.uk/research/researchers/starting/ facilities/cbal/core_biochemical_assay.html) and microarray gene expression (http://cambridge-brc.org.uk/new/ infrastructure-infrastructure-laboratoryfacilities.htm). 4 The BRC BioRepository (NIHR BRC-MRC BioRepository, F & G Block, Level 2, Addenbrooke's Hospital) has been established with the MRC Epidemiology Unit, to provide smart, automated sample (DNA, plasma, serum) storage facilities of BRC- and MRC-associated samples. 52 Cambridge BRC Other Information Honours, Awards & Personal Fellowships Principal Investigators: David Clayton: Wellcome Trust Principal Research Fellowship, renewed December 2010; Honorary Chair, Department of Epidemiology and Population Health, Medical Statistics Unit, London School of Hygiene and Tropical Medicine Research Fellowship, March 2012 Fellowship Award, March 2011 Sergey Nezhentsev (Affiliated): Wellcome Trust Senior Research Fellowship in Basic Biomedical Science, June 2011 Geoffrey Hesketh (Luzio): Canadian Institute of Health Research Fellowship, August 2011 Katrin Ottersbach: British Society for Haematology Early Stage Investigator Fellowship; Leukaemia & Lymphoma Research Bennett Senior Fellowship, September 2011 Nick Matheson (Lehner): Wellcome Trust Clinical Research Training Fellowship, April 2011 Janet Deane: Royal Society University Research Fellowship, October 2011 David Owen: Wellcome Trust Principal Research Fellowship, October 2010 Anna Petrunkina Harrison: Extraordinary Professorship (‘Ausserplanmaessige Professeur’). University of Veterinary Medicine, Hanover, 2011 Sadaf Farooqi (Affiliated): Professorship in the University of Cambridge, October 2011 Randy Read: William A Bridger Lectureship in Biochemistry, University of Alberta, Edmonton, Canada, 2010 Rhys Roberts (Luzio): Wellcome-Beit Prize/Wellcome Trust Intermediate Clinical Fellowship, June 2011 Andres Floto: BUPA Foundation Researcher of the Year award 2010 David Rubinsztein: Wellcome Trust Principal Research Fellowship, March 2012 Chris Wallace (DIL): Wellcome Trust Career Development Fellowship, February 2010 Bertie Göttgens: 2010 McCulloch & Till Award by the International Society for Hematology and Stem Cells; Professorship in the University of Cambridge, October 2011 Christopher Rudd: Wellcome Trust Principal Research Fellowship, extended December 2010 Lena Wartosch (Luzio): FEBS Longterm Fellowship, April 2010; EMBO Fellowship, April 2011 Tony Green: Elected Distinguished Visiting Professor Cancer Science Institute, Singapore, 2009–10; Elected Newton Abraham Visiting Professorship, University of Oxford, 2011 Peter St George-Hyslop: Fellow of the Society of Biology Michael Weekes (Lehner): Wellcome Trust Intermediate Clinical Fellowship, July 2011 John Todd: David Rumbough Award for Scientific Excellence from the JDRF Fiona Gribble: Minkowski Prize, EASD 2010; University of Cambridge Readership, October 2010 Geoff Woods: Professorship in the University of Cambridge, October 2011 Aiwu Zhou (Read): BHF Senior Basic Science Research Fellowship, January 2011 Research Scientists: James Huntington: ISTH 2011 WrightSchulte Memorial Lecture; Professorship in the University of Cambridge, October 2011 Mark Dawson (Huntly): Wellcome-Beit Prize/Wellcome Trust Intermediate Clinical Fellowship, 2010 David Lomas: Patron, Alpha-1 Awareness; Visiting Professor, University of Florida and Cleveland Clinic Jyoti Evans (Green): Kay Kendall Leukaemia Fund Junior Clinical Research Fellowship, April 2011 Stefan Marciniak: MRC Senior Clinical Shaun Flint (Smith): ACT Clinical 53 Editorial Boards of Journals David Clayton: Annals of Human Genetics Andres Floto: PLoS ONE Bertie Göttgens: Experimental Hematology (Associate Editor) Fiona Gribble: British Journal of Pharmacology; Biochemical Journal Gillian Griffiths: Journal of Cell Biology; Traffic; Current Opinions in Cell Biology; Biomed Central Biology; Centrosome James Huntington: Journal of Biological Chemistry; Biochemical Journal Brian Huntly: Experimental Hematology David Lomas: American Journal of Respiratory Cell and Molecular Biology; Journal of Chronic Obstructive Pulmonary Disease (COPD); Clinical Medicine Paul Luzio: Traffic Stefan Marciniak: World Journal of Diabetes Randy Read: Acta Crystallographica Section D Margaret Robinson: Traffic David Rubinsztein: Autophagy (Associate Editor); Human Molecular Genetics; Journal of Applied Biomedicine; PLoS ONE (Academic Editor); Cell Death and Differentiation Christopher Rudd: Current Biology; Immunology; International Journal of Biochemistry; Proceedings National Academy of Science (PNAS); PLoS ONE; Frontiers in Bioscience; European Journal of Immunology; Self/Non-Self; Seminars in Immunopathology; Frontiers in T Cell Biology 54 Peter St George-Hyslop: Neurodegenerative Diseases; Molecular Neurodegeneration; American Journal of Alzheimer’s Disease Staff Affiliations Symeon Siniossoglou: Journal of Biological Chemistry David Clayton: MRC ASTRAL Trail Steering Committee; SNP Steering Group, Cancer Research UK; Council of the International Biometrics Society; GeneLibrary Ireland Scientific Advisory Board John Todd: Expert Reviews in Molecular Medicine; Human Molecular Genetics John Trowsdale: European Journal of Immunology; Tissue Antigens; Human Immunology Linda Wicker: Journal of Experimental Medicine (Advisory Editor) Folma Buss: Cell Biological Committee of the Biochemical Society Andres Floto: European Cystic Fibrosis Society Nontuberculous Mycobacteria (NTM) Group (Chair); ECFS–CF Foundation Guidelines Committee for NTM Infection (Co-chair) Bertie Göttgens: International Society for Hematology and Stem Cells (Director); BBSRC – Pool of experts/ member of core grant committees Tony Green: Membership of National/ International Bodies: Academy of Medical Science; American Society of Haematology; American Association of Physicians; Association of Physicians; British Society for Haematology; British Association for Cancer Research; European Haematology Association; European Haematology Association Scientific Program Committee; International Society for Experimental Haematology; 1942 Club; Selected Committees/ Research Administration: Head, University of Cambridge Department of Haematology; Chairman: Addenbrooke’s Hospital Haematology Senior Staff Group; Scientific Advisers to the Kay Kendall Leukaemia Fund (2004–); Member: European School of Haematology Executive Committee (2002–); NCRI Myeloproliferative Disorder Study Group (1997–); American Society of Hematology, Scientific Committee for Hematopoietic Cytokines and Factors (2008–); Scientific Advisory Board, Hemato–Linné Stem Cell Programme; European Haematology Association Board (2009–); Clinical Research Fellow Committee (2009–) Fiona Gribble: 2010– SAB member for OXION – Ion Channels and Disease Initiative, Universities of Oxford, Cambridge, London and MRC Harwell; 2011 – Aeres Visiting Committee, Dijon, France; External SAB Roche, Basel; EFSD/Amylin Grants Committee; WT/ NIH PhD Studentship grant panel member Gillian Griffiths: Wellcome Trust Sir Henry Wellcome Postdoctoral Fellowship Committee; EMBO Membership Committee Brian Huntly: CRUK Discovery Committee (2006–); LRF Medical and Scientific Advisory Panel (2009–); European Hematology Association, Membership Committee, (2006–); European Hematology Association, Member of the Scientific Advisory Committee for the 15th Annual European Hematology Association meeting, Barcelona 2010; Invited Faculty member, Faculty of 1000, (2009–) Fiona Karet: University WiSETI steering group (2007–); Wellcome Trust Clinical Interview Committee (2006–2011); Academy of Medical Sciences Clinical Research Champion (2007–); UK Renal Association Executive and Chair of Research Committee (2010–); UK Kidney Research Consortium lead: monogenics clinical study group (2009–); Kidney Research UK Trustee (2011–); Salt wasting alkaloses Rare Renal Disease Group lead (2011); Medical Education England 'Shape of Medical Training' Steering Group (2011–) Paul Lehner: Wellcome Trust Expert Review Group: ‘Immune System in Health and Disease’ (Chair); Wellcome Trust 4 Yr PhD Programme in Infection and Immunity (Director); Trustee British Society of Immunology David Lomas: British Lung Foundation Grants Committee (Chair); MRC Population and Systems Medicine Board (Chair); British Lung Foundation (Trustee); Academy of Medical Sciences Sectional Committee; Alpha-1 Foundation (US) Grants Committee (Chair); GSK Respiratory CEDD (Centre of Excellence in Drug Discovery) (Board member); Alpha-one Antitrypsin Laurell Training Award (ALTA); Wellcome Trust Clinical PhD Programme (Director); Wellcome Trust/MRC 4 Yr PhD Programme, CIMR (Director); BRC Fellowships and Capacity Building (Theme Lead); Cambridge University Clinical Research Society (Senior Treasurer & Patron); External Advisory Committee, HRB PhD Scholars Programme (Ireland); MRC PhD Studentship skills portfolio advisory group (member) Paul Luzio: MRC Molecular & Cellular Medicine Board (Chair) (until March 2012); MRC Strategy Board (member) (until March 2012) Stefan Marciniak: Fellow of the Royal College of Physicians; Fellow of St Catherine’s College, Cambridge Lucy Raymond: Action Medical Research Grants Committee Randy Read: wwPDB X-ray Validation Task Force (Chair); PDB-Europe Scientific Advisory Committee (Chair); CCP4 Executive Committee (member) Evan Reid: Scientific Advisory Board of American Spastic Paraplegia Foundation (member); Cell Biology Theme Panel of Biochemical Society (member) Margaret Robinson: Wellcome Trust Cell and Developmental Biology Expert Review Group (member) David Rubinsztein: Wellcome Trust Investigator Awards Expert Review Group (co-Chair) (2011–); Wellcome Trust Molecular & Cellular Neuroscience Committee (2007–11); Parkinson’s Disease Brain Bank Assessment Panel (2009–); MRC Brain Banks Network Users Group (2009–) Christopher Rudd: Council Member, International Union of Immunological Societies (IUIS) (2007–); Member, Organising Committee, 15th International Congress of Immunology (ICI) (2009–) (Rome, August, 2013); Mentor, Dual University Scheme, National University of Singapore and University of Cambridge (2009–); Member, Education Committee, International Union of Immunological Societies (IUIS) (2010–); Chairman and Founder, Immune Ventures plc, Cambridge (2008–); Consultant, AstraZeneca, UK (2009–); Consultant, Mewburn Ellis LLP (2010–); Scientific Advisory Board, Akanas Therapeutics Inc., Boston, USA (2010–); Consultant, HLBB Shaw, Epping, Essex (2011–) Peter St George-Hyslop: Alzheimer Research Trust Scientific Advisory Board, Institut du Cerveau et de la Moelle Epiniere de France, Agence Nationale de la Recherche, Department of Health, Ministerial Advisory Group on Dementia, Deutsches Zentrum Fur Neurodegenerative Erkrankungen, Scientific Advisory Board, GlaxoSmithKline Neuroscience John Todd: Type 1 Diabetes Genetics Consortium Steering Committee; Cambridge Computational Biology Institute Management Committee, University of Cambridge; Wellcome Trust Case Control Consortium Management Committee; NIHR Cambridge Biomedical Research Centre Cambridge BioResource Management Committee; Institute of Metabolic Science-Metabolic Research Laboratory Strategy Committee; Professorial Pay Committee, Clinical School, University of Cambridge; Scientific Advisory Board of NIHR Biomedical Research Centre Core Biochemical Assay Lab; Juvenile Diabetes Research Foundation UK Scientific Advisory Committee; Senior Academic Promotions 55 Committee, Clinical School, University of Cambridge; Council of School, Clinical School, University of Cambridge; Board of Electors, Professorship of Statistics in Biomedicine, University of Cambridge; Planning Steering Group, Clinical School, University of Cambridge; Scientific Advisory Board of NIHR Cambridge Biomedical Research Centre Cambridge BioResource; Management Committee (Chairman) of the MRC– NIHR Cambridge Biomedical Research Centre, University of Cambridge Eastern Sequence and Informatics Hub; Cambridge University Hospitals NHS Foundation Trust Biomedical Research Centre Executive Committee; Scientific Advisory Board of the Centre for Dermatology and Genetic Medicine, Medical Sciences Institute, Dundee; Member of the Advisory Committee for the Regius Professorship of Physic; Board of Electors, Genzyme Professorship of Experimental Medicine, University of Cambridge Linda Wicker: Type 1 Diabetes Repository Advisory Committee, National Institutes of Health (Chair and member) (2002–) Fellows of the Royal Society Stephen O’Rahilly, Peter St GeorgeHyslop, John Todd Fellows of the Academy of Medical Sciences Jenefer Blackwell, Tony Green, Gillian Griffiths, Fiona Karet, Paul Lehner, David Lomas, Paul Luzio, Stephen O’Rahilly, Margaret Robinson, David Rubinsztein, Christopher Rudd, Peter St GeorgeHyslop, Ken Smith, John Todd, John Trowsdale EMBO Members Gillian Griffiths, Stephen O’Rahilly, David Owen, Margaret Robinson, David Ron, David Rubinsztein 56 Publications for Scientists in CIMR since 2010 Abdollahi, M. R., Morrison, E., Sirey, T., Molnar, Z., Hayward, B. E., Carr, I. M., Springell, K., Woods, C. G., Ahmed, M., Hattingh, L. et al. (2009). Mutation of the variant alpha-tubulin TUBA8 results in polymicrogyria with optic nerve hypoplasia. Am J Hum Genet 85, 737–744. Albers, C. A., Cvejic, A., Favier, R., Bouwmans, E. E., Alessi, M. C., Bertone, P., Jordan, G., Kettleborough, R. N. W., Kiddle, G., Kostadima, M. et al. (2011). Exome sequencing identifies NBEAL2 as the causative gene for gray platelet syndrome. Nat Genet 43, 735–737. Adams, P. D., Afonine, P. V., Bunkóczi, G., Chen, V. B., Davis, I. W., Echols, N., Headd, J., Hung, L.-W., Kapral, G. J., Grosse-Kunstleve, R. W. et al. (2010). PHENIX: a comprehensive Pythonbased system for macromolecular structure solution. Acta Crystallogr D Biol Crystallogr 66, 213–221. Anand, S., Stedham, F., Beer, P., Gudgin, E., Ortmann, C. A., Bench, A., Erber, W., Green, A. R. and Huntly, B. J. P. (2011). Effects of the JAK2 mutation on the hematopoietic stem and progenitor compartment in human myeloproliferative neoplasms. Blood 118, 177–181. Adams, P. D., Afonine, P. V., Bunkóczi, G., Chen, V. B., Echols, N., Headd, J. J., Hung, L.-W., Jain, S., Kapral, G. J., Grosse-Kunstleve, R. W. et al. (2011). The Phenix software for automated determination of macromolecular structures. Methods 55, 94–106. Anand, S., Stedham, F., Gudgin, E., Campbell, P., Beer, P., Green, A. R. and Huntly, B. J. P. (2011). Increased basal intracellular signaling patterns do not correlate with JAK2 genotype in human myeloproliferative neoplasms. Blood 118, 1610–1621. Aguado, C., Sarkar, S., Korolchuk, V. I., Criado, O., Vernia, S., Boya, P., Sanz, P., de Cordoba, S. R., Knecht, E. and Rubinsztein, D. C. (2010). Laforin, the most common protein mutated in Lafora disease, regulates autophagy. Hum Mol Genet 19, 2867–2876. Andersson, A. K., Sumariwalla, P. F., McCann, F. E., Amjadi, P., Chang, C., McNamee, K., Tornehave, D., Haase, C., Agersø, H., Stennicke, V. W. et al. (2011). Blockade of NKG2D ameliorates disease in mice with collagen-induced arthritis: A potential pathogenic role in chronic inflammatory arthritis. Arthritis Rheum 63, 2617–2629. Agusti, A., Calverley, P. M. A., Celli, B., Coxson, H. O., Edwards, L. D., Lomas, D. A., MacNee, W., Miller, B. E., Rennard, S., Silverman, E. K. et al. (2010). Characterisation of COPD heterogeneity in the ECLIPSE cohort. Respir Res 11, 122. Ahn, H., Dib-Hajj, S. D., Cox, J. J., Tyrrell, L., Elmslie, F. V., Clarke, A. A., Drenth, J. P. H., Woods, C. G. and Waxman, S. G. (2010). A new Nav1.7 sodium channel mutation I234T in a child with severe pain. Eur J Pain 14, 944–950. Anichtchik, O., Diekmann, H., Fleming, A., Roach, A., Goldsmith, P. and Rubinsztein, D. C. (2011). Expression of PINK1 in the brain, eye and ear of mouse during embryonic development. J Chem Neuroanat 42, 99–100. Babayan, S. A., Allen, J. E., Bradley, J. E., Geuking, M. B., Graham, A. L., Grencis, R. K., Kaufman, J., McCoy, K. D., Paterson, S., Smith, K. G. C. et al. (2011). Wild immunology: converging on the real world. Ann N Y Acad Sci 1236, 17–29. Bachmann, P. S., Piazza, R. G., Janes, M. E., Wong, N. C., Davies, C., Mogavero, A., Bhadri, V. A., Szymanska, B., Geninson, G., Magistroni, V. et al. (2010). Epigenetic silencing of BIM in glucocorticoid poorresponsive pediatric acute lymphoblastic leukemia, and its reversal by histone deacetylase inhibition. Blood 116, 3013–3022. Bafadhel, M., McKenna, S., Terry, S., Mistry, V., Reid, C., Haldar, P., Kebadze, T., Duvoix, A., Lindblad, K., Patel, H. et al. (2011). Acute exacerbations of COPD: identification of biological clusters and their biomarkers. Am J Respir Crit Care Med (epub). Bakircioglu, M., Carvalho, O. P., Khurshid, M., Cox, J. J., Tuysuz, B., Barak, T., Yilmaz, S., Caglayan, O., Dincer, A., Nicholas, A. K. et al. (2011). The essential role of centrosomal NDE1 in human cerebral cortex neurogenesis. Am J Hum Genet 88, 523–535. Bakke, P. S., Zhu, G., Gulsvik, A., Kong, X., Agusti, A. G. N., Calverley, P. M. A., Donner, C. F., Levy, R. D., Make, B. J., Paré, P. D. et al. (2011). Candidate genes for COPD in two large data sets. Eur Respir J 37, 255–263. Barrow, A. D., Raynal, N., Andersen, T. L., Slatter, D. A., Bihan, D., Pugh, N., Cella, M., Kim, T., Rho, J., NegishiKoga, T. et al. (2011). OSCAR is a collagen receptor that costimulates osteoclastogenesis in DAP12-deficient humans and mice. J Clin Invest 121, 3505–3516. Beer, P. A., Campbell, P. J. and Green, A. R. (2010). Comparison of different criteria for the diagnosis of primary myelofibrosis reveals limited clinical utility for measurement of serum lactate dehydrogenase. Haematologica 95, 1960–1963. 57 Beer, P. A., Delhommeau, F., LeCouédic, J. P., Dawson, M. A., Chen, E., Bareford, D., Kušec, R., McMullin, M. F., Harrison, C. N., Vannucchi, A. M. et al. (2010). Two routes to leukemic transformation after a JAK2 mutationpositive myeloproliferative neoplasm. Blood 115, 2891–2900. Beer, P. A., Erber, W. N., Campbell, P. J. and Green, A. R. (2011). Essential thrombocythemia: seeing the wood for the trees Response. Blood 118, 1180–1181. Beer, P. A., Erber, W. N., Campbell, P. J. and Green, A. R. (2011). How I treat essential thrombocythemia. Blood 117, 1472–1482. Beer, P. A., Ortmann, C. A., Campbell, P. J. and Green, A. R. (2010). Independently acquired biallelic JAK2 mutations are present in a minority of patients with essential thrombocythemia. Blood 116, 1013–1014. Beer, P. A., Ortmann, C. A., Stegelmann, F., Guglielmelli, P., Reilly, J. T., Larsen, T. S., Hasselbalch, H. C., Vannucchi, A. M., Möller, P, Döhner, K. and Green, A. R. (2010). Molecular mechanisms associated with leukemic transformation of MPLmutant myeloproliferative neoplasms. Haematologica 95, 2153–2156. Belorgey, D., Hägglöf, P., Onda, M. and Lomas, D. A. (2010). pH-dependent stability of neuroserpin is mediated by histidines 119 and 138; implications for the control of beta-sheet A and polymerization. Protein Sci 19, 220–228. Belorgey, D., Irving, J. A., Ekeowa, U. I., Freeke, J., Roussel, B. D., Miranda, E., Pérez, J., Robinson, C. V., Marciniak, S. J., Crowther, D. C., Michel, C. H. and Lomas, D. A. (2011). Characterisation of serpin polymers in vitro and in vivo. Methods 53, 255–266. Bennett, N. J., Ashiru, O., Morgan, F. J., Pang, Y., Okecha, G., Eagle, R. A., Trowsdale, J., Sissons, J. G. P. and Wills, M. R. (2010). Intracellular 58 sequestration of the NKG2D ligand ULBP3 by human cytomegalovirus. J Immunol 185, 1093–1102. Blackstone, C., O'Kane, C. J. and Reid, E. (2011). Hereditary spastic paraplegias: membrane traffic and the motor pathway. Nat Rev Neurosci 12, 31–42. Blyth, M., Raponi, M., Treacy, R., Raymond, F. L., Yates, J. R. W. and Baralle, D. (2010). Expanding the tuberous sclerosis phenotype: mild disease caused by a TSC1 splicing mutation. J Neurol Neurosur PS 81, 350–352. Bonadies, N., Foster, S. D., Chan, W. I., Kvinlaug, B. T., Spensberger, D., Dawson, M. A., Spooncer, E., Whetton, A. D., Bannister, A. J., Huntly, B. J. P. and Göttgens, B. (2011). Genome-wide analysis of transcriptional reprogramming in mouse models of acute myeloid leukaemia. PLoS One 6, e16330. Boname, J. M. and Lehner, P. J. (2011). What has the study of the K3 and K5 viral ubiquitin E3 ligases taught us about ubiquitin-mediated receptor regulation? Viruses-Basel 3, 118–131. Boname, J. M., Thomas, M., Stagg, H. R., Xu, P., Peng, J. and Lehner, P. J. (2010). Efficient internalization of MHC I requires lysine-11 and lysine-63 mixed linkage polyubiquitin chains. Traffic 11, 210–220. Bond, L. M., Brandstaetter, H., Sellers, J. R., Kendrick-Jones, J. and Buss, F. (2011). Myosin motor proteins are involved in the final stages of the secretory pathways. Biochem Soc Trans 39, 1115–1119. Bond, L. M., Peden, A. A., KendrickJones, J., Sellers, J. R. and Buss, F. (2011). Myosin VI and its binding partner optineurin are involved in secretory vesicle fusion at the plasma membrane. Mol Biol Cell 22, 54–65. Bowsher, D., Woods, C. G., Nicholas, A. K., Carvalho, O. M., Haggett, C. E., Tedman, B., Mackenzie, J. M., Crooks, D., Mahmood, N., Twomey, J. A. et al. (2009). Absence of pain with hyperhidrosis: A new syndrome where vascular afferents may mediate cutaneous sensation. Pain 147, 287–298. Boyd, E. M., Bench, A. J., GodayFernández, A., Anand, S., Vaghela, K. J., Beer, P., Scott, M. A., Bareford, D., Green, A. R., Huntly, B. J. P. and Erber, W. N. (2010). Clinical utility of routine MPL exon 10 analysis in the diagnosis of essential thrombocythaemia and primary myelofibrosis. Br J Haematol 149, 250–257. Brehm, J. M., Hagiwara, K., Tesfaigzi, Y., Bruse, S., Mariani, T. J., Bhattacharya, S., Boutaoui, N., Ziniti, J. P., Soto-Quiros, M. E., Avila, L. et al. (2011). Identification of FGF7 as a novel susceptibility locus for chronic obstructive pulmonary disease. Thorax (epub). Brorsson, A. C., Bolognesi, B., Tartaglia, G. G., Shammas, S. L., Favrin, G., Watson, I., Lomas, D. A., Chiti, F., Vendruscolo, M., Dobson, C. M., Crowther, D. C. and Luheshi, L. M. (2010). Intrinsic determinants of neurotoxic aggregate formation by the amyloid beta peptide. Biophys J 98, 1677–1684. Brorsson, A. C., Kumita, J. R., MacLeod, I., Bolognesi, B., Speretta, E., Luheshi, L. M., Knowles, T. P. J., Lomas, D. A., Dobson, C. M. and Crowther, D. C. (2010). Methods and models in neurodegenerative and systemic protein aggregation diseases. Front Biosci 15, 373–396. Bruni, A. C., Bernardi, L., Colao, R., Rubino, E., Smirne, N., Frangipane, F., Terni, B., Curcio, S. A. M., Mirabelli, M., Clodomiro, A. et al. (2010). Worldwide distribution of PSEN1 Met146Leu mutation: a large variability for a founder mutation. Neurology 74, 798–806. Bunkóczi, G. and Read, R. J. (2011). Improvement of molecular-replacement models with Sculptor. Acta Crystallogr D Biol Crystallogr 67, 303–312. Burr, M. L., Cano, F., Svobodova, S., Boyle, L. H., Boname, J. M. and Lehner, P. J. (2011). HRD1 and UBE2J1 target misfolded MHC class I heavy chains for endoplasmic reticulumassociated degradation. Proc Natl Acad Sci USA 108, 2034–2039. Burren, O. S., Adlem, E. C., Achuthan, P., Christensen, M., Coulson, R. M. and Todd, J. A. (2011). T1DBase: update 2011, organization and presentation of large-scale data sets for type 1 diabetes research. Nucleic Acids Res 39, D997–1001. Buss, F. and Kendrick-Jones, J. (2011). Multifunctional myosin VI has a multitude of cargoes. Proc Natl Acad Sci USA 108, 5927–5928. Butler, R., Beattie, B. L., Thong, U. P., Dwosh, E., Guimond, C., Feldman, H. H. et al. (2010). A novel PS1 gene mutation in a large Aboriginal kindred. Can J Neurol Sci 37, 359–364. Caffarel, M. M., Zaragoza, R., Pensa, S., Li, J., Green, A. R. and Watson, C. J. (2011). Constitutive activation of JAK2 in mammary epithelium elevates Stat5 signalling, promotes alveologenesis and resistance to cell death, and contributes to tumourigenesis. Cell Death Differ (epub). Calero-Nieto, F. J., Wood, A. D., Wilson, N. K., Kinston, S., Landry, J. R. and Göttgens, B. (2010). Transcriptional regulation of Elf-1: locus-wide analysis reveals four distinct promoters, a tissue-specific enhancer, control by PU.1 and the importance of Elf-1 downregulation for erythroid maturation. Nucleic Acids Res 38, 6363–6374. Cano, F., Miranda-Saavedra, D. and Lehner, P. J. (2010). RNA-binding E3 ubiquitin ligases: novel players in nucleic acid regulation. Biochem Soc Trans 38, 1621–1626. Carvalho, O. P., Thornton, G. K., Hertecant, J., Houlden, H., Nicholas, A. K., Cox, J. J., Rielly, M., Al-Gazali, L. and Woods, C. G. (2011). A novel NGF mutation clarifies the molecular mechanism and extends the phenotypic spectrum of the HSAN5 neuropathy. J Med Genet 48, 131–135. Castaldi, P. J., Demeo, D. L., Hersh, C. P., Lomas, D. A., Soerheim, I. C., Gulsvik, A., Bakke, P., Rennard, S., Pare, P., Vestbo, J., AATGM Investigators, ICGN Investigators and Silverman, E. K. (2011). Impact of non-linear smoking effects on the identification of gene-by-smoking interactions in COPD genetics studies. Thorax 66, 903–909. Castanet, M., Mallya, U., Agostini, M., Schoenmakers, E., Mitchell, C., Demuth, S., Raymond, F. L., Schwabe, J., Gurnell, M. and Chatterjee, V. K. (2010). Maternal isodisomy for chromosome 9 causing homozygosity for a novel FOXE1 mutation in syndromic congenital hypothyroidism. J Clin Endocrinol Metab 95, 4031–4036. Castellucci, L., Jamieson, S. E., Miller, E. N., de Almeida, L. F., Oliveira, J., Magalhães, A., Guimaraes, L., Lessa, M., Lago, E., de Jesus, A., Carvalho, E. and Blackwell, J. M. (2011). FLI1 polymorphism affects susceptibility to cutaneous leishmaniasis in Brazil. Genes Imm 12, 589–594. Castellucci, L., Jamieson, S. E., Miller, E. N., Menezes, E., Oliveira, J., Magalhães, A., Guimaraes, L., Lessa, M., Ribeiro, S., Reale, J., de Jesus, A., Carvalho, E. and Blackwell, J. M. (2010). CXCR1 and SLC11A1 polymorphisms affect susceptibility to cutaneous leishmaniasis in Brazil: a case-control and family-based study. BMC Med Genet 11, 10. Catapano, F., Chaudhry, A. N., Jones, R. B., Smith, K. G. C. and Jayne, D. W. (2010). Long-term efficacy and safety of rituximab in refractory and relapsing systemic lupus erythematosus. Nephrol Dial Transplant 25, 3586–3592. Cetica, V., Pende, D., Griffiths, G. M. and Aricò, M. (2010). Molecular basis of familial hemophagocytic lymphohistiocytosis. Haematologica 95, 538–541. Cetica, V., Santoro, A., Gilmour, K. C., Sieni, E., Beutel, K., Pende, D., Marcenaro, S., Koch, F., Grieve, S., Wheeler, R. et al. (2010). STXBP2 mutations in children with familial haemophagocytic lymphohistiocytosis type 5. J Med Genet 47, 595–600. Chacko, A. R., Arifullah, M., Sastri, N. P., Jeyakanthan, J., Ueno, G., Sekar, K., Read, R. J., Dodson, E. J., Rao, D. C. and Suguna, K. (2011). A novel pentameric structure of the diarrhea-inducing region of the rotavirus enterotoxigenic protein NSP4. J Virol 85, 12721–12732. Chan, W. I., Hannah, R. L., Dawson, M. A., Pridans, C., Foster, D., Joshi, A., Göttgens, B., Van Deursen, J. M. and Huntly, B. J. P. (2011). The transcriptional coactivator Cbp regulates self-renewal and differentiation in adult hematopoietic stem cells. Mol Cell Biol (epub). Chen, E., Beer, P. A., Godfrey, A. L., Ortmann, C. A., Ahn, M., Li, J., CostaPereira, A, Ingle, C. E., Dermitzakis, E. T. and Green, A. R. (2011). Distinct clinical phenotypes associated with JAK2V617F reflect differential STAT1 signalling. Exp Hematol 39, S46. Chen, E., Beer, P. A., Godfrey, A. L., Ortmann, C. A., Li, J., Costa-Pereira, A., Ingle, C. E., Dermitzakis, E. T., Campbell, P. J. and Green, A. R. (2010). Distinct clinical phenotypes associated with JAK2V617F reflect differential STAT1 signaling. Cancer Cell 18, 524–535. Chen, J., Morgan, A. J., StewartJones, G., Shepherd, D., Bossi, G., Wooldridge, L., Hutchinson, S. L., Sewell, A. K., Griffiths, G. M., van der Merwe, P. A. et al. (2010). Ca2+ release from the endoplasmic reticulum of NY-ESO-1-specific T cells is modulated by the affinity of TCR and by the use of the CD8 coreceptor. J Immunol 184, 1829–1839. 59 Chibalina, M. V., Poliakov, A., Kendrick-Jones, J. and Buss, F. (2010). Myosin VI and optineurin are required for polarized EGFR delivery and directed migration. Traffic 11, 1290–1303. Chilvers, E. R. Lomas, D. A. (2010). Diagnosing COPD in non-smokers: splitting not lumping. Thorax 65, 465–466. Chinthalapudi, K., Taft, M. H., Martin, R., Heissler, S. M., Preller, M., Hartmann, F. K., Brandstaetter, H., Kendrick-Jones, J., Tsiavaliaris, G., Gutzeit, H. O. et al. (2011). Mechanism and specificity of pentachloropseudilinmediated inhibition of myosin motor activity. J Biol Chem 286, 29700–29708. Cho, M. H., Boutaoui, N., Klanderman, B. J., Sylvia, J. S., Ziniti, J. P., Hersh, C. P., DeMeo, D. L., Hunninghake, G. M., Litonjua, A. A., Sparrow, D. et al. (2010). Variants in FAM13A are associated with chronic obstructive pulmonary disease. Nat Genet 42, 200–202. Choi, H. S., Su, W. M., Morgan, J. M., Han, G. S., Xu, Z., Karanasios, E., Siniossoglou, S. and Carman, G. M. (2011). Phosphorylation of phosphatidate phosphatase regulates its membrane association and physiological functions in Saccharomyces cerevisiae: identification of SER(602), THR(723), AND SER(744) as the sites phosphorylated by CDC28 (CDK1)encoded cyclin-dependent kinase. J Biol Chem 286, 1486–1498. Cipriani, V., Matharu, B. K., Khan, J. C., Shahid, H., Hayward, C., Wright, A. F., Armbrecht, A. M., Dhillon, B., Harding, S. P., Bishop, P. N., Bunce, C., Clayton, D. G., Moore, A. T. and Yates, J. R. W. (2011). No evidence of association between complement factor I genetic variant rs10033900 and age-related macular degeneration. Eur J Hum Genet (epub). Cipriani, V., Matharu, B. K., Khan, J. C., Shahid, H., Stanton, C. M., Hayward, C., Wright A. F., Bunce, C., Clayton, D. G., Moore, A. T. and Yates, J. R. W. (2011). Genetic variation in complement 60 regulators and susceptibility to age-related macular degeneration. Immunobiol (epub). Clark, R. D., Graham, J. M., Friez, M. J., Hoo, J. J., Jones, K. L., McKeown, C., Moeschler, J. B., Raymond, F. L., Rogers, R. C., Schwartz, C. E. et al. (2009). FG syndrome, an X-linked multiple congenital anomaly syndrome: The clinical phenotype and an algorithm for diagnostic testing. Genet Med 11, 769–775. Clatworthy, M. R., Espeli, M., Torpey, N. and Smith, K. G. C. (2010). The generation and maintenance of serum alloantibody. Curr Opin Immunol 22, 669–681. Clayton, D. (2010). On inferring presence of an individual in a mixture: a Bayesian approach. Biostatistics 11, 661–673. Cluett, C., Brayne, C., Clarke, R., Evans, G., Matthews, F., Rubinsztein, D. C., Huppert, F., Llewellyn, D. J., Rice, N., Henley, W. et al. (2010). Polymorphisms in LMNA and near a SERPINA gene cluster are associated with cognitive function in older people. Neurobiol Aging 31, 1563–1568. Cooper, J. D., Smyth, D. J., Walker, N. M., Stevens, H., Burren, O. S., Wallace, C., Greissl, C., Ramos-Lopez, E., Hyppönen, E., Dunger, D. B. et al. (2011). Inherited variation in vitamin D genes is associated with predisposition to autoimmune disease type 1 diabetes. Diabetes 60, 1624–1631. Corrochano, S., Renna, M., Carter, S., Chrobot, N., Kent, R., Stewart, M., Cooper, J., Brown, S. D., Rubinsztein, D. C. and Acevedo-Arozena, A. (2011). Alpha-synuclein levels modulate Huntington’s disease in mice. Hum Mol Genet (epub). Cotsapas, C., Voight, B. F., Rossin, E., Lage, K., Neale, B. M., Wallace, C., Abecasis, G. R., Barrett, J. C., Behrens, T., Cho, J. et al. (2011). Pervasive sharing of genetic effects in autoimmune disease. PLoS Genet 7, e1002254. Cox, J. J., Sheynin, J., Shorer, Z., Reimann, F., Nicholas, A. K., Zubovic, L., Baralle, M., Wraige, E., Manor, E., Levi, J., Woods, C. G. and Parvari, R. (2010). Congenital insensitivity to pain: novel SCN9A missense and in-frame deletion mutations. Hum Mutat 31, E1670–E1686. Cox, J. J., Willatt, L., Homfray, T. and Woods, C. G. (2011). A SOX9 duplication and familial 46,XX developmental testicular disorder. N Engl J Med 364, 91–93. Craddock, N., Hurles, M. E., Cardin, N., Pearson, R. D., Plagnol, V., Robson, S. and Wellcome Trust Case Control. (2010). Genome-wide association study of CNVs in 16,000 cases of eight common diseases and 3,000 shared controls. Nature 464, 713–720. da Silva Xavier, G., Farhan, H., Kim, H., Caxaria, S., Johnson, P., Hughes, S., Bugliani, M., Marselli, L., Marchetti, P., Birzele, F. et al. (2011). Per-arnt-sim (PAS) domain-containing protein kinase is downregulated in human islets in type 2 diabetes and regulates glucagon secretion. Diabetologia 54, 819–827. Daniele, T., Hackmann, Y., Ritter, A. T., Wenham, M., Booth, S., Bossi, G., Schintler, M., Auer-Grumbach, M. and Griffiths, G. M. (2011). A role for Rab7 in the movement of secretory granules in cytotoxic T lymphocytes. Traffic 12, 902–911. Davidson, J. E., Irizarry, M. C., Bray, B. C., Wetten, S., Galwey, N., Gibson, R., Borrie, M., Delisle, R., Feldman, H. H., Hsiung, G. et al. (2010). An exploration of cognitive subgroups in Alzheimer's disease. J Int Neuropsych Soc 16, 233–243. Davies, J. E. and Rubinsztein, D. C. (2011). Over-expression of BCL2 rescues muscle weakness in a mouse model of oculopharyngeal muscular dystrophy. Hum Mol Genet 20, 1154– 1163. Davies, J. E., Rose, C., Sarkar, S. and Rubinsztein, D. C. (2010). Cystamine suppresses polyalanine toxicity in a mouse model of oculopharyngeal muscular dystrophy. Sci Transl Med 2, 34ra40. Davison, L. J., Wallace, C., Cooper, J. D., Cope, N. F., Wilson, N. K., Smyth, D. J., Howson, J. M., Saleh, N., Al-Jeffery, A., Angus, K. L. et al. (2011). Long-range DNA looping and gene expression analyses identify DEXI as an autoimmune disease candidate gene. Hum Mol Genet (epub). Dawson, M. A., Prinjha, R. K., Dittman, A., Giotopoulos, G., Bantscheff, M., Chan, W. I., Robson, S. C., Chung, C., Hopf, C., Savitski, M. M. et al. (2011). Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature 478, 529–533. Dawson, M. A., Curry, J. E., Barber, K., Beer, P. A., Graham, B., Lyons, J. F., Richardson, C. J., Scott, M. A., Smyth, T., Squires, M. S. et al. (2010). AT9283, a potent inhibitor of the Aurora kinases and Jak2, has therapeutic potential in myeloproliferative disorders. Br J Haematol 150, 46–57. De Marinis, Y. Z., Salehi, A., Ward, C. E., Zhang, Q. A., Abdulkader, F., Bengtsson, M., Braha, O., Braun, M., Ramracheya, R., Amisten, S. et al. (2010). GLP-1 inhibits and adrenaline stimulates glucagon release by differential modulation of N- and L-type Ca2+ channel-dependent exocytosis. Cell Metab 11, 543–553. Deane, J. E., Abrusci, P., Johnson, S. and Lea, S. M. (2010). Timing is everything: the regulation of type III secretion. Cell Mol Life Sci 67, 1065–1075. Deane, J. E., Graham, S. C., Kim, N. N., Stein, P. E., McNair, R., CachónGonzález, M. B., Cox, T. M. and Read, R. J. (2011). Insights into Krabbe disease from structures of galactocerebrosidase. Proc Natl Acad Sci USA 108, 15169–15173. Dickens, J. A. and Lomas, D. A. (2011). Why has it been so difficult to prove the efficacy of alpha-1-antitrypsin replacement therapy? Insights from the study of disease pathogenesis. Drug Des Devel Ther 5, 391–405. mechanism of polymerization in the serpinopathies. Proc Natl Acad Sci USA 107, 17146–17151. DiMaio, F., Terwilliger, T. C., Read, R. J., Wlodawer, A., Oberdorfer, G., Wagner, U., Valkov, E., Alon, A., Fass, D., Axelrod, H. L. et al. (2011). Improved molecular replacement by densityand energy-guided protein structure optimization. Nature 473, 540–543. Ekeowa, U. I., Marciniak, S. J. and Lomas, D. A. (2011). "(1)-antitrypsin deficiency and inflammation. Expert Rev Clin Immunol 7, 243–252. Downes, K., Pekalski, M., Angus, K. L., Hardy, M., Nutland, S., Smyth, D. J., Walker, N. M., Wallace, C and Todd, J. A. (2010). Reduced expression of IFIH1 is protective for type 1 diabetes. PLoS One 5, e12646. Duncan, L. M., Nathan, J. A. and Lehner, P. J. (2010). Stabilization of an E3 ligase-E2-ubiquitin complex increases cell surface MHC class I expression. J Immunol 184, 6978–6985. Durrington, H. J., Upton, P. D., Hoer, S., Boname, J., Dunmore, B. J., Yang, J., Crilley, T. K., Butler, L. M., Blackbourn, D. J., Nash, G. B., Lehner, P. J. and Morrell, N. W. (2010). Identification of a lysosomal pathway regulating degradation of the bone morphogenetic protein receptor type II. J Biol Chem 285, 37641–37649. Duvoix, A., Miranda, E., Perez, J., Sorensen, G. L., Holmskov, U., Trapnell, B. C., Madsen, J., Clark, H. W., Edwards, L. D., Miller, B. E., TalSinger, R. M. and Lomas, D. A. (2011). Evaluation of full-length, cleaved and nitrosylated serum surfactant protein D as biomarkers for COPD. COPD 8, 79–95. Ehsani, S., Huo, H., Salehzadeh, A., Pocanschi, C. L., Watts, J. C., Wille, H., Westaway, D., Rogaeva, E., St George-Hyslop, P. H. and SchmittUlms, G. (2011). Family reunion-the ZIP/prion gene family. Prog Neurobiol 93, 405–420. Ekeowa, U. I., Freeke, J., Miranda, E., Gooptu, B., Bush, M. F., Pérez, J., Teckman, J., Robinson, C. V. and Lomas, D. A. (2010). Defining the Ellingsgaard, H., Hauselmann, I., Schuler, B., Habib, A. M., Baggio, L. L., Meier, D. T., Eppler, E., Bouzakri, K., Wueest, S., Muller, Y. D. et al. (2011). Interleukin-6 enhances insulin secretion by increasing L cell and " cell glucagonlike peptide-1 secretion. Nat Med 17, 1481–1489. Eriguchi, M., Mizuta, H., Luo, S., Kuroda, Y., Hara, H. and Rubinsztein, D. C. (2010). Alpha Pix enhances mutant huntingtin aggregation. J Neurol Sci 290, 80–85. Espeli, M., Bökers, S., Giannico, G., Dickinson, H. A., Bardsley, V., Fogo, A. B. and Smith, K. G. C. (2011). Local renal autoantibody production in lupus nephritis. J Am Soc Nephrol 22, 296–305. Espeli, M., Niederer, H. A., Traherne, J. A., Trowsdale, J. and Smith, K. G. C. (2010). Genetic variation, Fc! receptors, KIRs and infection: the evolution of autoimmunity. Curr Opin Immunol 22, 715–722. Fabani, M. M., Abreu-Goodger, C., Williams, D., Lyons, P. A., Torres, A. G., Smith, K. G. C., Enright, A. J., Gait, M. J. and Vigorito, E. (2010). Efficient inhibition of miR-155 function in vivo by peptide nucleic acids. Nucleic Acids Res 38, 4466–4475. Fakiola, M.†, Mishra, A.†, Rai, M., Singh, S. P., O’Leary, R., Ball, S., Francis, R. W., Miller, E. N., Sundar, S.* and Blackwell, J. M.* (2010). Genome wide linkage study for visceral leishmaniasis in India. PLoS One 5, e15807. † equal first authorship; *equal contributions 61 Fakiola, M., Miller, E. N., Fadl, M., Mohamed, H. S., Jamieson, S. E., Francis, R. W., Cordell, H. J., Peacock, C. S., Raju, M., Khalil, E. A. et al. (2011). Genetic and functional evidence implicating DLL1 as the gene that influences susceptibility to visceral leishmaniasis at chromosome 6q27. J Infect Dis 204, 467–477. Fitch, S., Kimber, G. M., Wilson, N. K., Göttgens, B., Dzierzak, E. and Ottersbach, K. (2011). A role for GATA3 and the sympathetic nervous system innthe emergence of haematopoietic stem cells. Exp Hematol 39, S86. Fleming, A. and Rubinsztein, D. C. (2011). Zebrafish as a model to understand autophagy and its role in neurological disease. Biochim Biophys Acta 1812, 520–526. Fleming, A., Noda, T., Yoshimori, T. and Rubinsztein, D. C. (2011). Chemical modulators of autophagy as biological probes and potential therapeutics. Nat Chem Biol 7, 9–17. Foreman, M. G., Kong, X., DeMeo, D. L., Pillai, S. G., Hersh, C. P., Bakke, P., Gulsvik, A., Lomas, D. A., Litonjua, A. A., Shapiro, S. D. et al. (2011). Polymorphisms in surfactant protein-D are associated with chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 44, 316–322. Fraser, H. I., Dendrou, C. A., Healy, B., Rainbow, D. B., Howlett, S., Smink, L. J., Gregory, S., Steward, C. A., Todd, J. A., Peterson, L. B. and Wicker, L. S. (2010). Nonobese diabetic congenic strain analysis of autoimmune diabetes reveals genetic complexity of the Idd18 locus and identifies Vav3 as a candidate gene. J Immunol 184, 5075–5084. Friedlander, R. S., Moss, C. E., Mace, J., Parker, H. E., Tolhurst, G., Habib, A. M., Wachten, S., Cooper, D. M., Gribble, F. M.* and Reimann, F.* (2011). Role of phosphodiesterase and adenylate cyclase isozymes in murine colonic glucagon-like peptide 1 secreting cells. Br J Pharmacol 163, 261–271. 62 Fung, E., Esposito, L., Todd, J. A. and Wicker, L. S. (2010). Multiplexed immunophenotyping of human antigenpresenting cells in whole blood by polychromatic flow cytometry. Nat Protoc 5, 357–370. Gabryelewicz, T., Masellis, M., Berdynski, M., Bilbao, J. M., Rogaeva, E., St George-Hyslop, P., Barczak, A., Czyzewski, K., Barcikowska, M., Wszolek, Z. et al. (2010). Intra-familial clinical heterogeneity due to FTLD-U with TDP-43 proteinopathy caused by a novel deletion in progranulin gene (PGRN). J Alzheimers Dis 22, 1123–1133. Galluzzi, L., Vitale, I., Abrams, J. M., Alnemri, E. S., Baehrecke, E. H., Blagosklonny, M. V., Dawson, T. M., Dawson, V. L., El-Deiry, W. S., Fulda, S. et al. (2011). Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ (epub). García-Arencibia, M., Hochfeld, W. E., Toh, P. P. C. and Rubinsztein, D. C. (2010). Autophagy, a guardian against neurodegeneration. Semin Cell Dev Biol 21, 691–698. Garcia-Ortega, A. M., Cañete, A., Quinter, C., Silberstein, L., PiquerGil, M., Alvarez-Dolado, M., Dekel, B., Göttgens, B. and Sánchez, M. J. (2010). Enhanced hematovascular contribution of SCL 3' enhancer expressing fetal liver cells uncovers their potential to integrate in extramedullary adult niches. Stem Cells 28, 100–112. Garstka, M. A., Fritzsche, S., Lenart, I., Hein, Z., Jankevicius, G., Boyle, L. H., Elliott, T., Trowsdale, J., Antoniou, A. N., Zacharias, M. and Springer, S. (2011). Tapasin dependence of major histocompatibility complex class I molecules correlates with their conformational flexibility. FASEB J 25, 3989–3998. George-Hyslop, P. S. and SchmittUlms, G. (2010). Selectively tuning !-secretase. Nature 467, 36–37. Gerold, K. D., Zheng, P. L., Rainbow, D. B., Zernecke, A., Wicker, L. S. and Kissler, S. (2011). The soluble CTLA-4 splice variant protects from type 1 diabetes and potentiates regulatory T-cell function. Diabetes 60, 1955–1963. Gerrish, A., Russo, G., Richards, A., Moskvina, V., Ivanov, D., Harold, D., Sims, R., Abraham, R., Hollingworth, P., Chapman, J. et al. (2011). The role of variation at A#PP, PSEN1, PSEN2, and MAPT in late onset Alzheimer’s Disease. J Alzheimers Dis (in press). Gibbons, D. L., Abeler-Dörner, L., Raine, T., Hwang, I. Y., Jandke, A., Wencker, M., Deban, L., Rudd, C. E., Irving, P. M., Kehrl, J. H. and Hayday, A. C. (2011). Cutting edge: regulator of g protein signaling-1 selectively regulates gut T cell trafficking and colitic potential. J Immunol 187, 2067–2071. Gordon, D. E., Bond, L. M., Sahlender, D. A. and Peden, A. A. (2010). A targeted siRNA screen to identify SNAREs required for constitutive secretion in mammalian cells. Traffic 11, 1191–1204. Göttgens, B., Ferreira, R., Sanchez, M. J., Ishibashi, S., Li, J., Spensberger, D., Lefevre, P., Ottersbach, K., Chapman, M., Kinston, S. et al. (2010). Cisregulatory remodeling of the SCL locus during vertebrate evolution. Mol Cell Biol 30, 5741–5751. Green, P., Wiseman, M., Crow, Y. J., Houlden, H., Riphagen, S., Lin, J. P., Raymond, F. L., Childs, A., Sheridan, E., Edwards, S. and Josifova, D. J. (2010). Brown-Vialetto-Van Laere syndrome, a Ponto-Bulbar Palsy with deafness, is caused by mutations in C20orf54. Am J Hum Genet 86, 485–489. Gribble, F. M. (2010). Alpha2Aadrenergic receptors and type 2 diabetes. N Engl J Med 362, 361–362. Griffiths, D. S., Li, J., Dawson, M. A., Trotter, M. W., Cheng, Y. H., Smith, A. M., Mansfield, W., Liu, P., Kouzarides, T., Nichols, J. et al. (2011). LIF- independent JAK signalling to chromatin in embryonic stem cells uncovered from an adult stem cell disease. Nat Cell Biol 13, 13–21. Griffiths, G. M., Tsun, A. and Stinchcombe, J. C. (2010). The immunological synapse: a focal point for endocytosis and exocytosis. J Cell Biol 189, 399–406. Gudgin, E. J. and Huntly, B. J. P. (2011). Acute myeloid leukemia: leukemia stem cells write a prognostic signature. Stem Cell Res Ther 2, 21. Hackett, A., Tarpey, P. S., Licata, A., Cox, J., Whibley, A., Boyle, J., Rogers, C., Grigg, J., Partington, M. Stevenson, R. E. et al. (2010). CASK mutations are frequent in males and cause X-linked nystagmus and variable XLMR phenotypes. Eur J Hum Genet 18, 544–552. Hall, I. P. and Lomas, D. A. (2010). The genetics of obstructive lung disease: big is beautiful. Thorax 65, 760–761. Halsall, S., Nicholas, A. K., Thornton, G., Martin, H. and Woods, C. G. (2010). Critical consequences of finding three pathogenic mutations in an individual with recessive disease. J Med Genet 47, 769–770. Hamilton-Williams, E. E., Wong, S. B. J., Martinez, X., Rainbow, D. B., Hunter, K. M., Wicker, L. S. and Sherman, L. A. (2010). Idd9.2 and Idd9.3 protective alleles function in CD4(+) T-cells and nonlymphoid cells to prevent expansion of pathogenic islet-specific CD8(+) T-cells. Diabetes 59, 1478–1486. Hannah, R., Joshi, A., Wilson, N. K., Kinston, S. and Göttgens, B. (2011). A compendium of genome-wide hematopoietic transcription factor maps supports the identification of gene regulatory control mechanisms. Exp Hematol 39, 531–541. Harbour, M. E., Breusegem, S. Y., Antrobus, R., Freeman, C., Reid, E. and Seaman, M. N. J. (2010). The cargo-selective retromer complex is a recruiting hub for protein complexes that regulate endosomal tubule dynamics. J Cell Sci 123, 3703–3717. Harrison, C. N., Bareford, D., Butt, N., Campbell, P., Conneally, E., Drummond, M., Erber, W., Everington, T., Green, A. R., Hall, G. W. et al. (2010). Guideline for investigation and management of adults and children presenting with a thrombocytosis. Br J Haematol 149, 352–375. Harwood, D. G., Kalechstein, A., Barker, W. W., Strauman, S., St George-Hyslop, P., Iglesias, C., Loewenstein, D. and Duara, R. (2010). The effect of alcohol and tobacco consumption, and apolipoprotein E genotype, on the age of onset in Alzheimer's disease. Int J Geriatr Psychiatry 25, 511–518. Heap, G. A., Yang, J. H., Downes, K., Healy, B. C., Hunt, K. A., Bockett, N., Franke, L., Dubois, P. C., Mein, C. A., Dobson, R. J. et al. (2010). Genomewide analysis of allelic expression imbalance in human primary cells by high-throughput transcriptome resequencing. Hum Mol Genet 19, 122–134. Heard, E., Tishkoff, S., Todd, J. A., Vidal, M., Wagner, G. P., Wang, J., Weigel, D. and Young, R. (2010). Ten years of genetics and genomics: what have we achieved and where are we heading? Nat Rev Genet 11, 723–733. Heinig, M., Petretto, E., Wallace, C., Bottolo, L., Rotival, M., Lu, H., Li, Y., Sarwar, R., Langley, S. R., Bauerfeind, A. et al. (2010). A trans-acting locus regulates an anti-viral expression network and type 1 diabetes risk. Nature 467, 460–464. Hersh, C. P., Pillai, S. G., Zhu, G. H., Lomas, D. A., Bakke, P., Gulsvik, A., DeMeo, D. L., Klanderman, B. J., Lazarus, R., Litonjua, A. A. et al. (2010). Multistudy fine mapping of chromosome 2q identifies XRCC5 as a chronic obstructive pulmonary disease susceptibility gene. Am J Respir Crit Care Med 182, 605–613. Hersh, C. P., Silverman, E. K., Gascon, J., Bhattacharya, S., Klanderman, B. J., Litonjua, A. A., Lefebvre, V., Sparrow, D., Reilly, J. J., Anderson, W. H., Lomas, D. A. and Mariani, T. J. (2011). SOX5 is a candidate gene for chronic obstructive pulmonary disease susceptibility and is necessary for lung development. Am J Respir Crit Care Med 183, 1482–1489. Heuser, M., Yun, H., Berg, T., Yung, E., Argiropoulos, B., Kuchenbauer, F., Park, G., Hamwi, I., Palmqvist, L., Lai, C. K. et al. (2011). Cell of origin in AML: susceptibility to MN1-induced transformation is regulated by the MEIS1/AbdB-like HOX protein complex. Cancer Cell 20, 39–52. Heuser, M., Yung, E., Yun, H., Berg, T., Argiropoulos, B., Kuchenbauer, F., Hamwi, I., Palmqvist, L., Lai, C. K., Leung, M. et al. (2011). The potent oncogenes MN1 and MEIS1 co-localize at a large proportion of their chromatin target sites suggestive of a higher order leukemogenic regulatory complex. Exp Hematol 39, S66–S67. Hiemstra, T. F., Charles, P. D., Hester, S. S., Karet, F. E.* and Lilley, K. S.* (2011). Uromodulin exclusion list improves urinary exosomal protein identification. J Biomol Techniques (in press). Higgins, J., Midgley, C., Bergh, A. M., Bell, S. M., Askham, J. M., Roberts, E., Binns, R. K., Sharif, S. M., Bennett, C., Glover, D. M., Woods, C. G., Morrison, E. E. and Bond, J. (2010). Human ASPM participates in spindle organisation, spindle orientation and cytokinesis. BMC Cell Biol 11, 85. Hirose, K., Inukai, T., Kikuchi, J., Furukawa, Y., Ikawa, T., Kawamoto, H., Oram, S. H. Göttgens, B., Kiyokawa, N., Miyagawa, Y. et al. (2010). Aberrant induction of LMO2 by the E2A-HLF chimeric transcription factor and its implication in leukemogenesis of B-precursor ALL with t(17;19). Blood 116, 962–970. 63 Hirst, J., Francisco, G. C., Sahlender, D. A., Seaman, M. N. J., Dacks, J. B. and Robinson, M. S. (2011). The fifth adaptor protein complex. PLoS Biol 9, e1001170. Hisadome, K., Reimann, F., Gribble, F. M. and Trapp, S. (2010). Leptin directly depolarizes preproglucagon neurons in the nucleus tractus solitarius electrical properties of glucagon-like peptide 1 neurons. Diabetes 59, 1890–1898. Hisadome, K., Reimann, F., Gribble, F. M. and Trapp, S. (2011). CCK stimulation of GLP-1 neurons involves alpha-1-adrenoceptor-mediated increase in glutamatergic synaptic inputs. Diabetes 60, 2701–2709. Höglinger, G. U., Melhem, N. M., Dickson, D. W., Sleiman, P. M. A., Wang, L., Klei, L., Rademakers, R., De Silva, R., Litvan, I., Riley, D. E. et al. (2011). Identification of common variants influencing risk of the tauopathy progressive supranuclear palsy. Nat Genet 43, 699–705. Hollingworth, P., Harold, D., Sims, R., Gerrish, A., Lambert, J. C., Carrasquillo, M. M., Abraham, R., Hamshere, M. L., Pahwa, J. S., Moskvina, V. et al. (2011). Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer's disease. Nat Genet 43, 429–435. Hopkins, T. G., Maher, E. R., Reid, E. and Marciniak, S. J. (2011). Recurrent pneumothorax. Lancet 377, 1624. Hov, J. R., Kosmoliaptsis, V., Traherne, J. A., Olsson, M., Boberg, K. M., Bergquist, A., Schrumpf, E., Bradley, J. A., Taylor, C. J., Lie, B. A., Trowsdale, J. and Karlsen, T. H. (2011). Electrostatic modifications of the human leukocyte antigen-DR P9 peptide-binding pocket and susceptibility to primary sclerosing cholangitis. Hepatol 53, 1967–1976. Howson, J. M., Rosinger, S., Smyth, D. J., Boehm, B. O., the ADBW-END Study Group and Todd, J. A. (2011). Genetic analysis of adult-onset 64 autoimmune diabetes. Diabetes 60, 2645–2653. Howson, J. M., Stevens, H., Smyth, D. J., Walker, N. M., Chandler, K. A., Bingley, P. J. and Todd, J. A. (2011). Evidence that HLA Class I and II associations with type 1 diabetes, autoantibodies to GAD and autoantibodies to IA-2, are distinct. Diabetes 60, 2635–2644. Hudson, T. J., Anderson, W., Aretz, A., Barker, A. D., Bell, C., Bernabe, R. R. and the International Cancer Genome Consortium. (2010). International network of cancer genome projects. Nature 464, 993–998. Huntington, J. A. (2011). Serpin structure, function and dysfunction. J Thromb Haemost 9, 26–34. Huntington, J. A. (2011). Thrombin plasticity. Biochimica et Biophysica Acta - Proteins and Proteomics (epub ahead of print). Huntington, J. A. and Whisstock, J. C. (2010). Molecular contortionism - on the physical limits of serpin 'loop-sheet' polymers. Biol Chem 391, 973–982. Huntington, J. A. and Yamasaki, M. (2011). Serpin polymerization in vitro. Methods Enzymol 501, 379–420. Hurst, J. R., Vestbo, J., Anzueto, A., Locantore, N., Mullerova, H., TalSinger, R., Miller, B., Lomas, D. A., Agusti, A., Macnee, W. et al. (2010). Susceptibility to exacerbation in chronic obstructive pulmonary disease. N Engl J Med 363, 1128–1138. Irving, J. A., Ekeowa, U. I., Belorgey, D., Haq, I., Gooptu, B., Miranda, E., Pérez, J., Roussel, B. D., Ordóñez, A., Dalton, L. E. et al. (2011). The serpinopathies studying serpin polymerization in vivo. Methods Enzymol 501, 421–466. Isachenko, V., Maettner, R., Petrunkina, A. M., Sterzik, K., Mallmann, P., Rahimi, G., Sanchez, R., Risopatron, J., Hancke, K., Damjanovski, I., Kreinberg, R. and Isachenko, E. (2011). Cryoprotectantfree vitrification of human spermatozoa in large (up to 0.5 ml) volume: a novel technology. Clin Lab 57, 643–650. Jackson, L. P., Kelly, B. T., McCoy, A. J., Gaffry, T., James, L. C., Collins, B. M., Honing, S., Evans, P. R. and Owen, D. J. (2010). A large-scale conformational change couples membrane recruitment to cargo binding in the AP2 clathrin adaptor complex. Cell 141, 1220–1229. Jahn, T. R., Kohlhoff, K. J., Scott, M., Tartaglia, G. G., Lomas, D. A., Dobson, C. M., Vendruscolo, M. and Crowther, D. C. (2011). Detection of early locomotor abnormalities in a Drosophila model of Alzheimer's disease. J Neurosci Methods 197, 186–189. Jahn, T. R., Malzer, E., Roote, J., Vishnivetskaya, A., Imarisio, S., Giannakou, M., Panser, K., Marciniak, S. and Crowther, D. C. (2011). Modeling serpin conformational diseases in Drosophila melanogaster. Methods Enzymol 499, 227–258. Jamieson, S. E.*, Peixoto-Rangel, A. L.*, Hargrave, A. C., de Roubaix, L. A., Mui, E. J., Boulter, N. R., Miller, E. N., Fuller, S. J., Wiley, J. S., Castellucci, L. et al. (2010). Evidence for association between purinergic receptor P2X7 (P2RX7) and congenital toxoplasmosis. Genes Immun 11, 374–383. Jayakumar, A., Castilho, T. M., Park, E., Goldsmith-Pestana, K., Blackwell, J. M. and McMahon-Pratt, D. (2011). TLR1/2 activation during heterologous prime-boost vaccination (DNA-MVA) enhances CD8+ T cell responses providing protection against Leishmania (Viannia). PLoS Neg Trop Dis 5, e1204. Jenkins, M. R. and Griffiths, G. M. (2010). The synapse and cytolytic machinery of cytotoxic T cells. Curr Opin Immunol 22, 308–313. Jimenez-Sanchez, M., Thompson, F., Zavodsky, E. and Rubinsztein, D. C. (2011). Autophagy and polyglutamine diseases. Prog Neurobiol (in press). Jin, H., Sanjo, N., Uchihara, T., Watabe, K., St George-Hyslop, P., Fraser, P. E. and Mizusawa, H. (2010). Presenilin-1 holoprotein is an interacting partner of sarco endoplasmic reticulum calciumATPase and confers resistance to endoplasmic reticulum stress. J Alzheimers Dis 20, 261–273. cholesterol metabolism in the aetiology of Alzheimer's disease. PLoS One 6. Jing, J. A., Junutula, J. R., Wu, C., Burden, J., Matern, H., Peden, A. A. and Prekeris, R. (2010). FIP1/ RCP binding to Golgin-97 regulates retrograde transport from recycling endosomes to the trans-Golgi network. Mol Biol Cell 21, 3041–3053. Jun, G., Naj, A. C., Beecham, G. W., Wang, L. S., Buros, J., Gallins, P. J., Buxbaum, J. D., Ertekin-Taner, N., Fallin, M. D., Friedland, R. et al. (2010). Meta-analysis confirms CR1, CLU, and PICALM as alzheimer disease risk loci and reveals interactions with APOE genotypes. Arch Neurol 67, 1473–1484. Johnson, D. J., Langdown, J. and Huntington, J. A. (2010). Molecular basis of factor IXa recognition by heparin-activated antithrombin revealed by a 1.7-A structure of the ternary complex. Proc Natl Acad Sci USA 107, 645–650. Jones, A. V., Campbell, P. J., Beer, P. A., Schnittger, S., Vannucchi, A. M., Zoi, K., Percy, M. J., McMullin, M. F., Scott, L. M., Tapper, W. et al. (2010). The JAK2 46/1 haplotype predisposes to MPL-mutated myeloproliferative neoplasms. Blood 115, 4517–4523. Jones, D. C., Kosmoliaptsis, V., Apps, R., Lapaque, N., Smith, I., Kono, A., Chang, C., Boyle, L. H., Taylor, C. J., Trowsdale, J. and Allen, R. L. (2011). HLA class I allelic sequence and conformation regulate leukocyte Ig-like receptor binding. J Immunol 186, 2990–2997. Jones, L., Holmans, P. A., Hamshere, M. L., Harold, D., Moskvina, V., Ivanov, D., Pocklington, A., Abraham, R., Hollingworth, P., Sims, R. et al. (2010). Genetic evidence implicates the immune system and cholesterol metabolism in the aetiology of Alzheimer's disease. PLoS One 5, e13950. Jones, L., Holmans, P. A., Hamshere, M. L., Harold, D., Moskvina, V., Ivanov, D., Pocklington, A., Abraham, R., Hollingworth, P., Sims, R. et al. (2011). Correction: genetic evidence implicates the immune system and Joshi, A. and Göttgens, B. (2011). Maximum parsimony analysis of gene expression profiles permits the reconstruction of developmental cell lineage trees. Dev Biol 353, 440–447. Kamath, P., Huntington, J. A. and Krishnaswamy, S. (2010). Ligand binding shuttles thrombin along a continuum of zymogen-and proteinase-like states. J Biol Chem 285, 28651–28658. Karanasios, E., Han, G. S., Xu, Z., Carman, G. M. and Siniossoglou, S. (2010). A phosphorylation-regulated amphipathic helix controls the membrane translocation and function of the yeast phosphatidate phosphatase. Proc Natl Acad Sci USA 107, 17539– 17544. Karet, F. E. (2011). Disorders of water and acid-base homeostasis. Nephron Physiol 118, 28–34. Karet, F. E. (2011). The renal tubular acidoses. In Netter's Textbook of the Kidney (in press). Keage, H. A., Matthews, F. E., Yip, A., Gao, L., McCracken, C., McKeith, I. G., Rubinsztein, D. C., Brayne, C. and the MRC Cognitive Function and Ageing Study. (2010). APOE and ACE polymorphisms and dementia risk in the older population over prolonged follow-up: 10 years of incidence in the MRC CFA Study. Age Ageing 39, 104–111. Kerzendorfer, C., Whibley, A., Carpenter, G., Outwin, E., Chiang, S. C., Turner, G., Schwartz, C., El-Khamisy, S., Raymond, F. L. and O'Driscoll, M. (2010). Mutations in Cullin 4B result in a human syndrome associated with increased camptothecin-induced topoisomerase I-dependent DNA breaks. Hum Mol Genet 19, 1324–1334. Kim, S. H., Fraser, P. E., Westaway, D., St George-Hyslop, P. H., Ehrlich, M. E. and Gandy, S. (2010). Group II metabotropic glutamate receptor stimulation triggers production and release of Alzheimer's amyloid(beta)42 from isolated intact nerve terminals. J Neurosci 30, 3870–3875. Klionsky, D. J., Baehrecke, E. H., Brumell, J. H., Chu, C. T., Codogno, P., Cuervo, A. M., Debnath, J., Deretic, V., Elazar, Z., Eskelinen, E. L. et al. (2011). A comprehensive glossary of autophagy-related molecules and processes. Autophagy 7, 1273–1294. Knecht, E., Aguado, C., Sarkar, S., Korolchuk, V. I., Criado-García, O., Vernia, S., Boya, P., Sanz, P., Rodríguez de Córdoba, S. and Rubinsztein, D. C. (2010). Impaired autophagy in Lafora disease. Autophagy 6, 991–993. Knezevic, K., Bee, T., Wilson, N. K., Janes, M. E., Kinston, S., Polderdijk, S., Kolb-Kokocinski, A., Ottersbach, K., Pencovich, N., Groner, Y. et al. (2011). A Runx1-Smad6 rheostat controls Runx1 activity during embryonic hematopoiesis. Mol Cell Biol 31, 2817–2826. Knobbe, C. B., Revett, T. J., Bai, Y., Chow, V., Jeon, A. H., Böhm, C., Ehsani, S., Kislinger, T., Mount, H. T., Mak, T. W., St George-Hyslop, P. H. and Schmitt-Ulms, G. (2011). Choice of biological source material supersedes oxidative stress in its influence on DJ-1 in vivo interactions with Hsp90. J Proteome Res 10, 4388–4404. Kelly, B. T. and Owen, D. J. (2011). Endocytic sorting of transmembrane protein cargo. Curr Opin Cell Biol 23, 404–412. 65 Kohlhoff, K. J., Jahn, T. R., Lomas, D. A., Dobson, C. M., Crowther, D. C. and Vendruscolo, M. (2011). The iFly tracking system for an automated locomotor and behavioural analysis of Drosophila melanogaster. Integr Biol (Camb) 3, 755–760. Kong, X. Y., Cho, M. H., Anderson, W., Coxson, H. O., Muller, N., Washko, G., Hoffman, E. A., Bakke, P., Gulsvik, A., Lomas, D. A. et al. (2011). Genomewide association study identifies BICD1 as a susceptibility gene for emphysema. Am J Respir Crit Care Med 183, 43–49. Koo, S., Huntly, B. J. P., Wang, Y. A., Chen, J., Brumme, K., Ball, B., McKinney-Freeman, S. L., Yabuuchi, A., Scholl, C., Bansal, D. et al. (2010). Cdx4 is dispensable for murine adult hematopoietic stem cells but promotes MLL-AF9-mediated leukemogenesis. Haematol-Hematol J 95, 1642–1650. Korolchuk, V. I. and Rubinsztein, D. C. (2011). Regulation of autophagy by lysosomal positioning. Autophagy 7, 927–928. Korolchuk, V. I., Menzies, F. M. and Rubinsztein, D. C. (2010). Mechanisms of cross-talk between the ubiquitinproteasome and autophagy-lysosome systems. FEBS Lett 584, 1393–1398. Korolchuk, V. I., Saiki, S., Lichtenberg, M., Siddiqi, F. H., Roberts, E. A., Imarisio, S., Jahreiss, L., Sarkar, S., Futter, M., Menzies, F. M. et al. (2011). Lysosomal positioning coordinates cellular nutrient responses. Nat Cell Biol 13, 453–460. Kozik, P., Francis, R. W., Seaman, M. N. J.* and Robinson, M. S.* (2010). A screen for endocytic motifs. Traffic 11, 843–855. Kremeyer, B., Lopera, F., Cox, J. J., Momin, A., Rugiero, F., Marsh, S., Woods, C. G., Jones, N. G., Paterson, K. J., Fricker, F. R. et al. (2010). A gainof-function mutation in TRPA1 causes familial episodic pain syndrome. Neuron 66, 671–680. Kvinlaug, B. T., Chan, W. I., Bullinger, L., Ramaswami, M., Sears, C., Foster, 66 D., Lazic. S. E., Okabe, R., Benner, A., Lee. B. H. et al. (2011). Common and overlapping oncogenic pathways contribute to the evolution of acute myeloid leukemias. Cancer Res 71, 4117–4129. D. A., Zhang, J., Basseres, D. S. et al. (2011). RUNX1 regulates the CD34 gene in haematopoietic stem cells by mediating interactions with a distal regulatory element. EMBO J 30, 4059–4070. Kwong, T., Robinson, C., Spencer, D., Norden, A., Shah, N., Wiseman, O. J., Karet, F. E. et al. (2010). Accuracy of urine pH testing in a regional Metabolic stone clinic. Is the dipstick accurate enough? J Endourol 24, A180. Li, J., Kent, D. G., Chen, E. and Green, A. R. (2011). Mouse models of myeloproliferative neoplasms: JAK of all grades. Dis Model Mech 4, 311–317. Lawrence, S. P., Bright, N. A., Luzio, J. P.* and Bowers, K.* (2010). The sodium/proton exchanger NHE8 regulates late endosomal morphology and function. Mol Biol Cell 21, 3540– 3551. Lechtenberg, B. C., Johnson, D. J., Freund, S. M. and Huntington, J. A. (2010). NMR resonance assignments of thrombin reveal the conformational and dynamic effects of ligation. Proc Natl Acad Sci USA 107, 14087–14092. Leddin, M., Perrod, C., Hoogenkamp, M., Ghani, S., Assi, S., Heinz, S., Wilson, N. K., Follows, G., Schönheit, J., Vockentanz, L. et al. (2011). Two distinct auto-regulatory loops operate at the PU.1 locus in B cells and myeloid cells. Blood 117, 2827–2838. Lee, J. H., Cheng, R., Barral, S., Reitz, C., Medrano, M., Lantigua, R., Jiménez-Velazquez, I. Z., Rogaeva, E., St George-Hyslop, P. H. and Mayeux, R. (2011). Identification of novel loci for Alzheimer disease and replication of CLU, PICALM, and BIN1 in Caribbean Hispanic individuals. Arch Neurol 68, 320–328. Lee, J. C., Lyons, P. A., McKinney, E. F., Sowerby, J. M., Carr, E. J., Bredin, F., Rickman, H. M., Ratlamwala, H., Hatton, A., Rayner, T. F., Parkes, M. and Smith, K. G. C. (2011). Gene expression profiling of CD8+ T cells predicts prognosis in patients with Crohn disease and ulcerative colitis. J Clin Invest 121, 4170–4179. Levantini, E., Lee, S., Radomska, H. S., Hetherington, C. J., Alberich-Jorda, M., Amabile, G., Zhang, P., Gonzalez, Li, J., Spensberger, D., Ahn, J. S., Anand, S., Beer, P. A., Ghevaert, C., Chen, E., Forrai, A., Scott, L. M., Ferreira, R. et al. (2010). JAK2 V617F impairs hematopoietic stem cell function in a conditional knock-in mouse model of JAK2 V617F-positive essential thrombocythemia. Blood 116, 1528–1538. Li, P., Burke, S., Wang, J., Chen, X., Ortiz, M., Lee, S. C., Lu, D., Campos, L., Goulding, D., Ng, B. L. et al. (2010). Reprogramming of T cells to natural killer-like cells upon Bcl11b deletion. Science 329, 85–89. Li, W., Johnson, D. J., Adams, T. E., Pozzi, N., De Filippis, V. and Huntington, J. A. (2010). Thrombin inhibition by serpins disrupts exosite II. J Biol Chem 285, 38621–38629. Lichtenberg, M., Mansilla, A., Zecchini, V. R., Fleming, A. and Rubinsztein, D. C. (2011). The Parkinson's disease protein LRRK2 impairs proteasome substrate clearance without affecting proteasome catalytic activity. Cell Death Dis 2, e196. Linterman, M. A., Pierson, W., Lee, S. K., Kallies, A., Kawamoto, S., Rayner, T. F., Srivastava, M., Divekar, D. P., Beaton, L., Hogan, J. J. et al. (2011). Foxp3(+) follicular regulatory T cells control the germinal center response. Nat Med 17, 975–982. Lipman, P. J., Cho, M. H., Bakke, P., Gulsvik, A., Kong, X. Y., Lomas, D. A., Anderson, W., Silverman, E. K. and Lange, C. (2011). On the follow-up of genome-wide association studies: an overall test for the most promising SNPs. Genet Epidemiol 35, 303–309. Liu, B., Moloney, A., Meehan, S., Morris, K., Thomas, S. E., Serpell, L. C., Hider, R., Marciniak, S. J., Lomas, D. A. and Crowther, D. C. (2011). Iron promotes the toxicity of amyloid beta peptide by impeding its ordered aggregation. J Biol Chem 286, 4248–4256. Liu, H., Purbhoo, M. A., Davis, D. M. and Rudd, C. E. (2010). SH2 domain containing leukocyte phosphoprotein of 76-kDa (SLP-76) feedback regulation of ZAP-70 microclustering. Proc Natl Acad Sci USA 107, 10166–10171. Llewellyn-Smith, I. J., Reimann, F., Gribble, F. M. and Trapp, S. (2011). Preproglucagon neurons project widely to autonomic control areas in the mouse brain. Neurosci 180, 111–121. Lomas, D. A. (2010). 25th anniversary Transatlantic Airway Conference: Protein misfolding and obstructive lung disease. Proc Am Thorac Soc 7, 343–345. Lugtenberg, D., Zangrande-Vieira, L., Kirchhoff, M., Whibley, A. C., Oudakker, A. R., Kjaergaard, S., Vianna-Morgante, A. M., Kleefstra, T., Ruiter, M., Jehee, F. S. et al. (2010). Recurrent deletion of ZNF630 at Xp11.23 is not associated with mental retardation. Am J Med Genet A 152A, 638–645. Luheshi, L. M., Hoyer, W., de Barros, T. P., van Dijk Härd, I., Brorsson, A. C., Macao, B., Persson, C., Crowther, D. C., Lomas, D. A., Ståhl, S., Dobson, C. M. Härd, T. (2010). Sequestration of the Abeta peptide prevents toxicity and promotes degradation in vivo. PLoS Biol 8, e1000334. Lumb, J. H., Connell, J. W., Allison, R. and Reid, E. (2011). The AAA ATPase spastin links microtubule severing to membrane modelling. Biochim Biophys Acta (epub). Luo, S. and Rubinsztein, D. C. (2010). Apoptosis blocks Beclin 1-dependent autophagosome synthesis: an effect rescued by Bcl-xL. Cell Death Differ 17, 268–277. Luzio, J. P., Gray, S. R. and Bright, N. A. (2010). Endosome-lysosome fusion. Biochem Soc Trans 38, 1413–1416. Mascarenhas, M. and Ottersbach, K. (2011). Role of cytokines in HSC emergence. Exp Hematol 39, S85. Lyons, P. A., McKinney, E. F., Rayner, T. F., Hatton, A., Woffendin, H. B., Koukoulaki, M., Freeman, T. C., Jayne, D. R. W., Chaudhry, A. N. and Smith, K. G. C. (2010). Novel expression signatures identified by transcriptional analysis of separated leucocyte subsets in systemic lupus erythematosus and vasculitis. Ann Rheum Dis 69, 1208–1213. Mason, M. J., Schaffner, C., Floto, R. A. and Teo, Q. A. (2011). Constitutive expression of a Mg2+ inhibited K+ current and a TRPM7-like current in human erythroleukemia cells. Am J Physiol Cell Physiol (epub). Malzer, E., Daly, M. L., Moloney, A., Sendall, T. J., Thomas, S. E., Ryder, E., Ryoo, H. D., Crowther, D. C., Lomas, D. A. and Marciniak, S. J. (2010). Impaired tissue growth is mediated by checkpoint kinase 1 (CHK1) in the integrated stress response. J Cell Sci 123, 2892–2900. Manes, T. D., Hoer, S., Muller, W. A., Lehner, P. J. and Pober, J. S. (2010). Kaposi's sarcoma-associated herpesvirus K3 and K5 proteins block distinct steps in transendothelial migration of effector memory CD4+T cells by targeting different endothelial proteins. J Immunol 184, 5186–5192. Marciniak, S. J. and Lomas, D. A. (2010). Alpha1-antitrypsin deficiency and autophagy. N Engl J Med 363, 1863–1864. Marcinak, S. J. and Ron, D. (2010). The unfolded protein response in lung disease. Proc Am Thorac Soc 7, 356–362. Marsh, S. G., Albert, E. D., Bodmer, W. F., Bontrop, R. E., Dupont, B., Erlich, H. A., Fernández-Viña, M., Geraghty, D. E., Holdsworth, R., Hurley, C. K. et al. (2010). An update to HLA nomenclature, 2010. Bone Marrow Transplant 45, 846–848. Marsh, S. G., Albert, E. D., Bodmer, W. F., Bontrop, R. E., Dupont, B., Erlich, H. A., Fernández-Viña, M., Geraghty, D. E., Holdsworth, R., Hurley, C. K. et al. (2010). Nomenclature for factors of the HLA system, 2010. Tissue Antigens 75, 291–455. McCoy, A. J. and Read, R. J. (2010). Experimental phasing: best practice and pitfalls. Acta Crystallogr D Biol Crystallogr 66, 458–469. McDowell, M. A., Johnson, S., Deane, J. E., Cheung, M., Roehrich, A. D., Blocker, A. J., McDonnell, J. M. and Lea, S. M. (2011). Structural and functional studies on the N-terminal domain of the Shigella type III secretion protein MxiG. J Biol Chem 286, 30606–30614. McEwan, W. A., Mallery, D. L., Rhodes, D. A., Trowsdale, J. and James, L. C. (2011). Intracellular antibody-mediated immunity and the role of TRIM21. BioEssays 33, 803–809. McGilvray, R. W., Eagle, R. A., Rolland, P., Jafferji, I., Trowsdale, J. and Durrant, L. G. (2010). ULBP2 and RAET1E NKG2D ligands are independent predictors of poor prognosis in ovarian cancer patients. Int J Cancer 127, 1412–1420. McKinney, E. F., Lyons, P. A., Carr, E. J., Hollis, J. L., Jayne, D. R., Willcocks, L. C., Koukoulaki, M., Brazma, A., Jovanovic, V., Kemeny, D. M. et al. (2010). A CD8+ T cell transcription signature predicts prognosis in autoimmune disease. Nat Med 16, 586–591. McLarren, K. W., Severson, T. M., du Souich, C., Stockton, D. W., Kratz, L. E., Cunningham, D., Hendson, G., Morin, R. D., Wu, D., Paul, J. E. et al. (2010). Hypomorphic temperaturesensitive alleles of NSDHL cause CK syndrome. Am J Hum Genet 87, 905–914. 67 Mduff, F. K., Hook, C. E., Tooze, R. M., Huntly, B. J. P., Pandolfi, P. P. and Turner, S. D. (2011). Determining the contribution of NPM1 heterozygosity to NPM-ALK-induced lymphomagenesis. Lab Invest 91, 1298–1303. Mehrotra, S., Oommen, J., Mishra, A., Sudharshan, M., Tiwary, P., Jamieson, S. E., Fakiola, M., Selvi Rani, D. S., Thangaraj, K., Rai, M., Sundar, S. and Blackwell. J. M. (2011). No evidence for association between SLC11A1 and visceral leishmaniasis in India. BMC Med Genet 12, 71. Menzies, F. M. and Rubinsztein, D. C. (2010). Broadening the therapeutic scope for rapamycin treatment. Autophagy 6, 286–287. Menzies, F. M., Hourez, R., Imarisio, S., Raspe, M., Sadiq, O., Chandraratna, D., O’Kane, C., Rock, K. L., Reits, E., Goldberg, A. L. and Rubinsztein, D. C. (2010). Puromycinsensitive aminopeptidase protects against aggregation-prone proteins via autophagy. Hum Mol Genet 19, 4573–4586. Menzies, F. M., Huebener, J., Renna, M., Bonin, M., Riess, O. and Rubinsztein, D. C. (2010). Autophagy induction reduces mutant ataxin-3 levels and toxicity in a mouse model of spinocerebellar ataxia type 3. Brain 133, 93–104. Menzies, F. M., Moreau, K. and Rubinsztein, D. C. (2011). Protein misfolding disorders and macroautophagy. Curr Opin Cell Biol 23, 190-197. Metcalf, D. J., García-Arencibia, M., Hochfeld, W. E. and Rubinsztein, D. C. (2010). Autophagy and misfolded proteins in neurodegeneration. Exp Neurol (epub). Miller, S. E., Sahlender, D., Graham, S. C., Hoening, S., Robinson, M. S., Peden, A. A.* and Owen, D. J.* (2011). The molecular basis for the endocytosis of small R-SNAREs by the clathrin adaptor CALM. Cell 147, 1118–1131. 68 Miranda, E., Pérez, J., Ekeowa, U. I., Hadzic, N., Kalsheker, N., Gooptu, B., Portmann, B., Belorgey, D., Hill, M., Chambers, S. et al. (2010). A novel monoclonal antibody to characterize pathogenic polymers in liver disease associated with alpha1-antitrypsin deficiency. Hepatol 52, 1078–1088. Miranda, K. C., Karet, F. E. and Brown, D. (2010). An extended nomenclature for mammalian V-ATPase subunit genes and splice variants. PLoS One 5, e9531. Mohammed, J. P., Fusakio, M. E., Rainbow, D. B., Moule, C., Fraser, H. I., Clark, J., Todd, J. A., Peterson, L. B., Savage, P. B., Wills-Karp, M. et al. (2011). Identification of Cd101 as a susceptibility gene for Novosphingobium aromaticivorans-induced liver autoimmunity. J Immunol 187, 337–349. Molloy, A., Nair, S., Cooke, F. J., Wain, J., Farrington, M., Lehner, P. J. and Torok, M. E. (2010). First report of Salmonella enterica serotype paratyphi A azithromycin resistance leading to treatment failure. J Clin Microbiol 48, 4655–4657. Moloney, A., Sattelle, D. B., Lomas, D. A. and Crowther, D. C. (2010). Alzheimer's disease: insights from Drosophila melanogaster models. Trends Biochem Sci 35, 228–235. Montenegro, G., Rebelo, A., Connell, J., Allison, R., Babalini, C., Schüle, R., Ishiura, H., D’Aloia, M., Montieri, P., Price, J. et al. (2012). Mutations in the ER-shaping protein Reticulon2 cause the axonal degenerative disorder Hereditary Spastic Paraplegia type 12. J Clin Invest (in press). Morahan, G., Mehta, M., James, I., Chen, W. M., Akolkar, B., Erlich, H. A., Hilner, J. E., Julier, C., Nerup, J., Nierras, C. et al. (2011). Tests for genetic interactions in type 1 diabetes linkage and stratification analyses of 4,422 affected sib-pairs. Diabetes 60, 1030–1040. Moreau, K., Luo, S. and Rubinsztein, D. C. (2010). Cytoprotective roles for autophagy. Curr Opin Cell Biol 22, 206–211. Moreau, K., Ravikumar, B., Renna, M., Puri, C. and Rubinsztein, D. C. (2011). Autophagosome precursor maturation requires homotypic fusion. Cell 146, 303–317. Moritoki, Y., Tsuda, M., Tsuneyama, K., Zhang, W., Yoshida, K., Lian, Z., Yang, G. X., Ridgway, W. M., Wicker, L. S., Ansari, A. A. and Gershwin, M. E. (2011). B cells promote hepatic inflammation, biliary cyst formation, and salivary gland inflammation in the NOD. c3c4 model of autoimmune cholangitis. Cell Immunol 268, 16–23. Morris, H., Morgan, M. D., Wood, A. M., Smith, S. W., Ekeowa, U. I., Herrmann, K., Holle, J. U., Guillevin, L., Lomas, D. A., Perez, J. et al. (2011). ANCA-associated vasculitis is linked to carriage of the Z allele of "1 antitrypsin and its polymers. Ann Rheum Dis 70, 1851–1856. Munuswamy-Ramanujam, G., Dai, E. B., Liu, L. Y., Shnabel, M., Sun, Y. M., Bartee, M., Lomas, D. A. and Lucas, A. R. (2010). Neuroserpin, a thrombolytic serine protease inhibitor (serpin), blocks transplant vasculopathy with associated modification of T-helper cell subsets. Thromb Haemost 103, 545–555. Murakami, T., Yang, S. P., Xie, L., Kawano, T., Fu, D., Mukai, A., Bohm, C., Chen, F., Robertson, J., Suzuki, H. et al. (2011). ALS mutations in FUS causes neuronal dysfunction and death in C. elegans by a dominant gain-offunction mechanism. Hum Mol Genet (epub). Naj, A. C., Jun, G., Beecham, G. W., Wang, L. S., Vardarajan, B. N., Buros, J., Gallins, P. J., Buxbaum, J. D., Jarvik, G. P., Crane, P. K. et al. (2011). Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer's disease. Nat Genet 43, 436–441. Narula, J., Smith, A. M., Göttgens, B. and Igoshin, O. A. (2010). Modeling reveals bistability and low-pass filtering in the network module determining blood stem cell fate. PLoS Comput Biol 6, 1–16. Nicholas, A. K., Khurshid, M., Désir, J., Carvalho, O. P., Cox, J. J., Thornton, G., Kausar, R., Ansar, M., Ahmad, W., Verloes, A. et al. (2010). Wdr62 is associated with the spindle pole and is mutated in human microcephaly. Nat Genet 42, 1010–1014. Niederer, H. A., Clatworthy, M. R., Willcocks, L. C. and Smith, K. G. C. (2010). FcgammaRIIB, FcgammaRIIIB, and systemic lupus erythematosus. Ann N Y Acad Sci 1183, 69–88. Niederer, H. A., Willcocks, L. C., Rayner, T. F., Yang, W., Lau, Y. L., Williams, T. N., Scott, J. A. G., Urban, B. C., Peshu, N., Dunstan, S. J. et al. (2010). Copy number, linkage disequilibrium and disease association in the FCGR locus. Hum Mol Genet 19, 3282–3294. Noor, A., Whibley, A., Marshall, C. R., Gianakopoulos, P. J., Piton, A., Carson, A. R., Orlic-Milacic, M., Lionel, A. C., Sato, D., Pinto, D. et al. (2010). Disruption at the PTCHD1 locus on Xp22.11 in autism spectrum disorder and intellectual disability. Sci Transl Med 2, 49ra68. Norwood, S. J., Shaw, D. J., Cowieson, N. P., Owen, D. J., Teasdale, R. D. and Collins, B. M. (2011). Assembly and solution structure of the core retromer protein complex. Traffic 12, 56–71. Ohashi, M., Eagle, R. A. and Trowsdale, J. (2010). Posttranslational modification of the NKG2D ligand RAET1G leads to cell surface expression of a glycosylphosphatidylinositol-linked isoform. J Biol Chem 285, 16408– 16415. Oracki, S. A., Tsantikos, E., Quilici, C., Light, A., Schmidt, T., Lew, A. M., Martin, J. E., Smith, K. G. C., Hibbs, M. L. and Tarlinton, D. M. (2010). CTLA4Ig alters the course of autoimmune disease development in Lyn(-/-) mice. J Immunol 184, 757–763. Oram, S. H., Thoms, J. A. I., Pridans, C., Janes, M. E., Kinston, S. J., Anand, S., Landry, J.-R., Lock, R. B., Jayaraman, P.-S., Huntly, B. J. P., Pimanda, J. E. and Göttgens, B. (2010). A previously unrecognized promoter of LMO2 forms part of a transcriptional regulatory circuit mediating LMO2 expression in a subset of T-acute lymphoblastic leukaemia patients. Oncogene 29, 5796–5808. Orlacchio, A., Babalini, C., Borreca, A., Patrono, C., Massa, R., Basaran, S., Munhoz, R. P., Rogaeva, E. A., St George-Hyslop, P. H., Bernardi, G. and Kawarai, T. (2010). SPATACSIN mutations cause autosomal recessive juvenile amyotrophic lateral sclerosis. Brain 133, 591–598. Ottersbach, K. and Dzierzak, E. (2010). The placenta as a haematopoietic organ. Int J Dev Biol 54, 1099–1106. Ottersbach, K., Mirshekar-Syahkal, B., Haak, E., Kimber, G. M., van Leusden, K., Harvey, K., O’Rourke, J., de Bruijn, M. F., Ferguson-Smith, A. C. and Dzierzak, E. (2011). DLK1 is a negative regulator of emerging hematopoietic stem cells. Exp Hematol 39, S24. Ottersbach, K., Smith, A., Wood, A. and Göttgens, B. (2010). Ontogeny of haematopoiesis: Recent advances and open questions. Br J Haematol 148, 343–355. Owen, D. J. and Collins, B. M. (2010). Vesicle transport: a new player in APP trafficking. Curr Biol 20, R413–R415. Papaemmanuil, E., Cazzola, M., Boultwood, J., Malcovati, L., Vyas, P., Bowen, D., Pellagatti, A., Wainscoat, J. S., Hellstrom-Lindberg, E., Gambacorti-Passerini, C. et al. (2011). Somatic mutation of SF3B1 in myelodysplasia with ring sideroblasts. N Engl J Med 365, 1384–1395. Parker, H. E., Reimann, F. and Gribble, F. M. (2010). Molecular mechanisms underlying nutrient-stimulated incretin secretion. Expert Rev Mol Med 12, e1. Parker, H. E., Wallis, K., le Roux, C. W., Wong, K. Y., Reimann, F. and Gribble, F. M. (2011). Molecular mechanisms underlying bile acid stimulated glucagon-like peptide-1 secretion. Br J Pharmacol (epub). Passamonti, F., Elena, C., Schnittger, S., Skoda, R. C., Green, A. R., Girodon, F., Kiladjian, J. J., McMullin, M. F., Ruggeri, M., Besses, C. et al. (2011). Molecular and clinical features of the myeloproliferative neoplasm associated with JAK2 exon 12 mutations. Blood 117, 2813–2816. Paul, D. S., Nisbet, J. P., Yang, T. P., Meacham, S., Rendon, A., Hautaviita, K., Tallila, J., White, J., Tijssen, M. R., Sivapalaratnam, S. et al. (2011). Maps of open chromatin guide the functional follow-up of genome-wide association signals: application to hematological traits. PLoS Genet 7, e1002139. Pellicano, F., Cilloni, D., Helgason, G. V., Messa, F., Panuzzo, C., Arruga, F., Bracco, E., Allan, E., Huntly, B. J. P., Holyoake, T. L. and Saglio, G. (2010). FOXO transcription factor activity is partially retained in quiescent CML stem cells and induced by tyrosine kinase inhibitors in CML progenitor cells. Blood 115, 2983. Persengiev, S., Koeleman, B. P. C., Downes, K., Valdigem, G., van der Slik, A. R., Eerligh, P., Monsuurb, A., Bruining, G. J., Wijmengae, C., Todd, J. A., Roep, B. A. and Alizadeh, B. Z. (2010). Association analysis of myosin IXB and type 1 diabetes. Hum Immunol 71, 598–601. Petrunkina, A. M. and Harrison, R. A. P. (2010). Systematic misestimation of cell subpopulations by flow cytometry: a mathematical analysis. Theriogenology 73, 839-847. Petrunkina, A. M. and Harrison, R. A. P. (2011). Cytometric solutions in veterinary andrology: Developments, advantages, and limitations. Cytometry A 79, 338–348. Petrunkina, A. M. and Harrison, R. A. P. (2011). Mathematical analysis of mis-estimation of cell subsets in flow cytometry: viability staining revisited. J Immunol Methods 368, 71–79. 69 Petrunkina, A. M., Waberski, D., Bollwein, H. and Sieme, H. (2010). Identifying non-sperm particles during flow cytometric physiological assessment: a simple approach. Theriogenology 73, 995–1000. Pettersen, J. A., Patry, D. G., St George-Hyslop, P. H. and Curry, B. (2011). Variant Alzheimer disease with spastic paraparesis: a rare presenilin-1 mutation. Can J Neurol Sci 38, 659–661. Pillai, S. G., Kong, X. Y., Edwards, L. D., Cho, M. H., Anderson, W. H., Coxson, H. O., Lomas, D. A., Silverman, E. K. and ECLIPSE and ICGN Investigators. (2010). Loci identified by genome-wide association studies influence different disease-related phenotypes in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 182, 1498–1505. Pimanda, J. E. and Göttgens, B. (2010). Gene regulatory networks governing haematopoietic stem cell development and identity. Int J Dev Biol 54, 1201–1211. Pimanda, J., Thoms, J., Birger, Y., Foster, S., Knezevic, K., Kirschenbaum, Y., Chandrakanthan, V., Lock, R., MacKenzie, K., Göttgens, B. and Izraeli, S. (2011). ERG promotes T-acute lymphoblastic leukemia and is transcriptionally regulated in leukemic cells by a stem cell enhancer. Exp Hematol 39, S108. Piper, R. C. and Lehner, P. J. (2011). Endosomal transport via ubiquitination. Trends Cell Biol 21, 647–655. Plagnol, V., Howson, J. M., Smyth, D. J., Walker, N., Hafler, J. P., Wallace, C., Stevens, H., Jackson, L., Simmonds, M. J., Type 1 Diabetes Genetics Consortium, Bingley, P. J., Goug, S. C. and Todd, J. A. (2011). Genome-wide association analysis of autoantibody positivity in type 1 diabetes cases. PLoS Genet 7, e1002216. Pociot, F., Akolkar, B., Concannon, P., Erlich, H. A., Julier, C., Morahan, G., Nierras, C. R., Todd, J. A., Rich, S. S. 70 and Nerup, J. (2010). Genetics of type 1 diabetes: what's next? Diabetes 59, 1561–1571. Pollen, D. A., Baker, S., Hinerfeld, D., Swearer, J., Evans, B. A., Evans, J. E., Caselli, R., Rogaeva, E. et al. (2010). Prevention of Alzheimer's disease in high risk groups: statin therapy in subjects with PSEN1 mutations or heterozygosity for apolipoprotein E epsilon 4. Alzheimers Res Ther 2, 31. Proitsi, P., Hamilton, G., Tsolaki, M., Lupton, M., Daniilidou, M., Hollingworth, P., Archera, N., Foya, C., Styliosa, F., McGuinness, B. et al. (2011). A multiple indicators multiple causes (MIMIC) model of behavioural and psychological symptoms in dementia (BPSD). Neurobiol Aging 32, 434–442. D. A. et al. (2011). Genetics of sputum gene expression in chronic obstructive pulmonary disease. PLoS One 6, e24395. Queiroz, J. W., Dias, G. D., Nobre, M. L., De Sousa Dias, M. C., Araújo, S. F., Barbosa, J. D., Trindade Neto, P., Blackwell, J. M. and Jeronimo, S. M. B. (2010). Geographic Information Systems and Applied Spatial Statistics are efficient tools to study Hansen’s Disease (leprosy) and to determine areas of greater risk of disease. Am J Trop Med Hyg 82, 306–314. Qutob, N., Balloux, F., Raj, T., Liu, H., Marion de Procé, S., Trowsdale, J. and Manica, A. (2011). Signatures of historical demography and pathogen richness on MHC class I genes. Immunogenet (epub). Proitsi, P., Lupton, M. K., Reeves, S. J., Hamilton, G., Archer, N., Martin, B. M., Iyegbea, C., Hollingworth, P., Lawlord, B., Gill, M. et al. (2010). Association of serotonin and dopamine gene pathways with behavioral subphenotypes in dementia. Neurobiol Aging (in press). Raab, M., Smith, X., Matthess, Y., Strebhardt, K. and Rudd, C. E. (2011). SKAP1 protein pH domain determines RapL membrane localization and Rap1 protein complex formation for T cell receptor (TCR) activation of LFA-1. J Biol Chem 286, 29663–29670. Purbhoo, M. A., Liu, H. B., Oddos, S., Owen, D. M., Neil, M. A. A., Pageon, S. V., French, P. M. W., Rudd, C. E. and Davis, D. M. (2010). Dynamics of subsynaptic vesicles and surface microclusters at the immunological synapse. Science Signal 3, ra36. Raab, M., Wang, H., Lu, Y., Smith, X., Wu, Z., Strebhardt, K., Ladbury, J. E. and Rudd, C. E. (2010). T cell receptor "inside-out" pathway via signaling module SKAP1-RapL regulates T cell motility and interactions in lymph nodes. Immun 32, 541–556. Puri, C., Chibalina, M. V., Arden, S. D., Kruppa, A. J., Kendrick-Jones, J. and Buss, F. (2010). Overexpression of myosin VI in prostate cancer cells enhances PSA and VEGF secretion, but has no effect on endocytosis. Oncogene 29, 188–200. Rainbow, D. B., Moule, C., Fraser, H. I., Clark, J., Howlett, S. K., Burren, O., Christensen, M., Moody, V., Steward, C. A., Mohammed, J. P. et al. (2011). Evidence that Cd101 is an autoimmune diabetes gene in nonobese diabetic mice. J Immunol 187, 325–336. Qi, X., Loiseau, F., Chan, W. L., Yan, Y., Wei, Z., Milroy, L. G., Myers, R. M., Ley, S. V., Read, R. J., Carrell, R. W. and Zhou, A. (2011). Allosteric modulation of hormone release from thyroxine and corticosteroid-binding globulins. J Biol Chem 286, 16163–16173. Ramasawmy, R., Menezes, E., Magalhães, A., Oliveira, J., Castellucci, L., Almeida, R., Rosa, M. E. A., Guimaraes, L. H., Lessa, M., Noronha, E. et al. (2010). The -2518 A/G promoter polymorphism in the gene (CCL2/MCP1) encoding monocyte chemotactic protein 1 influences susceptibility to mucosal but not localized cutaneous leishmaniasis in Brazil. Infect Genet Evol 10, 607–613. Qiu, W., Cho, M. H., Riley, J. H., Anderson, W. H., Singh, D., Bakke, P., Gulsvik, A., Litonjua, A. A., Lomas, Rashid, S. T., Corbineau, S., Hannan, N., Marciniak, S. J., Miranda, E., Alexander, G., Huang-Doran, I., Griffin, J., Ahrlund-Richter, L, Skepper, J. et al. (2010). Modeling inherited metabolic disorders of the liver using human induced pluripotent stem cells. J Clin Invest 120, 3127–3136. Ravikumar, B., Moreau, K. and Rubinsztein, D. C. (2010). Plasma membrane helps autophagosomes grow. Autophagy 6, 1184–1186. Ravikumar, B., Moreau, K., Jahreiss, L., Puri, C. and Rubinsztein, D. C. (2010). Plasma membrane contributes to the formation of pre-autophagosomal structures. Nat Cell Biol 12, 747–757. Ravikumar, B., Sarkar, S., Davies, J. E., Futter, M., Garcia-Arencibia, M., Green-Thompson, Z. W., JimenezSanchez, M., Korolchuk, V. I., Lichtenberg, M., Luo, S. et al. (2010). Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev 90, 1383–1435. Raymond, F. L., Whittaker, J., Jenkins, L., Lench, N. and Chitty, L. S. (2010). Molecular prenatal diagnosis: the impact of modern technologies. Prenat Diagn 30, 674–681. Read, R. J. (2010). From poor resolution to rich insight. Structure 18, 664–665. Read, R. J., Adams, P. D., Arendall, W. B., Brunger, A. T., Emsley, P., Joosten, R. P., Kleywegt, G. J., Krissinel, E. B., Lütteke, T., Otwinowski, Z. et al. (2011). A new generation of crystallographic validation tools for the protein data bank. Structure 19, 1395–1412. Read, R. J. and McCoy, A. J. (2011). Using SAD data in Phaser. Acta Crystallogr D Biol Crystallogr 67, 338–344. Reimann, F. (2010). Molecular mechanisms underlying nutrient detection by incretin-secreting cells. Int Dairy J 20, 236–242. Reimann, F., Cox, J. J., Belfer, I., Diatchenko, L., Zaykin, D. V., McHale, D. P., Drenth, J. P. H., Dai, F., Wheeler, J., Sanders, F. et al. (2010). Pain perception is altered by a nucleotide polymorphism in SCN9A. Proc Natl Acad Sci USA 107, 5148–5153. Reitz, C., Cheng, R., Rogaeva, E., Lee, J. H., Tokuhiro, S., Zou, F., Bettens, K., Sleegers, K., Tan, E. K., Kimura, R. et al. (2011). Meta-analysis of the association between variants in SORL1 and Alzheimer disease. Arch Neurol 68, 99–106. Reitz, C., Cheng, R., Schupf, N., Lee, J. H., Mehta, P. D., Rogaeva, E., St George-Hyslop, P. H. and Mayeux, R. (2010). Association between variants in IDE-KIF11-HHEX and plasma amyloid beta levels. Neurobiol Aging 33, 199.e13–199e17. Reitz, C., Tokuhiro, S., Clark, L. N., Conrad, C., Vonsattel, J. P., Hazrati, L. N., Palotás, A., Lantigua, R., Medrano, M., Jiménez-Velázquez, I. Z. et al. (2011). SORCS1 alters amyloid precursor protein processing and variants may increase Alzheimer's disease risk. Ann Neurol 69, 47–64. Renna, M., Jimenez-Sanchez, M., Sarkar, S. and Rubinsztein, D. C. (2010). Chemical inducers of autophagy that enhance the clearance of mutant proteins in neurodegenerative diseases. J Biol Chem 285, 11061–11067. Renna, M., Schaffner, C., Brown, K., Shang, S., Tamayo, M. H., Hegyi, K., Grimsey, N. J., Cusens, D., Coulter, S., Cooper, J. et al. (2011). Azithromycin blocks autophagy and may predispose cystic fibrosis patients to mycobacterial infection. J Clin Invest 121, 3554–3563. Renna, M., Schaffner, C., Winslow, A. R., Menzies, F. M., Peden, A. A., Floto, R. A. and Rubinsztein, D. C. (2011). Autophagic substrate clearance requires activity of the syntaxin-5 SNARE complex. J Cell Sci 124, 469–482. Rhodes, D. A., Boyle, L. H., Boname, J. M., Lehner, P. J. and Trowsdale, J. (2010). Ubiquitination of lysine-331 by Kaposi’s sarcoma-associated herpesvirus protein K5 targets HFE for lysosomal degradation. Proc Natl Acad Sci USA 107, 16240–16245. Riha, P. and Rudd, C. E. (2010). CD28 co-signaling in the adaptive immune response. Self/Nonself - Imm Recog Signal 1, 231–240. Roberts, R. C., Peden, A. A., Buss, F., Bright, N. A., Latouche, M., Reilly, M. M., Kendrick-Jones, J. and Luzio, J. P. (2010). Mistargeting of SH3TC2 away from the recycling endosome causes Charcot-Marie-Tooth disease type 4C. Hum Mol Genet 19, 1009–1018. Robin, C., Ottersbach, K., Boisset, J. C., Oziemlak, A. and Dzierzak, E. (2011). CD41 is developmentally regulated and differentially expressed on mouse hematopoietic stem cells. Blood 117, 5088–5091. Robinson, M. S., Sahlender, D. A. and Foster, S. D. (2010). Rapid inactivation of proteins by rapamycin-induced rerouting to mitochondria. Dev Cell 18, 324–331. Rodriguez-Cuenca, S., Cochemé, H. M., Logan, A., Abakumova, I., Prime, T. A., Rose, C., Vidal-Puiga, A., Smith, A. C., Rubinsztein, D. C., Fearnley, I. M. et al. (2010). Consequences of long-term oral administration of the mitochondriatargeted antioxidant MitoQ to wild-type mice. Free Radic Biol Med 48, 161–172. Rodriguez-Gonzalez, R., Millan, M., Sobrino, T., Miranda, E., Brea, D., de la Ossa, N. P., Blanco, M., Perez, J., Dorado, L., Castellanos, M., Lomas, D. A., Moro, M. A., Dávalos, A. and Castillo, J. (2011). The natural tissue plasminogen activator inhibitor neuroserpin and acute ischaemic stroke outcome. Thromb Haemost 105, 421–429. Rodriguez-Gonzalez, R., Sobrino, T., Rodriguez-Yanez, M., Millan, M., Brea, D., Miranda, E., Moldes, O., Pérez, J., Lomas, D. A., Leira, R., Dávalos, A. and Castillo, J. (2011). Association between neuroserpin and molecular markers of brain damage in patients with acute ischemic stroke. J Transl Med 9, 58. 71 Rogers, G. J., Tolhurst, G., Ramzan, A., Habib, A. M., Parker, H. E., Gribble, F. M.* and Reimann, F.* (2011). Electrical activity-triggered glucagon-like peptide-1 secretion from primary murine L-cells. J Physiol 589, 1081–1093. Rohr, J., Beutel, K., Maul-Pavicic, A., Vraetz, T., Thiel, J., Warnatz, K., Bondzio, I., Gross-Wieltsch, U., Schündeln, M., Schütz, B. et al. (2010). Atypical familial hemophagocytic lymphohistiocytosis due to mutations in UNC13D and STXBP2 overlaps with primary immunodeficiency diseases. Haematol-Hematol J 95, 2080–2087. Rose, C., Menzies, F. M., Renna, M., Acevedo-Arozena, A., Corrochano, S., Sadiq, O., Brown, S. D. and Rubinsztein, D. C. (2010). Rilmenidine attenuates toxicity of polyglutamine expansions in a mouse model of Huntington's disease. Hum Mol Genet 19, 2144–2153. Roussel, B. D., Irving, J. A., Ekeowa, U. I., Belorgey, D., Haq, I., Ordóñez, A., Kruppa, A. J., Duvoix, A., Rashid, S. T., Crowther, D. C., Marciniak, S. J. and Lomas, D. A. (2011). Unravelling the twists and turns of the serpinopathies. FEBS J 278, 3859–3867. Rubinsztein, D. C. (2010). Autophagy: where next? EMBO Rep 11, 3. Rubinsztein, D. C. (2010). Cdks regulate autophagy via Vps34. Mol Cell 38, 483–484. Rubinsztein, D. C. and Nixon, R. A. (2010). Rapamycin induces autophagic flux in neurons. Proc Natl Acad Sci USA 107, E181. Rubinsztein, D. C., Mariño, G. and Kroemer, G. (2011). Autophagy and aging. Cell 146, 682–695. Rudd, C. E. (2010). T-cell signaling and immunopathologies. Semin Immunopathol 32, 91–94. Rudd, C. E., Trevillyan, J. M., Dasgupta, J. D., Wong, L. L. and Schlossman, S. F. (2010). The CD4 receptor is complexed in detergent lysates to a protein-tyrosine kinase (pp58) from 72 human T lymphocytes (reprinted from Proc Nat Acad Sci 85, 5190–5194, 1988). J Immunol 185, 2645–2649. Rujirabanjerd, S., Nelson, J., Tarpey, P. S., Hackett, A., Edkins, S., Raymond, F. L., Schwartz, C. E., Turner, G, Iwase, S., Shi, Y., Futreal, P. A., Stratton, M. R. and Gecz, J. (2010). Identification and characterization of two novel JARID1C mutations: suggestion of an emerging genotype-phenotype correlation. Eur J Hum Genet 18, 330–335. St George-Hyslop, P. and Fraser, P. E. (2011). Assembly of the presenilin !-/'-secretase complex. J Neurochem (epub). St George-Hyslop, P. and SchmittUlms, G. (2010). Alzheimer's disease: Selectively tuning gamma-secretase. Nature 467, 36–37. Saleh, N. M., Raj, S. M., Smyth, D. J., Wallace, C., Howson, J. M., Bell, L., Walker, N. H., Stevens, H. E. and Todd, J. A. (2011). Genetic association analyses of atopic illness and proinflammatory cytokine genes with type 1 diabetes. Diabetes Metab Res Rev 27, 838–843. Samocha-Bonet, D., Wong, O., Synnott, E. L., Piyaratna, N., Douglas, A., Gribble, F. M., Holst, J. J., Chisholm, D. J. and Greenfield, J. R. (2011). Glutamine reduces postprandial glycemia and augments the glucagon-like peptide-1 response in type 2 diabetes patients. J Nutr 141, 1233–1238. Sanjo, N., Katayama, T., Hasegawa, H., Jin, H., Duthie, M., Mount, H. T., Mizusawa, H., St George-Hyslop, P. H. and Fraser, P. E. (2010). Localization and trafficking of endogenous anterior pharynx-defective 1, a component of Alzheimer's disease related gammasecretase. Neurosci Lett 483, 53–56. Sarkar, S., Korolchuk, V. I., Renna, M., Imarisio, S., Fleming, A., Williams, A., Garcia-Arencibia, M., Rose, C., Luo, S., Underwood, B. R. et al. (2011). Complex inhibitory effects of nitric oxide on autophagy. Mol Cell 43, 19–32. Saunders, A. M., Strittmatter, W. J., Schmechel, D., St George-Hyslop, P. H., Pericak-Vance, M. A., Joo, S. H., Rosi, B. L., Gusella, J. F., CrapperMacLachlan, D. R., Alberts, M. J., Hulette, C., Crain, B., Goldgaber, D. and Roses, A. D. (2011). Association of apolipoprotein E allele {varepsilon}4 with late-onset familial and sporadic Alzheimer's disease. Neurology 77, 950. Schiel, J. A., Park, K., Morphew, M. K., Reid, E., Hoenger, A. and Prekeris, R. (2011). Endocytic membrane fusion and buckling-induced microtubule severing mediate cell abscission. J Cell Sci 124, 1411–1424. Screaton, N. J., Miller, F. N., Patel, B. D., Groves, A., Tasker, A. D., Lomas, D. A. and Flower, C. D. (2011). The clinical impact of high resolution computed tomography in patients with respiratory disease. Eur Radiol 21, 225–231. Seaman, M. N. J. (2009). Enhanced SnapShot: Endosome-to-Golgi retrieval. Cell 139, 1198. Seaman, M. N. J. and Peden, A. A. (2010). Ricin toxin hits a retrograde roadblock. Cell 141, 222–224. Seki, N., Takahashi, Y., Tomiyama, H., Rogaeva, E., Murayama, S., Mizuno, Y., Hattori, N., Marras, C., Lang, A. E., George-Hyslop, P. S. et al. (2011). Comprehensive mutational analysis of LRRK2 reveals variants supporting association with autosomal dominant Parkinson's disease. J Hum Genet 56, 671–675. Settembre, C., Di Malta, C., Polito, V. A., Garcia Arencibia, M., Vetrini, F., Erdin, S., Erdin, S. U., Huynh, T., Medina, D., Colella, P., Sardiello, M., Rubinsztein, D. C. and Ballabio, A. (2011). TFEB links autophagy to lysosomal biogenesis. Science 332, 1429–1433. Shahid, H., Khan, J. C., Cipriani, V., Sepp, T., Matharu, B. K., Bunce, C., Harding, S. P., Clayton, D. G., Moore, A. T., Yates, J. R. W. for the Genetic Factors in AMD Study Group. (2011). Age-related macular degeneration: the importance of family history as a risk factor. Br J Ophthalmol (epub). Shoubridge, C., Tarpey, P. S., Abidi, F., Ramsden, S. L., Rujirabanjerd, S., Murphy, J. A., Boyle, J., Shaw, M., Gardner, A., Proos, A. et al. (2010). Mutations in the guanine nucleotide exchange factor gene IQSEC2 cause nonsyndromic intellectual disability. Nat Genet 42, 486–488. Siedlinski, M., Cho, M. H., Bakke, P., Gulsvik, A., Lomas, D. A., Anderson, W., Kong, X., Rennard, S. I., Beaty, T. H., Hokanson, J. E., Crapo, J. D. and Silverman, E. K. on behalf of the COPDGene Investigators and ECLIPSE Investigators. (2011). Genome-wide association study of smoking behaviours in patients with COPD. Thorax 66, 894–902. Sieni, E., Cetica, V., Mastrodicasa, E., Pende, D., Moretta, L., Griffiths, G. and. Aricò, M. (2011). Familial hemophagocytic lymphohistiocytosis: a model for understanding the human machinery of cellular cytotoxicity. Cell Mol Life Sci (epub). Sieni, E., Cetica, V., Santoro, A., Beutel, K., Mastrodicasa, E., Meeths, M., Ciambotti, B., Brugnolo, F., zur Stadt, U., Pende, D. et al. (2011). Genotype-phenotype study of familial haemophagocytic lymphohistiocytosis type 3. J Med Genet 48, 343–352. Silverman, E. K., Vestbo, J., Agusti, A., Anderson, W., Bakke, P. S., Barnes, K. C., Barr, R. G., Bleecker, E. R., Boezen, H. M., Burkart, K. M. et al. (2011). Opportunities and challenges in the Genetics of COPD 2010: An international COPD Genetics conference report. COPD: J Chron Obst Pulmon Dis 8, 121–135. Silverman, G. A., Whisstock, J. C., Bottomley, S. P., Huntington, J. A., Kaiserman, D., Luke, C. J., Pak, S. C., Reichhart, J.-M. and Bird, P. I. (2010). Serpins flex their muscle: I. Putting the clamps on proteolysis in diverse biological systems. J Biol Chem 285, 24299–24305. Sims, R., Dwyer, S., Harold, D., Gerrish, A., Hollingworth, P., Chapman, J., Jones, N., Abraham, R., Ivanov, D., Pahwa, J. S. et al. (2011). No evidence that extended tracts of homozygosity are associated with Alzheimer's disease. Am J Med Genet B Neuropsychiatr Genet 156, 764–771. Sin, D. D., Miller, B. E., Duvoix, A., Man, S. F., Zhang, X., Silverman, E. K., Connett, J. E., Anthonisen, N. A., Wise, R. A., Tashkin, D., Celli, B. R., Edwards, L. D., Locantore, N., MacNee, W., TalSinger, R. and Lomas, D. A. on behalf of the ECLIPSE Investigators. (2011). Serum PARC/CCL-18 concentrations and health outcomes in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 183, 1187–1192. Sir, J.-H., Barr, A. R., Nicholas, A. K., Carvalho, O. P., Khurshid, M., Sossick, A., Reichelt, S., D'Santos, C., Woods, C. G.*, Gergely, F.* (2011). A primary microcephaly protein complex forms a ring around parental centrioles. Nat Genet 43, 1147–1153. Smith, G. D., Robinson, C., Stewart, A. P., Edwards, E., Karet, H. I., Norden, A. G. W., Sandford, R N. and Karet Frankl, F. E. (2011). Characterization of a recurrent in-frame UMOD indel mutation causing late-onset autosomal dominant end-stage renal failure. Clin J Am Soc Nephrol (in press). Smith, I. A., Knezevic, B. R., Ammann, J. U., Rhodes, D. A., Aw, D., Palmer, D. B., Mather, I. H. and Trowsdale, J. (2010). BTN1A1, the mammary gland butyrophilin, and BTN2A2 are both inhibitors of T cell activation. J Immunol 184, 3514–3525. Smith, K. G. C. and Clatworthy, M. R. (2010). FcgammaRIIB in autoimmunity and infection: evolutionary and therapeutic implications. Nat Rev Immunol 10, 328–343. Smith, K. G. C., Willcocks, L. C. and Lyons, P. A. (2011). Comment on “the inhibiting Fc receptor for IgG, Fc!RIIB, is a modifier of autoimmune susceptibility”. J Immunol 187, 5473. Smyth, D. J., Cooper, J. D., Howson, J. M., Clarke, P., Downes, K., Mistry, T., Stevens, H., Walker, N. M. and Todd, J. A. (2011). FUT2 nonsecretor status links type 1 diabetes susceptibility and resistance to infection. Diabetes 60, 3081–3084. Sorheim, I. C., Bakke, P., Gulsvik, A., Pillai, S. C., Johannessen, A., Gaarder, P. I., Campbell, E. J., Agustí, A., Calverley, P. M. A., Donner, C. F. et al. (2010). alpha(1)-Antitrypsin protease inhibitor MZ heterozygosity is associated with airflow obstruction in two large cohorts. Chest 138, 1125–1132. Sorheim, I. C., DeMeo, D. L., Washko, G., Litonjua, A., Sparrow, D., Bowler, R., Bakke, P., Pillai, S. G., Coxson, H. O., Lomas, D. A., Silverman, E. K. and Hersh, C. P. (2010). Polymorphisms in the superoxide dismutase-3 gene are associated with emphysema in COPD. COPD 7, 262–268. Spruit, M. A., Polkey, M. I., Celli, B., Edwards, L. D., Watkins, M. L., PintoPlata, V., Vestbo, J., Calverley, P. M., Tal-Singer, R., Agusti, A. et al. (2011). Predicting outcomes from 6-minute walk distance in chronic obstructive pulmonary disease. J Am Med Dir Assoc (epub). Statham, H., Ponder, M., Richards, M., Hallowell, N. and Raymond, F. L. (2011). A family perspective of the value of a diagnosis for intellectual disability: Experiences from a genetic research study. Br J Learn Disab 39, 46–56. Stephens, P. J., Greenman, C. D., Fu, B., Yang, F., Bignell, G. R., Mudie, L. J., Pleasance, E. D., Lau, K. W., Beare, D., Stebbings, L. A. et al. (2011). Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144, 27–40. Stinchcombe, J. C., Salio, M., Cerundolo, V., Pende, D., Arico, M. and Griffiths, G. M. (2011). Centriole polarisation to the immunological synapse directs secretion from cytolytic cells of both the innate and adaptive immune systems. BMC Biol 9, 45. 73 Su, Y., Blake-Palmer, K. G., Fry, A. C., Best, A., Brown, A. C. N., Hiemstra, T. F., Horita, S., Zhou, A., Toye, A. M. and Karet, F. E. (2011). Glyceraldehyde 3-phosphate dehydrogenase is required for band 3 (anion exchanger 1) membrane residency in the mammalian kidney. Am J Physiol-Renal Physiol 300, F157-F166. Sun, G., Tarasov, A. I., McGinty, J., McDonald, A., Xavier, G. D., Gorman, T., Marley, A., French, P. M., Parker, H., Gribble, F. M. et al. (2010). Ablation of AMP-activated protein kinase alpha 1 and alpha 2 from mouse pancreatic beta cells and RIP2.Cre neurons suppresses insulin release in vivo. Diabetologia 53, 924–936. Swafford, A. D., Howson, J. M., Davison, L. J., Wallace, C., Smyth, D. J., Schuilenburg, H., Maisuria-Armer, M., Mistry, T., Lenardo, M. J. and Todd, J. A. (2011). An allele of IKZF1 (Ikaros) conferring susceptibility to childhood acute lymphoblastic leukemia protects against type 1 diabetes. Diabetes 60, 1041–1044. Swarbrick, J. D., Shaw, D. J., Chhabra, S., Ghai, R., Valkov, E., Norwood, S. J., Seaman, M. N. J. and Collins, B. M. (2011). VPS29 is not an active metallo-phosphatase but is a rigid scaffold required for retromer interaction with accessory proteins. PLoS One 6, e20420. Thomas, S. E., Dalton, L. E., Daly, M. L., Malzer, E. and Marciniak, S. J. (2010). Diabetes as a disease of endoplasmic reticulum stress. Diabetes Metab Res Rev 26, 611–621. Thomas, S. E., Dalton, L., Malzer, E. and Marciniak, S. J. (2011). Unravelling the story of protein misfolding in diabetes mellitus. World J Diabetes 2, 114–118. Thoms, J. A., Birger, Y., Foster, S., Knezevic, K., Kirschenbaum, Y., Chandrakanthan, V., Jonquieres, G., Spensberger, D., Wong, J. W., Oram, S. H. et al. (2011). ERG promotes T-acute lymphoblastic leukemia and is transcriptionally regulated in leukemic cells by a stem cell enhancer. Blood 117, 7079–7089. Tijssen, M. R., Cvejic, A., Joshi, A., Hannah, R. L., Ferreira, R., Forrai, A., Bellissimo, D. C., Oram, S. H., Smethurst, P. A., Wilson, N. K. et al. (2011). Genome-wide analysis of simultaneous GATA1/2, RUNX1, FLI1, and SCL binding in megakaryocytes identifies hematopoietic regulators. Dev Cell 20, 597–609. Swayamprakasam, A. P., Stover, E., Norgett, E., Blake-Palmer, K. G., Cunningham, M. J. and Karet, F. E. (2011). Importance of early audiologic assessment in distal renal tubular acidosis. Int Med Case Rep J 4, 7–11. Todd, J. A. (2010). D'oh! genes and environment cause Crohn's disease. Cell 141, 1114–1116. Taib, W. R. W., Smyth, D. J., Merriman, M. E., Dalbeth, N., Gow, P. J., Harrison, A. A., Highton, J., Jones, P. B. B., Stamp, L., Steer, S., Todd, J. A. and Merriman, T. R. (2010). The PTPN22 locus and rheumatoid arthritis: no evidence for an effect on risk independent of Arg620Trp. PLoS One 5, e13544. Todd, J. A., Knip, M. and Mathieu, C. (2011). Strategies for the prevention of autoimmune Type 1 diabetes. Diabet Med 28, 1141–1143. Tarpey, P., Thomas, S., Sarvananthan, N., Mallya, U., Lisgo, S., Talbot, C. J., 74 Roberts, E. O., Awan, M., Surendran, M., McLean, R. J. et al. (2011). Corrigendum: Mutations in FRMD7, a newly identified member of the FERM family, cause X-linked idiopathic congenital nystagmus. Nat Genet 4, 720. Todd, J. A. (2010). Etiology of type 1 diabetes. Immun 32, 457–467. Tolhurst, G., Zheng, Y., Parker, H. E., Habib, A. M., Reimann, F. and Gribble, F. M. (2011). Glutamine triggers and potentiates glucagon-like peptide-1 secretion by raising cytosolic Ca2+ and cAMP. Endocrinol 152, 405–413. Traherne, J. A., Martin, M., Ward, R., Ohashi, M., Pellett, F., Gladman, D., Middleton, D., Carrington, M. and Trowsdale, J. (2010). Mechanisms of copy number variation and hybrid gene formation in the KIR immune gene complex. Hum Mol Genet 19, 737–751. Trollet, C., Anvar, S. Y., Venema, A., Hargreaves, I. P., Foster, K., Vignaud, A., Ferry, A., Negroni, E., Hourde, C., Baraibar, M. A. et al. (2010). Molecular and phenotypic characterization of a mouse model of oculopharyngeal muscular dystrophy reveals severe muscular atrophy restricted to fast glycolytic fibres. Hum Mol Genet 19, 2191–2207. Trowsdale, J. (2011). The MHC, disease and selection. Immunol Lett 137, 1–8. Tsun, A., Qureshi, I., Stinchcombe, J. C., Jenkins, M. R., de la Roche, M., Kleczkowska, J., Zamoyska, R. and Griffiths, G. M. (2011). Centrosome docking at the immunological synapse is controlled by Lck signaling. J Cell Biol 192, 663–674. Underwood, B. R., Imarisio, S., Fleming, A., Rose, C., Krishna, G., Heard, P., Quick, M., Korolchuk, V. I., Renna, M., Sarkar, S., GarcíaArencibia, M., O'Kane, C. J., Murphy, M. P. and Rubinsztein, D. C. (2010). Antioxidants can inhibit basal autophagy and enhance neurodegeneration in models of polyglutamine disease. Hum Mol Genet 19, 3413–3429. van der Weyden, L., Giotopoulos, G., Rust, A. G., Matheson, L. S., van Delft, F. W., Kong, J., Corcoran, A. E, Greaves, M. F., Mullighan, C. G., Huntly, B. J. P. and Adams, D. J. (2011). Modeling the evolution of ETV6RUNX1-induced B-cell precursor acute lymphoblastic leukemia in mice. Blood 118, 1041–1051. van Ham, T. J., Holmberg, M. A., van der Goot, A. T., Teuling, E., Garcia-Arencibia, M., Kim, H. E., Du, D., Thijssen, K. L., Wiersma, M., Burggraaff, R. et al. (2010). Identification of MOAG-4/SERF as a regulator of age-related proteotoxicity. Cell 142, 601–612. Vestbo, J., Edwards, L. D., Scanlon, P. D., Yates, J. C., Agusti, A., Bakke, P., Calverley, P. M. A., Celli, B., Coxson, H. O., Crim, C., Lomas, D. A. et al. (2011). Changes in forced expiratory volume in 1 second over time in COPD. N Engl J Med 365, 1184–1192. Vezzoli, A., Bonadies, N., Allen, M. D., Freund, S. M., Santiveri, C. M., Kvinlaug, B. T., Huntly, B. J. P., Göttgens, B. and Bycroft, M. (2010). Molecular basis of histone H3K36me3 recognition by the PWWP domain of Brpf1. Nat Struct Mol Biol 17, 617–619. Vincent, B., Sunyach, C., Orzechowski, H. S., St GeorgeHyslop, P. and Checler, F. (2010). p53-dependent transcriptional control of cellular prion by presenilins. J Neurochem 115, 41–42. Virgin, H. W. and Todd, J. A. (2011). Metagenomics and Personalized Medicine. Cell 147, 44–56. Waberski, D., Henning, H. and Petrunkina, A. M. (2011). Assessment of storage effects in liquid preserved boar semen. Reprod Domest Anim 46, 45–48. Waisberg, M., Tarasenko, T., Vickers, B. K., Scott, B. L., Willcocks, L. C., Molina-Cruz, A., Piercea, M. A., Huangd, C., Torres-Veleze, F. J., Smith, K. G. C. et al. (2011). Genetic susceptibility to systemic lupus erythematosus protects against cerebral malaria in mice. Proc Natl Acad Sci USA 108, 1122–1127. Walker, B. A., Hunt, L. G., Sowa, A. K., Skjodt, K., Gobel, T. W., Lehner, P. J. and Kaufman, J. (2011). The dominantly expressed class I molecule of the chicken MHC is explained by coevolution with the polymorphic peptide transporter (TAP) genes. Proc Natl Acad Sci USA 108, 8396–8401. Wallace, C., Smyth, D. J., MaisuriaArmer, M., Walker, N. M., Todd, J. A. and Clayton, D. G. (2010). The imprinted DLK1-MEG3 gene region on chromosome 14q32.2 alters susceptibility to type 1 diabetes. Nat Genet 42, 68–71. Wan, E. S., Cho, M. H., Boutaoui, N., Klanderman, B. J., Sylvia, J. S., Ziniti, J. P., Won, S., Lange, C., Pillai, S. G., Anderson, W. H. et al. (2011). Genome-wide association analysis of body mass in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 45, 304–310. Wang, H., Lim, D. and Rudd, C. E. (2010). Immunopathologies linked to integrin signalling. Semin Immunopathol 32, 173–182. Wang, H., Lu, Y. and Rudd, C. E. (2010). SKAP1 is dispensable for chemokine-induced migration of primary T-cells. Immunol Lett 128, 148–153. Wang, J. G., Wicker, L. S. and Santamaria, P. (2009). IL-2 and its high-affinity receptor: Genetic control of immunoregulation and autoimmunity. Semin Immunol 21, 363–371. Wang, T. J., Zhang, F., Richards, J. B., Kestenbaum, B., van Meurs, J. B., Berry, D., Kiel, D. P., Streeten, E. A., Ohlsson, C., Koller, D. L. et al. (2010). Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet 376, 180–188. Weekes, M. P., Antrobus, R., Lill, J. R., Duncan, L. M., Hör, S. and Lehner, P. J. (2010). Comparative analysis of techniques to purify plasma membrane proteins. J Biomol Tech 21, 108–115. Weiss, J., Pyrski, M., Jacobi, E., Bufe, B., Willnecker, V., Schick, B., Zizzari, P., Gossage, S. J., Greer, C. A., LeindersZufall, T., Woods, C. G., Wood, J. N. and Zufall, F. (2011). Loss-of-function mutations in sodium channel Na(v)1.7 cause anosmia. Nature 472, 186–190. metazoan secretory pathway. EMBO J 29, 304–314. Wenham, M., Grieve, S., Cummins, M., Jones, M. L., Booth, S., Kilner, R., Ancliff, P. J., Griffiths, G. M. and Mumford, A. D. (2010). Two patients with Hermansky Pudlak syndrome type 2 and novel mutations in AP3B1. Haematologica 95, 333–337. Wheeler, D. W., Thompson, A. J., Corletto, F., Reckless, J., Loke, J. C., Lapaque, N., Grant, A. J., Mastroeni, P., Grainger, D. J., Padgett, C. L. et al. (2011). Anaesthetic impairment of immune function is mediated via GABA(A) receptors. PLoS One 6, e17152. Whibley, A. C., Plagnol, V., Tarpey, P. S., Abidi, F., Fullston, T., Choma, M. K., Boucher, C. A., Shepherd, L., Willatt, L., Parkin, G. et al. (2010). Finescale survey of X chromosome copy number variants and indels underlying intellectual disability. Am J Hum Genet 87, 173–188. Whibley, A., Urquhart, J., Dore, J., Willatt, L., Parkin, G., Gaunt, L., Black, G., Donnai, D. and Raymond, F. L. (2010). Deletion of MAOA and MAOB in a male patient causes severe developmental delay, intermittent hypotonia and stereotypical hand movements. Eur J Hum Genet 18, 1095–1099. Whisstock, J. C., Silverman, G. A., Bird, P. I., Bottomley, S. P., Kaiserman, D., Luke, C. J., Pak, S. C., Reichhart, J.-C. and Huntington, J. A. (2010). Serpins flex their muscle: II. Structural insights into target peptidase recognition, polymerization, and transport functions. J Biol Chem 285, 24307–24312. Wicker, L. S. (2010). Type 1 diabetes susceptibility genes: Expression and functional variations determined by genotype. Endocr J 57, S220. Wendler, F., Gillingham, A. K., Sinka, R., Rosa-Ferreira, C., Gordon, D. E., Franch-Marro, X., Peden, A. A., Vincent, J.-P. and Munro, S. (2010). A genome-wide RNA interference screen identifies two novel components of the 75 Willcocks, L. C., Carr, E. J., Niederer, H. A., Rayner, T. F., Williams, T. N., Yang, W., Scott, J. A. G., Urban, B. C., Peshub, N., Vyse, T. J., Lau, Y. L., Lyons, P. A. and Smith, K. G. C. (2010). A defunctioning polymorphism in FCGR2B is associated with protection against malaria but susceptibility to systemic lupus erythematosus. Proc Natl Acad Sci USA 107, 7881–7885. Willcocks, L. C., Lyons, P. A., Rees, A. J. and Smith, K. G. C. (2010). The contribution of genetic variation and infection to the pathogenesis of ANCAassociated systemic vasculitis. Arthritis Res Ther 12, 202. Wilson, N. K., Calero-Nieto, F. J., Ferreira, R. and Göttgens, B. (2011). Transcriptional regulation of haematopoietic transcription factors. Stem Cell Res Ther 2, 6. Wilson, N. K., Foster, S. D., Wang, X., Knezevic, K., Schütte, J., Kaimakis, P., Chilarska, P. M., Kinston, S., Ouwehand, W. H., Dzierzak, E. et al. (2010). Combinatorial transcriptional control in blood stem/progenitor cells: genome-wide analysis of ten major transcriptional regulators. Cell Stem Cell 7, 532–544. Wilson, N. K., Tijssen, M. R. and Göttgens, B. (2011). Deciphering transcriptional control mechanisms in hematopoiesis-The impact of highthroughput sequencing technologies. Exp Hematol 39, 961–968. Wilson, N. K., Timms, R. T., Kinston, S. J., Cheng, Y. H., Oram, S. H., Landry, J. R., Mullender, J., Ottersbach, K. and Göttgens, B. (2010). Gfi1 expression is controlled by five distinct regulatory regions spread over 100 kilobases, with Scl/Tal1, Gata2, PU.1, Erg, Meis1, and Runx1 acting as upstream regulators in early hematopoietic cells. Mol Cell Biol 30, 3853–3863. Wines, B. D., Trist, H. M., Farrugia, W., Ngo, C., Trowsdale, J., Areschoug, T., Lindahl, G., Fraser, J. D. and Ramsland, P. A. (2012). A conserved host and pathogen recognition site 76 on immunoglobulins: structural and functional aspects. Adv Exp Med Biol 946, 87–112. Winn, M. D., Ballard, C. C., Cowtan, K. D., Dodson, E. J., Emsley, P., Evans, P. R., Keegan, R. M., Krissinel, E. B., Leslie, A. G. W., McCoy, A. et al. (2011). Overview of the CCP4 suite and current developments. Acta Crystallogr D Biol Crystallogr 67, 235–242. Winslow, A. R. and Rubinsztein, D. C. (2011). The Parkinson disease protein "-synuclein inhibits autophagy. Autophagy 7, 429–431. Winslow, A. R., Chen, C. W., Corrochano, S., Acevedo-Arozena, A., Gordon, D. E., Peden, A. A., Lichtenberg, M., Menzies, F. M., Ravikumar, B., Imarisio, S., Brown, S., O’Kane, C. J. and Rubinsztein, D. C. (2010). "-Synuclein impairs macroautophagy: implications for Parkinson's disease. J Cell Biol 190, 1023–1037. Woods, C. G. (2011). Pain, pain genetics, and 'next-generation' pain genetics. Dev Med Child Neurol 53, 874–875. Yamanouchi, J., Puertas, M. C., Verdaguer, J., Lyons, P. A., Rainbow, D. B., Chamberlain, G., Hunter, K. M., Peterson, L. B., Wicker, L. S. and Santamaria, P. (2010). Idd9.1 locus controls the suppressive activity of FoxP3(+)CD4(+)CD25(+) regulatory T-cells. Diabetes 59, 272–281. Yamasaki, M., Sendall, T. J., Harris, L. E., Lewis, G. M. and Huntington, J. A. (2010). Loop-sheet mechanism of serpin polymerization tested by reactive center loop mutations. J Biol Chem 285, 30752–30758. Yamasaki, M., Sendall, T. J., Pearce, M. C., Whisstock, J. C. and Huntington, J. A. (2011). Molecular basis of "1-antitrypsin deficiency revealed by the structure of a domain-swapped trimer. EMBO Rep 12, 1011–1017. Yang, D. S., Stavrides, P., Mohan, P. S., Kaushik, S., Kumar, A., Ohno, M., Schmidt, S. D., Wesson, D. W., Bandyopadhyay, U., Jiang, Y. et al. (2011). Therapeutic effects of remediating autophagy failure in a mouse model of Alzheimer disease by enhancing lysosomal proteolysis. Autophagy 7, 788–789. Yang, D. S., Stavrides, P., Mohan, P. S., Kaushik, S., Kumar, A., Ohno, M., Schmidt, S. D., Wesson, D., Bandyopadhyay. U., Jiang. Y. et al. (2011). Reversal of autophagy dysfunction in the TgCRND8 mouse model of Alzheimer's disease ameliorates amyloid pathologies and memory deficits. Brain 134, 258–277. Yang, G., Wu, Y., Tsukamoto, H., Leung, P. S., Lian, Z., Rainbow, D. B., Hunter, K. M., Morris, G. A., Lyons, P. A., Peterson, L. B., Wicker, L. S., Gershwin, E. and Ridgway, W. M. (2011). CD8 T cells mediate direct biliary ductule damage in nonobese diabetic autoimmune biliary disease. J Immunol 186, 1259–1267. Yang, J. H., Downes, K., Howson, J. M., Nutland, S., Stevens, H. E., Walker, N. M. and Todd, J. A. (2011). Evidence of association with type 1 diabetes in the SLC11A1 gene region. BMC Med Genet 12, 59. Younis, R., Bingle, L. E. H., Rollauer, S., Munera, D., Busby, S. J., Johnson, S., Deane, J. E., Lea, S. M., Frankel, G. and Pallen, M. J. (2010). SepL resembles an aberrant effector in binding to a class 1 type III secretion chaperone and carrying an N-terminal secretion signal. J Bacteriol 192, 6093–6098. Yung, E., Sekulovic, S., Berg, T., Bilenky, M., Nielsen, C., Göttgens, B., Jones, S., Hirst, M. and Humphries, K. (2011). A comprehensive genome wide characterization of MEIS1 induced leukemic transformation. Exp Hematol 39, S45–S46. Yusa, K.*, Rashid, S. T.*, StrickMarchand, H., Varela, I., Liu, P.-Q., Paschon, D. E., Miranda, E., Ordóñez, A., Hannan, N., Rohani, F. et al. (2011). Targeted gene correction of "1-antitrypsin deficiency in induced pluripotent stem cells. Nature 478, 391–394. Zelmer, A., Martin, M. J., Gundogdu, O., Birchenough, G., Lever, R., Wren, B. W., Luzio, J. P. and Taylor, P. W. (2010). Administration of capsuleselective endosialidase E minimizes upregulation of organ gene expression induced by experimental systemic infection with Escherichia coli K1. Microbiology 156, 2205–2215. Zheng, S., Clabough, E. B., Sarkar, S., Futter, M., Rubinsztein, D. C. and Zeitlin, S. O. (2010). Deletion of the huntingtin polyglutamine stretch enhances neuronal autophagy and longevity in mice. PLoS Genet 6, e1000838. Zhou, A., Carrell, R. W., Murphy, M. P., Wei, Z., Yan, Y., Stanley, P. L., Stein, P. E., Broughton Pipkin, F. and Read, R. J. (2010). A redox switch in angiotensinogen modulates angiotensin release. Nature 468, 108–111. 77 The Wellcome Trust/MRC Building The Wellcome Trust/MRC Building which houses the Cambridge Institute for Medical Research and the MRC Mitochondrial Biology Unit. 78 Regulations for CIMR Management 1 The Cambridge Institute for Medical Research shall be an institution within the Faculty of Clinical Medicine and shall be under the general control of a Strategy Committee, which shall consist of: (a) the Director (Chairman) of the Institute; (b) the Deputy Director (Deputy Chairman) of the Institute; (c) the Regius Professor of Physic; (d) the Heads of the Departments from which staff working within the Institute are drawn; (e) the Director of the Diabetes and Inflammation Laboratory; (f) two persons appointed by the Faculty Board of Clinical Medicine. 2 The Regius Professor of Physic shall serve as Chairman of the Strategy Committee and the Deputy Director (Deputy Chairman) of the Institute shall serve as Secretary of the Strategy Committee. 3 Subject to the powers of the Council, the General Board, and the Faculty Board of Clinical Medicine, the duties of the Strategy Committee shall be as follows: (a) to promote research in, and at the interface of, the clinical and basic biomedical sciences that underpin the Institute’s major goal of determining and understanding the molecular mechanisms of disease; (b) to co-operate with outside bodies including the Wellcome Trust in the encouragement of such research; (c) to establish an Institute Management Committee and receive reports from it relating to the administration of funds allocated to the Institute for the purposes specified in (a) and (b) above and reports on the affairs of the Institute; (d) to convene such ad hoc or standing advisory groups as may be appropriate to support the Committee's work; (e) to nominate to the Faculty Board of Clinical Medicine for appointment or reappointment by that body the Director and Deputy Director. 4 There shall be an Institute Management Committee consisting of: (a) the Director (Chairman) of the Institute, who shall be Chairman of the Committee; (b) the Deputy Director (Deputy Chairman) of the Institute; (c) the Administrator of the Institute; (d) six Principal Investigators appointed by the Strategy Committee. The six principal investigators will serve on the Management Committee for periods not exceeding three years at any one time. The Management Committee may, with the agreement of the Strategy Committee, co-opt additional members. 5 The duties of the Management Committee shall be as follows: (a) to advise the Director (Chairman) of the Institute on strategic issues and implementation of strategy as agreed by the Strategy Committee and on other matters concerning the administration of the Institute including health and safety issues; (b) in consultation with the relevant Heads of Department, to select new Principal Investigators, using the criteria of scientific excellence and contribution to the aims of the Institute, and to approve applications from Principal Investigators wishing to seek extension of their externally funded fellowships; (c) to consider and make recommendations to the Director (Chairman) and Strategy Committee on allocation of space and resources; (d) to administer funds allocated to the Institute for the purposes specified in 3(a) above; (e) to formulate the Institute’s financial strategy, to prepare for the approval of the Faculty Board the Annual Estimates and year end reports, and applications to that Board for School funds; (f) to provide such data and reports as may be required by the Strategy Committee, the Faculty Board of Clinical Medicine and any outside bodies, including the Wellcome Trust; (g) to maintain records, to be updated at each meeting, of any developments in commercial exploitation, opportunities for the capture of IPR, or planned interactions with commercial companies, on the part of any Institute staff members and/or relating to research conducted by the Institute’s staff. Director (Chairman) of the Institute and Deputy Director (Deputy Chairman) of the Institute 2 There shall be a University office of Deputy Director (Deputy Chairman) of the Cambridge Institute for Medical Research, which may be held concurrently with another University office. 3 The Director (Chairman) and Deputy Director (Deputy Chairman) of the Cambridge Institute for Medical Research shall be appointed by the Faculty Board of Clinical Medicine on the recommendation of the Strategy Committee. Appointments and reappointments to the offices of Director (Chairman) and Deputy Director (Deputy Chairman) shall be for such periods not exceeding five years at a time as shall be determined by the Faculty Board on the recommendation of the Strategy Committee. 4 Under the general control of the Strategy Committee, and subject to the powers of the Management Committee, the Director (Chairman) of the Institute shall be the administrative Head of the Institute. 5 The Director (Chairman) or his or her nominated deputy shall also represent the Institute on the Wellcome Trust/MRC Building User’s Committee. Procedure for the appointment and reappointment of the Director (Chairman) and Deputy Director (Deputy Chairman) 1 The Strategy Committee shall designate one of their members to take soundings on their behalf from amongst the Heads of Departments from which staff of the Institute are drawn, the Principal Investigators based in the Institute, and to submit a nomination or nominations to the Strategy Committee. The Strategy Committee shall determine the nomination to be made to the Faculty Board of Clinical Medicine. For the Deputy Directorship the designated person shall be the Director unless the Strategy Committee shall determine otherwise. 2 This procedure shall apply also for re-appointments. Regulations approved by the Faculty Board of Clinical Medicine, University of Cambridge 1 There shall be a University office of Director (Chairman) of the Cambridge Institute for Medical Research, which may be held concurrently with another University office. (These regulations are currently under review, pending approval by the Faculty Board of Clinical Medicine) 79 Administrative, technical and other support staff Back cover illustration: A new “knocksideways” system, in which a coat protein, AP-2 (in red), gets rerouted to mitochondria in cells expressing a “Mitotrap” construct (green), while clathrin (blue) does not get rerouted. This rerouting occurs within seconds of addition of a small molecule, and causes the rerouted protein to be unavailable for its normal role and therefore non-functional. (Illustration courtesy of Margaret Robinson) www.cimr.cam.ac.uk