section home - The Gurdon Institute

Transcription

section home - The Gurdon Institute
The Wellcome Trust/Cancer Research UK
Gurdon Institute
2015 PROSPECTUS / ANNUAL REPORT 2014
Gurdon
INSTITUTE
PROSPECTUS 2015
ANNUAL REPORT 2014
http://www.gurdon.cam.ac.uk
CONTENTS
THE INSTITUTE IN 2014
INTRODUCTION
THE INSTITUTE IN 2014
INTRODUCTION........................................................................................................................................3
HISTORICAL BACKGROUND..........................................................................................................4
CENTRAL SUPPORT SERVICES....................................................................................................4
FUNDING.........................................................................................................................................................4
ENERGY AND ENVIRONMENT..........................................................................................................5
PUBLIC ENGAGEMENT..........................................................................................................5
POST-DOC ASSOCIATION..........................................................................................................5
RETREAT............................................................................................................................................................5
RESEARCH GROUPS.........................................................................................................6
MEMBERS OF THE INSTITUTE................................................................................42
CATEGORIES OF APPOINTMENT..............................................................................42
POSTGRADUATE OPPORTUNITIES..........................................................................42
SENIOR GROUP LEADERS.............................................................................................42
GROUP LEADERS.......................................................................................................................46
VISITING STUDENTS AND RESEARCHERS....................................................................47
ADMINISTRATION/SUPPORT STAFF........................................................48
INSTITUTE PUBLICATIONS.......................................................................................50
TALKS BY INSTITUTE RESEARCHERS.............................................................55
GURDON INSTITUTE SEMINAR SERIES..........................................................58
OTHER INFORMATION
STAFF AFFILIATIONS.............................................................................................................58
HONOURS AND AWARDS........................................................................................................59
EDITORIAL BOARDS OF JOURNALS.............................................................................59
INTERNATIONAL SCIENTIFIC ADVISORY BOARD...........................................59
CHAIRMAN OF MANAGEMENT COMMITTEE................................................59
LEAVERS DURING 2014................................................................................................60
ACKNOWLEDGEMENTS..............................................................Inside back cover
2
THE GURDON INSTITUTE
Much of our activity this year has been devoted to investigating ways that
the Gurdon Institute can expand to provide extra space for new research
directions. This effort has been stimulated in part by Tony Kouzarides’
sterling efforts to stimulate translational research in the University by
setting up a Therapeutics Centre as a platform for collaborations between
Cambridge academics and pharmaceutical companies. As our research
develops into novel areas, we also need more room for new facilities, such
as super-resolution imaging, genomics and tissue culture space for longterm organoid experiments. After much discussion and a generous offer
of financial support from Dr Jonathan Milner, a former member of the
Institute, the University has agreed to house the Therapeutics Centre within
a new building on the Cambridge Biomedical campus. We are delighted
that this new Centre will give a boost to translational research within
the University, as this has long been an area where the Gurdon Institute
has excelled. This was exemplified this year by the approval throughout
Europe and the USA of the ovarian cancer treatment Lynparza/Olaparib,
a drug initially developed by KuDOS, a start-up company founded by
Steve Jackson. Although the Therapeutic Centre will be based on the
Addenbrooke’s site, the Gurdon Institute will remain in the centre of town,
where we hope to build an extension to meet our other needs. Designing
and raising the funds for this venture will be a major goal for 2015.
The research in the Institute continues to flourish, and the outstanding
work of my colleagues was recognised with several prizes and awards
last year. Azim Surani was awarded a Jawaharlal Nehru Science Fellowship
by the Indian government to spend a year at the Institute for Stem
Cell Biology and Regenerative Medicine in Bangalore and also won the
International Society for Stem Cell Research McEwen Award for innovation;
John Gurdon was elected an honorary member of the Royal College of
Physicians, and received the Mike Hogg Award from the MD Anderson
Cancer Centre, Houston, Texas and an honorary degree from Rockefeller
University; Andrea Brand was made an honorary fellow of Brasenose
College, University of Oxford; Ben Simons was awarded the Franklin Medal
and Prize by the Institute of Physics; Steve Jackson was elected a Fellow
of the European Academy of Cancer Sciences and the Imperial College
Faculty of Medicine, and won the GSK Discovery Fast Track Challenge
with Dr. Delphine Larrieu; Meritxell Huch won the National Centre for
Replacement, Refinement and Reduction of Animals in Research (3Rs)
Prize for developing a method to grow mini-livers from adult stem cells; Phil
Zegerman was selected to join the EMBO Young Investigator programme
for the best young group leaders in Europe; Julie Ahringer and Nick Brown
were both awarded Professorships by the University, and Rick Livesey was
promoted to Senior Group Leader within the Gurdon Institute. Finally, the
Gurdon Institute received an Athena Swan Bronze Award “to recognise
and celebrate good practice in recruiting, retaining and promoting women
in SET within Higher Education.” Thanks are due to Julie Ahringer, Suzanne
Campbell, Ann Cartwright, Di Foster, Annabel Griffiths, Jon Pines and
Emmanuelle Vire on the Equality and Diversity committee for their hard
work in putting the application together and identifying ways to improve
the working environment for women in the Institute.
It was not just the group leaders who received awards this year and I
would like to commend Josep Foment (a postdoc in the Jackson group),
Laura Wagstaff (a postdoc in the Piddini group) and Helen Fox (a graduate student with Jenny Gallop) for winning prizes for the posters
they presented at conferences. I would also like to congratulate two
former members of the Institute: Ron Laskey (one of the founders of the
Institute) received a Cancer Research UK Lifetime Achievement Award for
“Challenging nuclear fiction: new roles for old nuclear proteins” and Sophie
Martin (a former PhD student in the St Johnston group, now a Professor at
the University of Lausanne) won the EMBO Gold Medal for “her research
to understand the organization and development of the cell”.
All of the research within the Institute depends on external grants, and
group leaders have been very successful at competing for funding this year.
Andrea Brand and Eric Miska have both received Wellcome Trust Senior
Investigator Awards to study “the nutritional control of neural stem-cell
quiescence and reactivation” and “transgenerational epigenetic inheritance:
adaptation, genome stability and evolution” respectively. Meritxell Huch,
who joined us in January, was awarded a Henry Dale Fellowship to
investigate the mechanisms of adult liver regeneration and also won a
Wellcome Trust Beit Prize Fellowship, which is given to the best researchers
at the beginning of their independent careers. Group leaders also secured
a number of other grants, including BBSRC grants to Andrea Brand and
Nick Brown. Most notably, Rick Livesey and his collaborators at the Institute
of Neurology (UCL) received an award of £2 million from Alzheimer’s
Research UK and the Alborada Trust to establish the Alzheimer’s Research
UK Stem Cell Research Centre, which was announced by the Prime
Minister in June at the G8 Dementia summit.
Under the excellent leadership of Dr Emmanuelle Vire, the Gurdon
Institute Postdoc Association (GIPA) has been playing an increasingly
important role in the life of the Institute. The past year was a particularly
active one for GIPA: it organised a one day symposium on “Building an
organism” with talks from Thomas Graf (CGR Barcelona), Greg Hannon
(Cold Spring Harbor Laboratory), Ewa Paluch (University College London)
and Jan-Michel Peters (IMP Vienna) and also hosted research seminars
from Susan Strome (University of California Santa Cruz), Andrew Jackson
(University of Edinburgh), Philippe Pasero (CNRS, Montpellier), Thijn
Brummelkamp (NCI, Amsterdam), Peter Campbell (The Sanger Institute)
and Jonathan Whetstine (Harvard Medical School, Boston). In addition,
GIPA organised six career pathways talks, in which former Gurdon Institute
postdocs described their careers outside academia. The highlight of the
postdoc calendar is the annual retreat and barbecue, which focused this
time on promoting new collaborations, with a speed-dating session that
used an algorithm developed by Rafael Carazo-Salas to pair up postdocs
with different research topics and expertise. This year also saw the launch
of an innovative postdoc mentoring programme, in which senior postdocs
mentor new postdocs and PhD students to help them settle into to
Cambridge and the Institute.
THE GURDON INSTITUTE
3
THE INSTITUTE IN 2014
Another person who deserves special mention this year is Hélène
Doerflinger, who does a fantastic job organising the Institute’s outreach
activities. Hélène launched the “Mobile Laboratory” this year, with
equipment funded by the Wellcome Trust Institutional Strategic Support
Fund. The Mobile Laboratory made four school visits in 2014, with a team
of Gurdon scientists going into local primary school classes to give year 5
and 6 children the chance to discover biology using real lab equipment. Our
outreach activities also cater for secondary school students, with six groups
of sixth-form students visiting the Gurdon Institute for two hour workshops
on “Cell division and cancer”, and fifteen students spending one week
shadowing an Institute postdoc or student to gain an insight into working in
a research laboratory. The highlight of the year was European Researchers’
Night at the Natural History Museum in London that was attended by
30,000 members of the public, at which Institute members ran a stand
explaining “What flies can tell us about human health”.
THE INSTITUTE IN 2014
The Institute is an integrated part of Cambridge University, and all group
leaders are also members of another University department within the
School of Biological Sciences, and contribute to both undergraduate and
graduate student teaching.
The University has also been generous in its support of the Institute,
particularly through various student and Herchel Smith schemes, and its
funding of equipment.
PUBLIC ENGAGEMENT
CENTRAL SUPPORT SERVICES
In 2014, the Gurdon Institute has undertaken many Public Engagement
activities for different audiences. With The Mobile Lab project, a team of
scientists ‘took a lab into local primary schools’ and ran a workshop using
microscopes. We organised workshops for sixth-form students to give them
the opportunity to visit the Institute and meet scientists. We welcomed
sixth-form students in our labs for work experience. We participated in the
Cambridge Alumni Festival. We organised core talks aimed at our nonscientist colleagues. We took part in the European Researchers’ Night at
the Natural History Museum, London.
The Institute’s ‘core staff ’ provides essential administrative, technical and
computing support to our scientists so that the scientists can spend as
much time as possible on their research.
The success of the Institute depends on all our core staff who do excellent
jobs supporting our research and keeping everything running smoothly.
It also depends on our great esprit de corps, I would like to thank the
many people who arrange various events during the year, particularly Ann
Cartwright, Suzanne Campbell, Emma Rawlins and Phil Zegerman for
organising the Institute retreat, the second-year PhD students for running
the weekly Happy Hours and the retreat treasure hunt, and the Social
Committee for parties. Finally, I am particularly grateful to Robb Krumlauf
and the rest of our International Scientific Advisory Board, who visited us
for two days in March to give us their usual dose of excellent advice and
support.
GURDON INSTITUTE POST-DOC ASSOCIATION
Grant income 1992 - 2014
Total number of staff (December 2014)
HISTORICAL BACKGROUND
The Institute was founded in 1989 to promote research in the areas of
developmental biology and cancer biology, and is situated in the middle of
the area containing the biological science departments of the University
of Cambridge, close to the newly-established Wellcome Trust Institute for
Stem Cell Research. The Institute hosts a number of independent research
groups in a purpose-built building designed to promote as much interaction
as possible. Developmental and cancer biology are complementary since
developmental biology is concerned with how cells, including stem cells,
acquire and maintain their normal function, whereas cancer is a result of a
cell breaking loose from its correct controls and becoming abnormal. Both
areas require a detailed knowledge of intra- and intercellular processes,
which need to be analysed at the scientific and technical levels. To
understand what goes wrong when a cell becomes cancerous requires
knowledge of the processes that ensure correct function in normal
development. At the technical level, the analysis of cellular and molecular
processes requires familiarity with techniques that no single person can
master, including molecular biology, biochemistry, microarray technology,
bioinformatics, cell culture, imaging and embryonic manipulations. There
is, therefore, a major benefit in having scientists with different but
complementary knowledge and technical skills working in close proximity
to one another as is the case in the Institute.
4
THE GURDON INSTITUTE
to boiling enough water to make 80 million cups of tea! We continue to
engage with the wider University community who wish to learn from our
achievements.
FUNDING
Our two major funding bodies, the Wellcome Trust and Cancer Research
UK, continue to offer the Institute vital backing in the form of Fellowships,
individual programme, project and equipment grants, in addition to our
invaluable core funding.
Other sources of funding, both direct and indirect, include the European
Commission, BBSRC, MRC, the Royal Society, NIH, the European
Molecular Biology Organization, HFSP, the Isaac Newton Trust, Alzheimer’s
Research UK, the Japan Society for the Promotion of Science, the March
of Dimes, the Erasmus Programme, the Amgen Scholars Programme, the
Darwin Trust, the Thai Government, the Liechtenstein Government, the
Turkish Government, the Cambridge Cancer Centre, Gates Cambridge
Scholarships, GSK, Boehringer Ingelheim, APART, Funai Foundation,
Worldwide Cancer Research, Dr Hadwen Trust, Swedish Society for
Medical Research, Taiwan Government, EPSRC, the Innovative Medicines
Initiative, Canadian Institutes of Health Research, Ataxia Telangiectasia
Society, Rubicon, Birax, Leukaemia and Lymphoma Research, Thouron
Award.
The Gurdon Institute Postdoc Association (GIPA) was established in 2008.
The aim of GIPA is to encourage scientific and social interactions, improve
postdocs’ welfare and provide researcher development opportunities. GIPA
organises a diverse set of events – annual Postdoc Retreats, Seminar Series
featuring world-leading scientists, Career Paths discussion panels, Mentoring
Scheme for junior postdocs and PhD students, and Happy Hours for the
whole Gurdon Institute. In 2014 GIPA organised the 1st Gurdon Institute
Postdoc Symposium, attended by 150 participants across the University,
and featuring 4 invited keynote talks, 10 short talks from early career
researchers and a poster session.
RETREAT
The 2014 Annual Retreat took place at the Five Lakes Hotel in Malden
on 1st and 2nd October. Our special guest was Sir Patrick Maxwell. The
Retreat is invaluable for both the exchange of scientific ideas and the social
opportunities that it provides.
Sources of funding 2014
ENERGY & ENVIRONMENT
In July we held a tea party to celebrate our first milestone achievement
of saving one million kWh of electricity since our energy saving campaign
launched in March 2012. Institute members continue to reduce energy
consumption, without affecting science, and they are key to the success
of our long-term aim to embed energy awareness within the Institute
culture. Our total savings to date are nearing 1.5 million kWh. This equates
Professor Daniel St Johnston
THE GURDON INSTITUTE
5
Julie Ahringer
The regulation of chromatin structure and function
Co-workers: Alex Appert, Fanelie Bauer, Ron Chen, Yan Dong, Csenge Gal, Carolina Gemma, Jürgen Jänes, Alicia McMurchy, Wei Qiang Seow,
Przemyslaw Stempor, Annette Steward, Christine Turner, Carson Woodbury, Brian Wysolmerski
Chromatin regulation plays a central role in the
determination and expression of cellular identity, and
chromatin disregulation is implicated in many diseases. We
use C. elegans to investigate the developmental regulation
of chromatin in transcription and genome organization.
Towards understanding the role and regulation of
chromatin during development, we have been mapping
the locations of accessible DNA, transcription initiation,
histone modifications, and chromatin regulators during
development in wild-type and chromatin mutants. We
found that that high promoter CpG density is a conserved
genomic signal for open chromatin. We are studying
the mechanism of CpG recognition and the function
of marking of these regions by H3K4me3. Mapping
transcription initiation revealed widespread bidirectional
initiation at C. elegans enhancers and promoters. We are
investigating the functions and developmental regulation
of enhancers and non-coding transcription, and their
relationship with promoters.
Within chromatin, particular sets of histone modifications
and/or chromatin proteins co-occur, and different
“chromatin states” are associated with different genomic
features. By generating and analysing a C. elegans
chromatin state map, we found that the genome is
organized into blocks of active and inactive chromatin
separated by boundary regions. We are investigating the
formation and function of different types of chromatin and
how this global genomic organization arises.
We also study the functions of C. elegans counterparts
of major chromatin regulatory complexes implicated
in human disease. In recent work, we found that
transcriptional repression by the Retinoblastoma/
DRM complex involves facilitating a high level of the
histone variant H2A.Z on the gene bodies of its targets.
These results suggest a new avenue for the study of
Retinoblastoma-related proteins.
6
THE GURDON INSTITUTE
Selected publications:
• Ho JW, 68 modENCODE consortium authors, Strome
S, Elgin SC, Liu XS, Lieb JD, Ahringer J, Karpen GH and Park
PJ (2014) Comparative analysis of metazoan chromatin
architecture. Nature, 512, 449-452
Fig 2. C. elegans CFP-1/CXXC1 is targeted to CpG-rich promoters marked by
H3K4me3
Fig 1. Genome-wide mapping of chromatin proteins and
histone modifications
• Weick E-M, Sarkies P, Silva N, Chen A-J, Moss SMM,
Cording AC, Ahringer J, Martinez-Perez E and Miska
EA (2014) PRDE-1 is a nuclear factor essential for the
biogenesis of Ruby motif- dependent piRNAs in C. elegans
Genes Dev, 28, 783-96
Fig 3. Chromatin states show that genes are organized
into active and inactive blocks
• Chen A-J, Stempor P, Down TA, Zeiser E, Feuer S and
Ahringer J (2014) Extreme HOT regions are CpG dense
promoters in C. elegans and human. Genome Research,
24: 1138-1146
• Chen A-J, Down TA, Stempor P, Chen QB, Egelhofer TA,
Hillier LW, Jeffers TE and Ahringer J (2013) The landscape
of RNA polymerase II transcription initiation in C. elegans
reveals enhancer and promoter architectures. Genome
Research, 8, 1339-47
Fig 4. Models for
enhancer functions
THE GURDON INSTITUTE
7
Andrea Brand
Stem cells to synapses: regulation of self-renewal and differentiation in the nervous system
Three-dimensional reconstruction of the blood-brain barrier glia (membranes in red) showing of the
glia overlying neural stem cells (grey).
Quiescent neural stem cells in the Drosophila central
nervous system (membranes in green; nuclei in blue).
Lola mutant neurons in the larval optic (labelled in green) dedifferentiate to a neural stem cell fate
and express Deadpan (labelled in red; arrowheads).
Without lola (green) to maintain repression, neurons
dedifferentiate, proliferate and form tumours.
Co-workers: Janina Ander, Josephine Bageritz, Elizabeth Caygill, Seth Cheetham, Melanie Cranston, Abhijit Das, Catherine Davidson, Paul Fox, Anna
Hakes, Jun Liu, Owen Marshall, Leo Otsuki, Chloe Shard, Pauline Spéder, Christine Turner, Jelle Van Den Ameele, Mo Zhao
Discovering how stem cells are maintained in a
multipotent state and how their progeny differentiate into
distinct cellular fates is a key step in the therapeutic use
of stem cells to repair tissues after damage or disease. We
are investigating the genetic networks that regulate neural
stem cell behaviour. Neural stem cells in the adult brain
exist primarily in a quiescent state but can be reactivated
in response to changing physiological conditions. How do
stem cells sense and respond to metabolic changes? In
the Drosophila central nervous system, quiescent neural
stem cells are reactivated synchronously in response to a
nutritional stimulus. We showed that feeding triggers insulin
production by blood-brain barrier glial cells, activating the
insulin/IGF pathway in underlying neural stem cells and
stimulating their growth and proliferation. More recently,
we discovered that gap junctions in the blood-brain barrier
glia mediate the influence of metabolic changes on stem
cell behaviour, enabling glia to respond to nutritional signals
and reactivate quiescent stem cells.
The ability to reprogram differentiated cells into a
pluripotent state has revealed that the differentiated state
is plastic and reversible. Mechanisms must be in place to
prevent neurons from dedifferentiating to a multipotent,
stem cell-like state. We discovered that the BTB-Zn
finger transcription factor, Lola, is required to maintain
neurons in a differentiated state. In lola mutants, neurons
dedifferentiate, turn on neural stem cell genes and begin
to divide, forming tumours. Thus, neurons rather than stem
cells or intermediate progenitors are the tumour-initiating
cells in lola mutants.
Cell-type specific transcriptional profiling is key to
understanding cell fate specification and function. We
developed ‘Targeted DamID’ (TaDa) to enable cell-specific
profiling without cell isolation. TaDa permits genome-wide
profiling of DNA- or chromatin-binding proteins without
cell sorting, fixation or affinity purification.
8
THE GURDON INSTITUTE
For more information, see the Brand lab home page:
http://www.gurdon.cam.ac.uk/research/brand
Selected publications:
• Spéder P and Brand AH (2014) Gap junction proteins
in the blood-brain barrier control nutrient-dependent
reactivation of Drosophila neural stem cells. Developmental
Cell 30, 309-321
• Southall TD, Davidson CM, Miller C, Carr A and Brand
AH (2014) Dedifferentiation of neurons precedes tumour
formation in lola mutants. Developmental Cell 28, 685-96
• Southall TD, Gold KS, Egger B, Davidson CM, Caygill
EE, Marshall OJ and Brand AH (2013) Cell type-specific
profiling of gene expression and chromatin binding
without cell isolation: Assaying RNA Pol II occupancy in
neural stem cells. Developmental Cell 26, 101-112
• Cheetham SW and Brand AH (2013) Insulin finds its
niche. Science 340, 817-818
• Chell JM and Brand AH (2010) Nutrition-responsive
glia control exit of neural stem cells from quiescence. Cell
143(7), 1161-1173
THE GURDON INSTITUTE
9
Nick Brown
Molecular analysis of morphogenesis
Co-workers: Natalia Bulgakova, Juan Manuel Gomez, Hannah Green, Benjamin Klapholz, Tarun Kumar, Miranda Landgraf, Aidan Maartens, John
Overton, Peerapat Thongneuk
Cellular adhesion and communication are vital during the
development of multicellular organisms. These processes
use proteins on the surface of cells, receptors, which
stick cells together (adhesion) and/or transmit signals
from outside the cell to the interior so that the cell can
respond to its environment. Our research is currently
focused on how adhesion receptors are linked with the
cytoskeleton to specify cell shape and movement within
the developing animal. This linkage between the adhesion
receptors and the major cytoskeletal filaments contains
many components, giving it the ability to grow or shrink
in response to numerous signals. For example, as the
cytoskeleton becomes contractile and exerts stronger
force on the adhesion sites, additional linker proteins are
recruited in to strengthen adhesion.
We use the fruit fly Drosophila as our model organism
to discover how the complex machinery linking cell
adhesion to the cytoskeleton works, and contributes
to morphogenesis. We are seeking to discover how
adhesion receptors form contacts of differing strength and
longevity, at one point mediating dynamic attachments as
the cell moves, and at another point stable connections
essential for the functional architecture of the body. A
good example of stable sites of adhesion is the integrindependent attachments of the muscles (Fig 1). Normally
integrin binding to extracellular ligands is the first step
in assembling a large complex of intracellular integrinassociated proteins into an adhesion complex. Of
particular interest are the mechanosensitive properties of
cell adhesion, where acto-myosin contraction with the cell
exerts force on sites of adhesion, causing the recruitment
of proteins like vinculin to strengthen adhesion. By
mimicking the action of force to generate an “activated”
vinculin, we find that this protein can trigger adhesion
complexes in the absence of integrins (Fig 2). We have
been puzzled by the apparent lack of contribution of some
integrin-associated proteins, as flies lacking these proteins
seemed normal. However, by examining the muscles
under the highest stress, the adult flight muscles, we are
10
THE GURDON INSTITUTE
discovering new phenotypes (Fig. 3). Using advanced
imaging methods including fluorescence lifetime imaging
and super-resolution (Fig. 4) is revealing how the adhesion
complex is assembled and functions.
Selected publications:
• Klapholz B, Herbert SL, Wellmann J, Johnson R, Parsons
M and Brown NH (2015) Alternative mechanisms for talin
to mediate integrin function. Curr Biol [in press]
• Maartens A and Brown NH (2015) The many faces of
cell adhesion during Drosophila muscle development. Dev
Biol [in press]
Fig 1. Attachment of muscles in the Drosophila embryo
is mediated by integrins, which are linked to the actin
cytoskeleton (red) by linker proteins such as vinculin (cyan).
• Huelsmann S, Ylänne J and Brown NH (2013) Filopodialike actin cables position nuclei in association with
perinuclear actin in Drosophila nurse cells. Dev Cell 26,
604-615.
Fig 3. New phenotypes of integrin-associated proteins are being revealed by examining
the highly-ordered flight muscles of the adult Drosophila (top normal [wild type], bottom
mutant). Actin is in magenta and tropomyosin in yellow.
• Bulgakova NA, Grigoriev I, Yap AS, Akhmanova A and
Brown NH (2013) Dynamic microtubules produce an
asymmetric E-cadherin-Bazooka complex to maintain
segment boundaries. J Cell Biol 201, 887-901.
• Bulgakova NA, Klapholz B and Brown NH (2012) Cell
adhesion in Drosophila: versatility of cadherin and integrin
complexes during development. Curr Opin Cell Biol 24,
702-712.
Fig 4. Super-resolution
view of the association
between integrins (cyan)
and actin (purple) at the
contacts between the
two epithelial cell layers
that make up the wing.
Fig 2. In the absence of integrins the muscles detach and round up
(actin is in red), but activated vinculin is able to bypass the normal
requirement for integrins to trigger the assembly of integrin-associated
proteins into complexes (cyan).
THE GURDON INSTITUTE
11
Jenny Gallop
Membranes, actin and morphogenesis
Co-workers: Guilherme Correia, Helen Fox, Lynn Froggett, Yoshiko Inoue, Iris Jarsch, Julia Mason, Daniel Saxton, Hanae Shimo
We are interested in the molecular basis of cell shape
and the changes that occur when cells move and tissues
develop. Cell shape is in large part determined by the
actin cytoskeleton and remodelling of the cytoskeleton
underlies the cell rearrangements that occur during
normal morphogenesis and also when morphogenetic
programs go wrong, for example in developmental defects
and during cancer metastasis. The machinery of the actin
cytoskeleton is also hijacked by various pathogens to
mediate infection.
Actin filaments are nucleated at cell membranes and are
elongated and bundled in different ways to form distinct
cytoskeletal structures. We have found that the membrane
environment influences which proteins are used to make
actin structures. Membranes are interesting to consider
in how cells change shape because they are the interface
between the outside and inside of the cell and therefore
are hubs of signalling activity, as well as being the boundary
of the cell that has to be moulded by links to the
cytoskeleton.
We are particularly concentrating on how actin
is polymerised during filopodia formation and
endocytosis (Fig 1). We take a two-pronged approach:
(1) reconstitution of actin polymerisation in vitro using
artificial membranes and Xenopus egg extracts (Fig 2)
and (2) investigation of how actin regulators are used by
cells in vivo in Drosophila melanogaster and during early
development in Xenopus laevis (Fig 3). This interdisciplinary
approach gives us the possibility of attaining a complete
molecular understanding and also testing those models
within the natural complement of physiological signals
provided by the whole organism.
Selected publications:
• Gallop JL, Walrant A, Cantley LC and Kirschner MW
(2013) Phosphoinositides and membrane curvature switch
the mode of actin polymerization via selective recruitment
of toca-1 and Snx9. Proc Natl Acad Sci 110: 7193-7198
Fig 1. Filopodia protrude from cells and are made of bundled actin,
vesicles bud inwards into cells and nucleate branched actin.
• Lee K*, Gallop JL*, Rambani K and Kirschner MW (2010)
Self-assembly of filopodia-like structures on supported
lipid bilayers. Science 329: 1341-1345
• Gallop JL*, Jao CC*, Kent HM, Butler PJ, Evans PR, Langen
R and McMahon HT (2006) Mechanism of endophilin
N-BAR domain-mediated membrane curvature. EMBO J
25: 2898-2910
• Gallop JL, Butler PJ and McMahon HT (2005) Endophilin
and CtBP/BARS are not acyl transferases in endocytosis or
Golgi fission. Nature 438: 675-678
• McMahon HT and Gallop JL (2005) Membrane
curvature and mechanisms of dynamic cell membrane
remodelling. Nature 438: 590-596
(* joint first authors)
Fig 2. Filopodia-like structures formed in vitro, with fluorescentlylabelled actin which grow from supported lipid bilayers.
Fig 3. Total internal reflection fluorescence microscopy
image of a Keller explant from a Xenopus gastrula,
showing that actin regulator Toca-1 localises to
lamellipodial edges, filopodia tips and endocytic vesicles.
12
THE GURDON INSTITUTE
THE GURDON INSTITUTE
13
John Gurdon
Nuclear reprogramming by oocytes and eggs
Co-workers: Dilly Bradford, Nigel Garrett, Eva Hormanseder, Jerome Jullien, Magdalena Koziol, Kei Miyamoto, Mami Oikawa, Angela Simeone,
Munender Vodnala, Ming-Hsuan Wen
The overall aim of our research programme is
to understand the mechanisms of somatic cell
reprogramming. When a somatic cell nucleus is
transplanted to an egg, it is within a few hours induced
to change its pattern of gene expression from that of
a somatic cell to that of an embryo. The same changes
occur when the highly condensed and specialised sperm
nucleus enters the egg at fertilisation and participation in
normal development. The difference is that nearly 100%
of sperm nuclei are reprogrammed perfectly whereas the
great majority of nuclei from specialised somatic cells make
this change imperfectly; they resist the reprogramming
effects of egg cytoplasm.
To analyse the mechanisms of reprogramming we
transplanted multiple somatic nuclei to the germinal
vesicles (nucleus) of an oocyte (the first meiotic prophase
progenitor of an egg). Nuclei transplanted to oocytes
do not divide or synthesise DNA, as they do when
transplanted to an egg, but nevertheless nearly all of them
are induced to undergo the first stages of new gene
expression.
During this year we have achieved a first full description of
major events that lead to reprogramming by oocytes. A
rapid sequence of steps is seen, at the single nucleus and
defined time course level, to lead in a hierarchical manner
to the binding and phosphorylation of RNA polymerase
II. The whole process is independent of new protein
synthesis.
We have also described the phenomenon of a “mitotic
advantage”, in which some genes in mitotic nuclei are up
to 100 times more efficiently reprogrammed by oocytes
than in similar somatic nuclei in interphase. We propose
that in normal development, mitosis is a stage in the
cell cycle when transcription factors in chromatin are
extensively exchanged leading to new directions of cell
differentiation.
14
THE GURDON INSTITUTE
Fig 1. New RNAs from transplanted reprogrammed nuclei.
Fig 2. Single nucleus analysis
of nuclear reprogramming
by oocytes.
Selected publications:
• Gurdon JB (2013) Secrets in the egg. Cell 153: 1179.
• Jullien J, Miyamoto K, Pasque V, Allen GE, Bradshaw CR,
Garrett NJ, Halley-Stott RP, Kimura H, Ohsumi K and
Gurdon JB (2014) Hierarchical molecular events driven
by oocyte-specific factors lead to rapid and extensive
reprogramming. Molecular Cell 55, 1-13.
• Halley-Stott RP, Jullien J, Pasque V and Gurdon JB (2014)
Mitosis gives a brief window of opportunity for a change in
gene transcription. PLoS Biology 12(7) e1001914.
(a)
(a)

(b)
• Christophorou MA, Castelo-Branco G, Halley-Stott
RP, Oliveira CS, Loos R, Radzisheuskaya A, Mowen KA,
Bertone P, Silva JCR, Zernicka-Goetz M, Nielsen ML,
Gurdon JB and Kouzarides T (2014) Citrullination regulates
pluripotency and histone H1 binding to chromatin. Nature
doi: 10.1038/nature12942.
(a)
• Teperek M, Miyamoto K, Simeone A, Feret, Deery,
Gurdon JB and Jullien J (2014) Sperm and spermatids
contain different proteins and bind distinct egg
factors. International Journal of Molecular Science
15(9):16719-16740.
Fig 4. Mitotic advantage
is maintained after
nuclear sonication.
Fig 3. (a) Design of mitotic advantage experiment.
(b) Mitotic chromatin is strongly reprogrammed to Sox2 transcription.
Fig 5. Procedure to achieve different levels of cell
permeabilization before nuclear transfer.
THE GURDON INSTITUTE
15
Meritxell Huch
Fig 1a
Fig 1b
Stem cells and tissue regeneration - implications in disease and cancer
Co-workers: Luigi Aloia, Laura Broutier, John Crang, Chris Hindley, Bart Kramer, Gianmarco Mastrogiovanni, Alessandra Merenda, Mikel Mckie
In adult mammals, self-renewal is required for the
maintenance of tissue homeostasis and tissue repair. In
organs with extensive self-renewal, such as the intestine
and stomach, adult stem cell populations are constantly
cycling to maintain cellular turnover. In organs with limited
proliferative capacity, though, such as the liver or pancreas,
we have recently described a population of stem/
progenitor cells that become activated exclusively upon
damage to repair the lost tissue and reinstall homeostasis.
However, the mechanism that regulates the activation
of the cells during regeneration, from the implication of
the niche to the epigenetic mechanisms regulating this
activation remains unsolved. One of our main goals is to
understand the mechanism of adult tissue regeneration,
using the liver and pancreas as model organs. Chronic liver
disease and liver and pancreas cancer are highly associated
to inflammation and tissue damage. Understanding the
mechanism regulating these processes holds promise to
extend our knowledge on tissue regeneration, disease and
cancer.
We are also interested in tissue engineering and disease
modelling. Despite the enormous regenerative capacity
of the liver in vivo, liver cells have resisted expansion in
culture. We have recently described a culture system
(liver organoid culture) that allows, for the first time,
the long-term (>1year) expansion of mouse liver stem/
progenitor cells into 3D structures that we have termed
‘liver organoids’. In this novel culture system adult liver
stem/progenitor cells maintain their ability of selfrenewal and differentiation towards functional liver cells.
When transplanted into a mouse model of liver disease
(FAH-/- mice), the cultured cells partially rescued the
liver phenotype, showing their therapeutic potential. We
have observed similar results using adult pancreas tissue.
Following on that discovery, we would now like to transfer
this technology to the study of liver diseases with the
aim of better understanding these and potentially finding
better therapeutic strategies.
16
THE GURDON INSTITUTE
Selected publications:
Fig 1c
• Huch M*, Bonfanti P*, Boj SF*, Sato T*, Loomans CJ, van
de Wetering M, Sojoodi M, Li VS, Schuijers J, Gracanin A,
Ringnalda F, Begthel H, Hamer K, Mulder J, van Es JH, de
Koning E, Vries RG, Heimberg H and Clevers H (2013)
Unlimited in vitro expansion of adult bi-potent pancreas
progenitors through the Lgr5/R-spondin axis. EMBO J
32(20):2708-21
• Huch M*, Dorrell C*, Boj SF, van Es JH, Li VSW, van
de Wetering M, Sato T, Hamer K, Sasaki N, Finegold MJ,
Haft A, Vries R, Grompe M and Clevers H (2013) In vitro
expansion of single Lgr5+ liver stem cells induced by Wntdriven regeneration. Nature 494: 247-50
• Barker N*, Huch M*, Kujala P, van de Wetering M,
Snippert HJ, van Es JH, Sato T, Stange DE, Begthel H, van
den Born M, Danenberg E, van den Brink S, Korving J, Abo
A, Peters PJ, Wright N, Poulsom R and Clevers H (2010)
Lgr5(+ve) stem cells drive self-renewal in the stomach
and build long-lived gastric units in vitro. Cell Stem Cell
6(1):25-36
(* denotes joint first authors)
Fig .1a–c, Lgr5-lacZ mice were injected i.p. with corn oil or CCl as in Fig.
1. Six days later, liver tissue was dissociated to single cells, loaded with
the fluorescent b-galactosidase (CMFDG) substrate and analysed by
fluorescent-activated cell sorting (FACS). Sorted isolated Lgr5-LacZ+ cells
were cultured at a ratio of onesingle Lgr5-LacZ+ cell per well (clonal) as
described in the Methods.
a, Scheme representing the protocol used.
b, Representative FACS plot of dissociatedsingle cells from CCl-treated
(with CCl) and non-treated (without CCl) livers. Cells were gated following
sequential selection by cell-size (forward scatter(FSC) versus side scatter
(SSC)) and propidium iodide (PI) exclusion. Viable CMFDG+PI− cells were
selected and sorted. Representative sorted cell is shown.
c, Serial differential interference contrast (DIC) images showing the
outgrowth of a single Lgr5-LacZ+ cell. Original magnifications were ×40
(days 0–5), ×20(day 7–11), ×10 (day 19) and ×4 (1 month onwards).
P, passage.
Fig 2. Human liver organoid derived from a human liver biopsy of
a healthy donor and grown for >4 months in culture
THE GURDON INSTITUTE
17
Steve Jackson
Maintenance of genome stability
Co-workers: Pallavi Agarwal, Gabriel Balmus, Linda Baskcomb, Rimma Belotserkovskaya, Andrew Blackford, Will Chiang, Julia Coates, Matt Cornwell,
Mukerrem Demir, Kate Dry, Josep Forment, Yaron Galanty, Nicola Geisler, Mareike Herzog, Satpal Jhujh, Delphine Larrieu, Carlos le Sage, Natalia
Lukashchuk, Francisco Muñoz-Martinez, Ryotaro Nishi, Fabio Puddu, Helen Reed, Israel Salguero, Christine Schmidt, Matylda Sczaniecka-Clift, Rohan
Sivapalan, David Weismann, Paul Wijnhoven
Our work focuses on the DNA-damage response (DDR),
the set of events that optimises cell survival and genome
integrity by detecting DNA damage, signalling its presence
and mediating its repair. As DDR defects are associated
with neurodegenerative diseases, immunodeficiencies,
premature ageing and cancer, our research is not only
providing academic insights but is also suggesting new ways
better to understand and alleviate such conditions.
This work extended the list of DUBs linked to the DDR
and highlighted their potential as cancer therapy targets.
One highlight for our research during the past year was us
identifying the small molecule “Remodelin” that improves
nuclear architecture, chromatin organisation and fitness of
human lamin A/C-depleted cells and cells from patients
with the premature-ageing disease Hutchinson-Gilford
progeria syndrome. These findings provided insights
into how the Remodelin target protein, NAT10, affects
nuclear architecture and suggested strategies for treating
laminopathies and cancer.
• Nishi R, Wijnhoven P, le Sage C, Tjeertes J, Galanty Y,
Forment JV, Clague MJ, Urbé S and Jackson SP (2014)
Systematic characterization of deubiquitylating enzymes for
roles in maintaining genome integrity. Nature Cell Biology
16, 1016-1026
In addition, we have shown that rolled-up transparent
microtubes can serve as cell culture scaffolds that precisely
define the space available for single cell growth. This
work established that the spatial confinement of mitotic
mammalian cells inside tubular architectures can perturb
metaphase plate formation, delay mitotic progression, and
cause chromosomal instability in both transformed and
nontransformed human cell lines. These findings could
provide important clues into how spatial constraints
dictate cellular behaviour and function.
Another particularly notable achievement for us in 2014
was our work systematically screening deubiquitylating
enzymes (DUBs) for roles in maintaining genome integrity.
We identified a number of DUBs with previously unknown
links to double-strand break (DSB) repair, the G2/M DNA
damage checkpoint and genome integrity maintenance.
Furthermore, we established that the DUB UCHL5
regulates DSB resection and homologous recombination
through protecting its interactor, NFRκB, from degradation.
18
THE GURDON INSTITUTE
Protein dynamics to and from sites of DNA breaks. DNA damage checkpoint and
repair factors and modulators of chromatin organization are recruited (green arrows)
to DNA breaks (SSB and DSB), while transcription machineries are excluded (red
arrows), and the dynamics of structural chromatin components operate in both
directions (orange arrows). HR, homologous recombination; NHEJ, non-homologous
end joining. Taken from Polo SE and Jackson SP (2011) Dynamics of DNA damage
response at DNA breaks: A focus on protein modifications. Genes Dev 25, 409-433
Selected publications:
• Larrieu D, Britton S, Demir M, Rodriguez R and Jackson
SP. (2014) Chemical inhibition of NAT10 corrects defects
of laminopathic cells. Science 344, 527-532
• Blasius M, Wagner SA, Choudhary C, Bartek J and
Jackson SP (2014) A quantitative 14-3-3 interaction screen
connects the nuclear exosome targeting complex to the
DNA damage response. Genes and Development 28,
1977-1982
Rolled up functionalised nanomembranes as three-dimensional
cavities for single cell studies
• Xi W, Schmidt CK*, Sanchez S*, Gracias DH, CarazoSalas RE, Jackson SP and Schmidt OG (2014) Rolled-up
functionalized nanomembranes as three-dimensional
cavities for single cell studies. NANOLetters 14(8) 41974204 *Co-corresponding authors
Image shows a laminopathic cell with
a mis-shapen nucleus before (left) and
after (right) chemical inhibiton of the
N-acetyl transferase NAT10.
THE GURDON INSTITUTE
19
Tony Kouzarides
Epigenetic modifications and cancer
Co-workers: Andrej Alendar, Paulo Amaral, Andy Bannister, Isaia Barbieri, Ester Cannizzaro, Ka Hing (Harvey) Che, Ali Cook, Namshik Han, Sri
Lestari, Nikki Mann, Valentina Migliori, Gonzalo Millan Zambrano, Sam Robson, Helena Santos Rosa, Meike Wiese
Our group is interested in defining the mechanisms
by which modifications of chromatin and non-coding
(nc) RNAs regulate cellular processes. Our attention
is focused on enzymes which regulate transcription by
covalently modifying histones or ncRNAs. We would like
to understand what biological processes these enzymes
control and the precise mechanism by which modifications
act. At the same time we are dissecting how modification
pathways are mis-regulated in cancer cells and exploring
avenues for treatment.
Our recent work has identified two new modification
pathways. The first involves methylation of miRNA 145 by
a new RNA modifying enzyme BCDN3D. This methylation
disrupts the binding of miRNA 145 to dicer and therefore
controls miRNA maturation. The BCDN3D enzyme is an
oncogene with pro-metastatic characteristics, indicating
that this pathway may be therapeutically important.
The second pathway involves a new class of chromatin
modifying enzyme, which is able to methylate a glutamine
residue within H2A. This modification is restricted to
the rDNA locus and has a role in transcription by RNA
polymerase I. We have also continued the characterisation
of arginine cirtrullination by the Padi4 enzyme, a
modification we described some years ago. We can now
show that this enzymatic activity has a role in pluripotency.
Our interest in the intervention of epigenetic pathways
involved in cancer has identified the acetyl-binding BET
proteins as a therapeutic target. A small molecule inhibitor
of BETs (I-BET) was used to prevent the binding of BET
proteins to acetylated histones and suppress a gene
expression program leading to MLL-leukaemia. This small
molecule effectively inhibits primary human leukaemias
and halts the process of leukaemia in model systems. I-BET
is currently in clinical trials.
20
THE GURDON INSTITUTE
Fig 1. Citrullination by PADI4 regulates pluripotency by
causing chromatin de-compaction
Selected publications:
• Christophorou M, Castelo-Branco G, Halley-Stott R,
Slade Oliveira C, Loos R, Bertone P, Silva J, Zernicka-Goetz
M, Nielsen M, Gurdon JB, Radzisheuskaya A, Mowen M and
Kouzarides T (2014) Citrullination regulates pluripotency
and H1 linker histone binding to chromatin. Nature
507(7490), 104-108
• Tessarz P, Santos-Rosa H, Robson SC, Sylvestersen KB,
Nelson CJ, Nielsen ML and Kouzarides T (2014) Glutamine
methylation in Histone H2A is an RNA Polymerase I
dedicated modification. Nature 505(7484), 564-568
• Xhemalce B, Robson S and Kouzarides T (2012) Human
RNA Methyltransferase BCDIN3D Regulates MicroRNA
Processing. Cell 151(2), 278-288
• Dawson M, Kouzarides T et al (2011) Inhibition of BET
recruitment to chromatin as an effective treatment for
MLL-fusion leukaemia. Nature 478(7370), 529-533
Fig 2. Glutamine methylation of H2A by the human Fibrillarin enzyme
is localised in the nucleus and regulates rDNA transcription.
• Bartke T, Vermeulen M, Xhemalce B, Robson SC, Mann
M and Kouzarides T (2010). Nucleosome-interacting
Proteins Regulated by DNA and Histone Methylation. Cell 143: 470 – 84
Fig 3. The small molecule I-BET displaces BET proteins
and represses genes that cause MLL-leukaemia.
THE GURDON INSTITUTE
21
Rick Livesey
Mammalian neural stem cell biology, fundamental and applied
Co-workers: Philipp Berg, Laura Brightman, Philip Brownjohn, Tatyana Dias, lewis Evans, Jayne Fisher, Alberto Frangini, Teresa Krieger, Ayiba Momoh,
Steven Moore, Tomoki Otani, Nathalie Saurat, James Smith, Victoria Stubbs
The cerebral cortex, which makes up three quarters
of the human brain, is the part of the nervous system
that integrates sensations, executes decisions and is
responsible for cognition and perception. Given its
functional importance, it is not surprising that diseases of
the cerebral cortex are major causes of morbidity and
mortality. Understanding the biology of cortical neural
stem cells is essential for understanding human evolution,
the pathogenesis of human neurodevelopmental disorders
and the rational design of neural repair strategies in adults.
During embryonic development, all of the neurons in
the cortex are generated from a complex population
of multipotent stem and progenitor cells. Much of the
research in the lab centres on the cell and molecular
biology of cortical stem cells. We are particularly
interested in the molecular mechanisms controlling
multipotency, self-renewal and neurogenesis, and how
these are coordinated to generate complex lineages in a
fixed temporal order. A number of ongoing projects in the
group address the functional importance of transcriptional
and epigenetic mechanisms in this system.
In the other major strand of research in the group, we
have developed methods for directing differentiation of
human pluripotent stem cells to cortical neurons, via a
cortical stem cell stage. Human stem-cell-derived cortical
neurons form functional networks of excitatory synapses
in culture. We are using this system for studies of human
neural stem cell biology and to generate models of
cortical diseases. Our initial focus has been on dementia,
where we have used stem cells from people with Down
syndrome and from patients with familial Alzheimer’s
disease to create cell culture models of Alzheimer’s disease
pathogenesis in cortical neurons. We are using those
models to study Alzheimer’s disease pathogenesis and the
efficacy of current therapeutic strategies.
22
THE GURDON INSTITUTE
Selected publications:
• Livesey FJ (2014) Human stem cell models of dementia.
Human Molecular Genetics doi:10.1093/hmg/ddu302
• Olsson B, Legros L, Guilhot F, Strömberg K, Livesey
FJ, Wilson DH, Zetterberg H and Blennow K (2014)
Imatinib treatment and Aß42 in humans. Alzheimer’s and
Dementia epub ahead of print.
• Alsiö JM, Tarchini B, Cayouette M, and Livesey FJ (2013)
Ikaros promotes early-born neuronal fates in the cerebral
cortex. PNAS 110: E716–E725.
• Shi Y, Kirwan P, Smith J, Robinson HP and Livesey FJ
(2012) Human cerebral cortex development from
pluripotent stem cells to functional excitatory synapses.
Nat Neurosci 15, 477-486
Fig 1. Human stem cell-derived neuroepithelial
rosettes in cell culture
Fig 2. Human stem cellderived excitatory neurons.
Fig 3. Axons of
human stem cellderived neurons in a
microfluidic device.
Figure 4.
Superresolution image
of GFP (green) and
mCherry (red) labelled
microtubule-binding
proteins on microtubules
(blue) in human axons
and dendrites.
THE GURDON INSTITUTE
23
Eric Miska
Fig 1. We have discovered that let-7, LIN-28 and the poly(U)
polymerase form an ultraconserved switch that regulates stem
cell decisions in C elegans
Non-coding RNA and genome dynamics
Co-workers: Alper Akay, Alyson Ashe, Fabian Braukmann, Tomas di Domenico, Tanay Ghosh, Sabrina Huber, Joanna Kosalka, Miranda Landgraf,
Jéremie le Pen, Milan Malinsky, Eyal Maori, Ragini Medhi, Marc Ridyard, Alexandra Sapetschnig, Mélanie Tanguy, Eva-Maria Weick
RNA is at the heart of cellular and organismal control with
non-coding RNA emerging as a major regulator of many
biological processes. We are interested in all aspects of
regulation by non-coding RNA. Our goal is to understand
how non-coding RNAs regulate development, physiology
and disease, in particular human cancer. Current research
themes include: miRNA biology and pathology, miRNA
mechanism, piRNA biology and the germline, endo-siRNAs
in epigenetic inheritance and environmental conditioning,
small RNA evolution, RNA modification and the role of
RNAi in host pathogen interaction. For example:
piRNAs are animal-specific small RNAs usually
restricted to the germline and required for fertility.
In 2008 we identified the piRNAs of C. elegans. We
have demonstrated that Piwi proteins and piRNAs
are important for gemline development and fertility. In
the absence of piRNAs germline genome integrity is
compromised and consequently piRNAs have been called
“guardians of the genome”. Now we are investigating how
piRNAs are generated in the germline and how they act
to silence their targets. We are also working to understand
how the piRNA pathway responds to foreign DNA/RNA
and how this pathway is evolving.
We are using a combination of molecular genetics,
biochemical and computational approaches to study RNA.
Model organisms have been key in unravelling almost
all biological processes. With a 100 Mb genome, three
days generation time and an established genetic toolkit,
C elegans is the ideal starting point to address many
questions in RNA biology. In addition, we are taking
advantage of mammalian models and human cell culture.
24
THE GURDON INSTITUTE
Selected publications:
• Gapp K, Jawaid A, Sarkies P, Bohacek J, Pelczar P, Prados J,
Farinelli L, Miska EA and Mansuy IM (2014) Implication of
sperm RNAs in transgenerational inheritance of the effects
of early trauma in mice. Nat Neurosci 17(5): 667-9
• Ashe A, Bélicard T, Le Pen J, Sarkies P, Frézal L, Lehrbach
NJ, Félix MA, Miska EA (2013) A deletion polymorphism
in the Caenorhabditis elegans RIG-I homolog disables viral
RNA dicing and antiviral immunity. E-Life 2:e00994
• Sarkies P and Miska EA (2012) Molecular biology. Is
there social RNA? Science 341(6145), 467 - 468
• Ashe A, Sapetschnig A, Weick EM, Mitchell J, Bagijn MP,
Cording AC, Doebley AL, Goldstein LD, Lehrbach NJ, Le
Pen J, Pintacuda G, Sakaguchi A, Sarkies P, Ahmed S and
Miska EA (2012) piRNAs can trigger a multigenerational
epigenetic memory in the germline of C. elegans. Cell 150,
88-99
• Bagijn MP, Goldstein LD, Sapetschnig A, Weick EM,
Bouasker S, Lehrbach NJ, Simard MJ and Miska EA (2012)
Function, targets, and evolution of Caenorhabditis elegans
piRNAs. Science 337, 574 - 578
Fig 2. An in-vivo assay for piRNA function in the germline. piRNAs and Piwi
proteins protect the germline. We are using molecular genetics, cell biology and
high-throughput sequencing to discover miRNA biogenesis and mechanisms.
Fig 3. RNA-mediated transgenerational
epigenetic inheritance in C elegans
THE GURDON INSTITUTE
25
Eugenia Piddini
Fig 1. Stress signaling and cell competition. Loser Minute -/+ mutant
cells chronically activate the JNK reporter Puc-LacZ and the JAK/STAT
ligand Unpaired-3 in the adult fly gut (top and bottom, respectively,
compare wt gut on the left to Minute -/+ gut on the right).
Competitive cell interactions in normal physiology and cancer
Co-workers: Michael Dinan, Maja Goschorska, Golnar Kolahgar, Kasia Kozyrska, Iwo Kucinski, Kathy Oswald, Saskia Suijkerbuijk, Silvia Vivarelli,
Laura Wagstaff
The elimination of suboptimal cells from tissues is an
important process that helps preserve tissue function.
Cell competition is a quality control mechanism that
achieves exactly that: when suboptimal cells are present,
they are recognised by surrounding fitter cells, which
eliminate them through competition. Much of the work in
our lab focuses on investigating the mechanisms and the
physiological role of this phenomenon, combining work in
Drosophila and in mammalian cell culture models.
Through transcriptional profiling we have identified a
molecular signature common to cells that are normally
outcompeted in Drosophila wing imaginal discs. This has
allowed us to identify a number of genes and signaling
pathways that we have found to play a role in cell
competition and which we are currently investigating
further.
Cell competition has been mostly studied in developing
tissues, but recently has also been observed in adult
tissues. This has important implications, as selection of fitter
cells during adult tissue maintenance can lead to improved
health and potentially delay tissue ageing. Our lab studies
the mechanism and relevance of cell competition in adult
tissues using the adult Drosophila gut as a model. Through
these studies we found that the JNK and JAK/STAT
signaling pathways, activated in loser cells, help promote
the proliferation of fitter stem cells.
In addition the lab investigates the role of cell competition
in cancer. Indeed it has been suggested that precancerous
cells could act as supercompetitors and kill surrounding
normal cells. For these studies we use a fly model of adult
intestinal adenoma and there we have shown that indeed
growing adenoma cells kill surrounding normal tissue. We
are currently investigating the relevance of this for tumour
growth.
26
THE GURDON INSTITUTE
Selected publications:
• Graml V, Studera X, Lawson JL, Chessel A, Geymonat
M, Bortfeld-Miller M, Walter T, Wagstaff L, Piddini E and
Carazo-Salas RE (2014) A genomic multi-process survey
of the machineries that control and link cell shape,
microtubule organisation and cell cycle progression. Dev
Cell 31(2):227-39 Journal Cover
• Wagstaff L, Kolahgar G and Piddini E (2013) Competitive
cell interactions in cancer: a cellular tug of war. Trends in
Cell Biology 23(4):160-7.
• Vincent JP*, Kolahgar G, Gagliardi M and Piddini E*
(2011) Steep differences in Wingless signalling trigger
Myc-independent competitive cell interactions. Dev Cell
366-374 * Corresponding authors
Fig 2. Adenoma cells suppress the growth of neighbouring
normal cells. We can generate fly guts in which normal and
mutated adenoma cells (identified by differential GFP/RFP
labeling) co-exist as shown in the top panels. This approach
shows that wt cell clones (RFP negative, bottom panels) are
much tinier if they grow alongside adenoma (right) cells than
if they grow next to other wild-type cells (left).
• Piddini E and Vincent JP (2009) Interpretation of the
Wingless gradient requires signalling-induced self-inhibition
(2009). Cell 136, 296-307
• Vivarelli S, Wagstaff L and Piddini E (2012) Cell wars:
regulation of cell survival and proliferation by cell
competition. Essays Biochem 10;53(1):69-82
Fig 3. Mechanical cell competition: we have discovered a new type of cell
competition whereby cells are eliminated because of hypersensitivity to
crowding. As shown here, while normal MDCK cells grow well and increase
in density with time, ScribbleKD loser cells grown at the same density are
inhibited in growth.
THE GURDON INSTITUTE
27
Jonathon Pines
How do cells control mitosis?
Co-workers: Barbara Di Fiore, Anja Hagting, Andrew Harrison, Daisuke Izawa, Mark Jackman, Chiara Marcozzi, Oxana Nashchekina, Bernhard Strauss, Jill Temple, Samuel Wieser, Claudia Wurzenberger, Keiko Yata
A dividing cell must ensure that the two daughter cells
receive an equal and identical copy of the genome, and
this is the focus of our research. We aim to determine how
cells regulate entry to mitosis and subsequently coordinate
chromosome segregation with cell separation.We know
that mitosis is controlled by the interplay between protein
kinases, protein phosphatases, and APC/C-mediated
proteolysis, and to understand the rapid and complex
dynamics of mitosis it is essential to study these processes
in living cells, complemented by biochemical analyses. We
introduce fluorescent tags into the genes encoding our
proteins of interest by homologous recombination to
enable us to measure protein numbers and kinetics in vivo,
which we can use to inform molecular models.
To understand how cells trigger mitosis we are analysing
the behaviour of the key mitotic kinases and their
regulators. We developed a FRET biosensor to assay
the dominant mitotic kinase, Cyclin B1-Cdk1, in vivo and
are using this to define the pathways that regulate the
timing of mitosis. To identify the proteins responsible for
regulating the Cyclin-Cdks, and provide insights into CyclinCdk substrates, we analyse protein complexes through the
cell cycle by SILAC mass spectrometry.
To understand how proteolysis regulates progress through
mitosis we complement the analysis of APC/C-dependent
degradation in living cells with biochemical analyses of
protein complexes and ubiquitination activity. These studies
are revealing how the APC/C is activated and how it is
able to select a particular protein for destruction at a
specific time. The crucial role of the Spindle Assembly
Checkpoint in controlling the APC/C to regulate
chromosome segregation has meant that our recent work
has elucidated the mechanisms of some of the key steps in
the checkpoint pathway, and revealed how the checkpoint
is able to rapidly inactivate the APC/C.
28
THE GURDON INSTITUTE
Mass spectroscopy analysis reveals the dynamic interactions of
the different cyclins through the cell cycle.
Credit: Felicia Walton-Pagliuca & Mark Collins (Sanger Institute)
Selected publications:
• Izawa D and Pines J (2015) The Mitotic Checkpoint
Complex binds a second CDC20 to inhibit active APC/C.
Nature 517, 631-634
• Collin P, Nashchekina O, Walker R and Pines J (2013) The
spindle assembly checkpoint works like a rheostat not a
toggle-switch Nat Cell Biol 15, 1378-1385
• Mansfeld J, Collin P, Collins MO, Choudhary J and Pines J
(2011) APC15 drives the turnover of MCC-Cdc20 to
make the spindle assembly checkpoint responsive to
kinetochore attachment. Nat Cell Biol 13, 1234-1244.
• Pagliuca F, Collins MO, Lichawska A, Zegerman P,
Choudhary JS and Pines J (2011) Quantitative proteomics
reveals the basis for the biochemical specificity of the cell
cycle machinery. Mol Cell 43, 406-417.
• Gavet O and Pines J (2010) Progressive activation of
Cyclin B1-Cdk1 coordinates entry to mitosis. Dev Cell 18,
533-543.
Mitotic RPE-1 cell with a single unattached
chromosome. Mad2 in red, Hec1 in green, DNA in
blue and spindle MTs in white. (Philippe Collin)
Montage of a prometaphase cell in which the Venus fluorescent protein
has been knocked into the Mad2 locus. Mad2 binds to unattached
kinetochores.The chromosomes are labelled with ectopically expressed
Histone H2B-mRuby. (Philippe Collin)
THE GURDON INSTITUTE
29
Emma Rawlins
Adult mouse lung section showing lineage-labelled
secretory cells (green) in the conducting airways.
Stem and progenitor cells in the mammalian lung
Co-workers: Gayan Balasooriya, Christoph Budjan, Jo-Anne Johnson, Usua Laresgoiti Garay, MarKo Nikolic
Our lungs have a complex three-dimensional structure
which facilitates respiration and host defence. Building this
structure requires that lung embryonic progenitor cells
produce the correct types and numbers of cells in the
correct sequence. How is this controlled? And how is the
final structure maintained in the adult? Our lab investigates
the cellular and molecular mechanisms which control
stem and progenitor cell fate decisions in the developing
and adult lungs. Key unanswered questions include what
mechanisms control the decision of lung progenitors
to self-renew or to differentiate? Which pathways are
required for cell lineage specification in the lung? Our
approach is to use the power of mouse genetics to
understand the control of lung progenitor cell behaviour
at the single cell level. This allows individual cells to be
analysed quantitatively in vivo, or by live-imaging in organ
culture systems.
We have previously shown that in the embryonic lung
there is a population of Id2+ multipotent epithelial
progenitor cells located at the distal tips of the budding
epithelium. The developmental potential, or competence,
of these cells changes during embryogenesis. At the
same time the cells undergo a change in gene expression
pattern. We are currently exploring the cellular and
molecular basis of this change in competence.
The identity of the epithelial stem and progenitor cells
in the postnatal lung remains controversial. Our previous
work has shown that each anatomical region (trachea,
bronchioles, alveoli) has its own progenitor cell population
and that the behaviour of these progenitors can change in
response to local conditions. Our current postnatal work
focuses on:
• Better characterising the adult lung progenitor cells.
This includes testing whether progenitor cell behaviour is
widespread or there are stem cells.
• Understanding the genetic regulation of the progenitors
under several different physiologically-relevant conditions.
30
THE GURDON INSTITUTE
In particular, we are focusing on genes that are
hypothesised to control the decision to self-renew or
differentiate.
Our long-term vision is to combine the developmental
and homeostatic aspects of our work to develop new
approaches to ameliorate human pulmonary disease. In
particular, we are working towards being able specifically
to direct endogenous lung stem cells to generate any lung
epithelial cell type.
Selected publications:
• Rawlins EL, Okubo T, Xue Y, Brass DM, Auten RL,
Hasegawa H, Wang F and Hogan BLM (2009) The role of
Scgb1a1+ Clara cells in the long-term maintenance and
repair of lung airway, but not alveolar, epithelium. Cell Stem
Cell 4 525-534
• Rawlins EL, Clark CP, Xue Y and Hogan BLM (2009)
The Id2 distal tip lung epithelium contains individual
multipotent embryonic progenitor cells. Development 136
3741-3745
• Rawlins EL (2011) The building blocks of mammalian
lung development. Developmental Dynamics 240 463-76
Mouse embryonic lung growing in culture. Blue (X-gal staining) shows
grafted stem cells which have been incorporated into the lung structure.
• Onaitis M, D’Amico TA, Clark C, Guinney J, Harpole
DH and Rawlins EL ( 2011) A 10-gene progenitor cell
signature predicts prognosis in lung adenocarcinoma.
Annals of Thoracic Surgery 91 1046-50
Mouse embryonic lung undergoing
branching morphogenesis, stained to
show the epithelium (E-cadherin).
A clone of mutant tracheal epithelial
cells labelled with GFP (green).
THE GURDON INSTITUTE
31
Ben Simons
Mechanisms of stem cell fate in development, maintenance, and diseased states
Co-workers: Juergen Fink (Stem Cell Institute), Philip Greulich (Cavendish), Edouard Hannezo (Cavendish), Teresa Krieger, Crystal McClain (Stem
Cell Institute), Steffen Rulands (Cavendish)
The coordination of cell proliferation and fate specification
is central to the development and maintenance of tissues. In
development, systems must be tightly-regulated to ensure that
precise numbers of lineage-specified cells are generated in the
correct sequence whilst, in adult, a delicate balance between
proliferation and differentiation is essential for homeostasis.
Through a programme of multidisciplinary and collaborative
research, our group develops biophysical approaches to
identify unifying principles of stem cell regulation in the
development and maintenance of tissues, and to use the
insights to resolve pathways leading to dysregulation in
diseased states.
Applying methods from statistical physics, we have shown
that strategies of stem cell self-renewal can be grouped into
one of four “universality” classes according to whether the
balance between proliferation and differentiation is achieved
at the level of individual cells or the population, and whether
regulation follows from intrinsic (cell-autonomous) processes
or is mediated by signals from the niche. By combining lineage
tracing studies with marker-based assays, we have used these
insights to show that different mammalian epithelial tissues,
including epidermis, oesophagus, intestine, trachea, and
germline conform to population asymmetry, in which stem
cells are lost and replaced, leading to “neutral drift” dynamics
and consolidation of clonal diversity.
Lately, by combining static clonal labeling assays with
intravital in vivo live-imaging approaches, we have found
that both intestinal and germline stem cells are functionally
heterogeneous, with cells moving reversibly between “primed”
states, biased for self-renewal or differentiation and loss.
This process of “dynamical heterogeneity” has established a
new paradigm for tissue maintenance, with ramifications for
stem cell identity and function. Building on the success of this
programme, we have used parallel approaches to identify
patterns of progenitor cell fate in the development of tissues,
including the neocortex and heart, as well as in processes
leading to dysregulation in diseased states.
Peter Dirks, Bill Harris, Meritxell Huch, Rick Livesey, Eugenia
Piddini, Emma Rawlins, Wolf Reik, Jacco van Rheenen, Songhai
Shi, Hongjun Song, and Shosei Yoshida.
Selected publications:
• Gao P, Postiglione MP, Krieger TG, Hernandez L, Wang C,
Han Z, Streicher C, Papusheva E, Insolera R, Chugh K, Kodish
O, Huang K, Simons BD, Luo L, Hippenmeyer S and Shi
SH (2014) Deterministic progenitor behavior and unitary
production of neurons in the neocortex. Cell 159, 775-88
• Hara K, Nakagawa T, Enomoto H, Suzuki M, Yamamoto
M, Simons BD and Yoshida S (2014) Mouse spermatogenic
stem cells continually interconvert between equipotent singly
isolated and syncytial states. Cell Stem Cell 14, 658-72
• Ritsma L, Ellenbroek SI, Zomer A, Snippert HJ, de Sauvage
FJ, Simons BD, Clevers H and van Rheenen J (2014) Intestinal
crypt homeostasis revealed at single-stem-cell level by in vivo
live imaging. Nature 507, 362-5
• Mascre G, Dekoninck S, Drogat B, Youssef KK, Brohee S,
Sotiropoulou PA, Simons BD and Blanpain C (2012) Distinct
contribution of stem and progenitor cells to epidermal
maintenance. Nature 489, 257-62
• Driessens G, Beck B, Caauwe A, Simons BD and Blanpain
C (2012) Defining the mode of tumour growth by clonal
analysis. Nature 488, 527-30
Studies of clonal fate using a multicolour inducible genetic labelling system
provide a vivid demonstration of neutral
drift dynamics and the progession
towards monoclonality in crypt. The
top image shows a section through
the base of the crypt showing the
clonal progeny of the stem/paneth cell
compartment at 7 days post-induction.
The bottom image shows the migration
streams of differentiated cells moving
up (fully-clonal crypts) and onto villi.
Inducible genetic labelling allows
the fate of progenitor cells and
their progeny to be traced in
epidermis both in normal and
diseased states. The figure
shows the progeny of a GFP
labelled cell in a squamous
tumour in mouse. Such lineage
tracing assays allows for the
in vivo characterisation of the
tumour-initiating potential of
tumour cells, and the study of
the progression from benign
papilloma to invasive squamous
carcinoma.
Genetic lineage tracing of mouse heart development using
a multicolour confetti reporter allows the potency of early
Mesp1 expressing cells to be resolved, and provides insight
into tissue morphogenesis.
Current collaborators include Cedric Blanpain, Hans Clevers,
32
THE GURDON INSTITUTE
THE GURDON INSTITUTE
33
Daniel St Johnston
Polarising epithelial cells and body axes
Co-workers: Dan Bergstralh, Catiá Carvalho, Jia Chen, Nicole Dawney, Hélène Doerflinger, Artur Fernandes, Weronika Fic, Jackie Hall, Holly
Lovegrove, Nick Lowe , Dmitry Nashchekin, Avik Mukherjee, Jennifer Richen, George Sirinakis, Vanessa Stefanak, Vitor Trovisco, Helen ZennerBranco
Cell polarity is essential for normal cell shape and function
and underpins key developmental processes, such as
cell migration, axis determination and asymmetric stem
cell divisions, whereas a loss of polarity is a hallmark of
tumours. We are using Drosophila and mammalian tissue
culture cells to analyse how cells polarise and how cortical
polarity factors regulate other polarised aspects of cell
behaviour.
Most organs in the body are composed of epithelial cells
that polarise along their apical-basal axes and adhere to
each other to form sheets of cells that act as barriers
between compartments. We use the follicular epithelium
that surrounds the developing Drosophila egg chamber as a
model to investigate how apical-basal polarity is established
in a secretory epithelium and how polarity factors control
the organisation of the microtubule cytoskeleton. For
example, we have analysed how the mitotic spindle is
oriented to ensure that both daughter cells remain within
epithelium, as mis-oriented spindles have been proposed
to contribute to tumour development. We have also
started to examine the relationship between epithelial
polarity and polarised secretion using custom-built, superresolution microscopes, so that we can image vesicles
targeted for apical or basal secretion that are too small to
see using conventional systems.
Almost all well-characterised epithelia are secretory and
little is known about the polarity of absorptive epithelia,
where the main direction of transcellular transport is
reversed. We have found that the absorptive enterocytes
of the adult midgut rely on different polarity factors
from secretory epithelia. Midgut cells have an inverted
arrangement of intercellular junctions compared to the
follicle cells, making them more similar to mammalian
epithelia. We are therefore using the gut as a model to
discover and analyse new polarity factors and pathways.
Another major goal of the group is to understand
how the Drosophila oocyte is polarised to define the
anterior-posterior axis of the embryo. This requires the
microtubule-dependent transport of bicoid and oskar
34
THE GURDON INSTITUTE
mRNAs to opposite ends of this very large cell, and we
use a range of live imaging techniques to visualise moving
mRNAs and growing microtubules in wildtype and mutant
oocytes.
Selected publications:
• Lowe N, St Johnston D et al (2014) Analysis of
the expression patterns, subcellular localisations and
interaction partners of Drosophila proteins using a pigP
protein trap library. Development 141, 3994-4005
Fig 1. Tracks of growing microtubules
on the apical side of the follicular
epithelium over a two-minute period
• Morais-de-Sa E, Mukherjee A, Lowe N and St Johnston
D (2014) Slmb antagonises the aPKC/Par-6 complex to
control oocyte and epithelial polarity. Development 141,
2984-2992
Fig 2. A clone of mutant follicle cells (marked by the loss of
nuclear GFP) that have lost their apical-basal polarity and
been extruded from the epithelium.
Fig 3. An egg chamber containing two types of follicle cell clones
homozygous for mutations that delay the switch between proliferation
and differentiation. One class of clones is marked by the loss GFP
(green), the other by the loss of RFP (red) and the nuclei have been
counterstained for DNA (blue). The two mutations are additive as the
cells in the double mutant clones (blue only) are smaller than either
single mutant.
• Gardiol A and St Johnston D (2014) Staufen targets
coracle mRNA to Drosophila neuromuscular junctions
and regulates GluRIIA synaptic accumulation and bouton
number. Developmental Biology 392, 153-167
• Bergstralh D T and St Johnston D (2014) Spindle
orientation: what if it goes wrong? Seminars in Cell &
Developmental Biology 34, 140-145
• Morais-de-Sa E, Vega-Rioja A, Trovisco V, St Johnston D
(2013) Oskar is targeted for degradation by the sequential
action of Par-1, GSK-3, and the SCF-Slimb ubiquitin ligase.
Developmental Cell 26: 303-314
Fig 4. A model showing the polarity factors
that mark different cortical domains in
epithelial cells and the inhibitory interactions
between them.
Fig 5. A stage 10 egg chamber expressing a marker for the
microtubule minus ends fused to Cherry fluorescent protein
(red), counterstained for DNA (blue). The minus ends of the
microtubules are anchored to the anterior cortex of the oocyte
and direct the localisation of bicoid mRNA.
THE GURDON INSTITUTE
35
Azim Surani
Germline - specification and programming for totipotency and development
Co-workers: Delphine Cougot, Dang Vinh Do, Lynn Froggett, Wolfram Gruhn, Ufük Günesdogan, Jamie Hackett, Yun Huang, Naoko Irie, Elena
Itskovich, Shinseog Kim, Toshihiro Kobayashi, Caroline Lee, Roopsha Sengupta, Walfred Tang, Thor Theunissen, Julia Tischler, Jan Zylicz
Specification of primordial germ cells (PGCs) occurs after
the development of equipotent post implantation epiblast
cells, which also give rise to all the somatic cells in mice.
Recent studies show that that BLIMP1, PRDM14 and AP2g
are necessary and sufficient for PGC specification (Fig 1).
This mutually interdependent tripartite genetic network
initiates PGC specification by repressing the somatic
programme but induces pluripotency genes and the germ
cell programme (Fig 2). These events can be captured
in vitro under specific conditions after they undergo
priming and gain competence for the specification of
cell fates. The network also initiates sequential and
dynamic changes in histone modifications, reactivation
of the X chromosome and comprehensive global DNA
demethylation, including imprints erasure (Fig 3). The latter
is important for the initiation of the imprinting cycle in
the germ line, and subsequently, establishment of parent
of origin specific imprints (Fig 4). The inheritance of
these epigenetic modifications after fertilisation results in
functional differences between parental genomes, which
following fertilisation is critical for the establishment of
totipotency. We are interested in the wider applications
of the knowledge gained from the specification of PGCs
and epigenetic reprogramming for the manipulation of
pluripotent state and cell fates
Selected publications:
• Irie N, Weinberger L, Tang WWC, Kobayashi T, Viukov
S, Manor Y, Dietmann S, Hanna JH and Surani MA (2015)
SOX17 is a critical specifier of human primordial germ cell
fate. Cell 160, 253-268
• Kim S, Günesdogan U, Zylicz JJ, Hackett JA, Cougot D,
Bao S, Lee C, Dietmann S, Allen GE, Sengupta R and Surani
MA (2014) PRMT5 protects genomic integrity during
global DNA demethylation in primordial germ cells and
preimplantation embryos. Molecular Cell 56(4) 564-579
Fig 2a. Differential occupancy and
combinatorial roles of BLIMP1, PRDM14 and
AP2g : BLIMP1 occupies promoters, PRDM14,
distal regulatory elements and AP2g bind
both. Venn diagram depicts genes bound by
the three factors
Fig 1. Founder population
of PGCs at E7.5 detected
by STELLA.
• Hackett JA, Sengupta R, Zylicz JJ, Murakami K, Lee
C, Down TA and Surani MA (2013) Germline DNA
demethylation dynamics and imprint erasure through
5-hydoxymethylcytosine. Science 339, 448-452
Fig 2b. The tripartite genetic network
regulates expression of genes at PGC
specification
• Magnüsdöttir E, Dietmann S, Murakami K, Günesdogan
U, Tang F, Bao S, Diamanti E, Lao K, Gottgens B, Surani MA
(2013) A tripartite transcription factor network regulates
primordial germ cell specification in mice. Nature Cell
Biology 15, 906-915
Fig 3. Parallel routes to
reprograming and the
establishment of the
epigenetic ground state
in primordial germ cells
Fig 4. Germline – Imprinting
cycle generates the totipotent/
pluripotent states with
parent of origin specific DNA
methylation imprints for the
transmission of epigenetic
information.
36
THE GURDON INSTITUTE
THE GURDON INSTITUTE
37
Philip Zegerman
The regulation of DNA replication initiation in eukaryotes
Co-workers: Geylani Can, Clara Collart, Vincent Gaggioli, Christine Hänni, Mark Johnson, Barbara Schöpf
To successfully pass on their genetic information, every
organism must make a perfect duplicate of their genome
in every cell cycle. Failure to copy every chromosome
faithfully leads to genomic instability, which is the
root cause of cancer. As a result, the process of DNA
replication must be strictly regulated, within the normal cell
cycle, after DNA damage and during development. Our
research takes advantage of a wide variety of organisms
to understand the molecular mechanism of how this strict
regulation of DNA replication is achieved.
Perfect genome duplication in eukaryotes is achieved by
coupling the assembly of the DNA replication apparatus
with the cell cycle. The fundamental regulator of the cell
cycle, Cyclin-Dependent Kinase (CDK) plays a pivotal role
in ensuring that replication initiation can only occur once
before cell division. We have previously shown that CDK
phosphorylates the two essential replication initiation
factors Sld2 and Sld3, which in turn allows binding to
another essential initiation factor called Dpb11. How CDK
phosphorylation of these targets facilitates replication
initiation is not known, but the transient association of
these factors at origins produces a switch that only allows
replication initiation in S-phase of the cell cycle.
Interestingly, the time it takes to copy the genome changes
during development. For example in many organisms
S-phase is fast in the embryo, but greatly slows down in
somatic cells. We have shown that it is the level of the
key CDK targets that determines the rate of genome
duplication in early vertebrate embryogenesis. Our work
has therefore pinpointed a fundamental step in replication
initiation that determines both the fidelity and the rate of
DNA replication across eukaryotes.
Selected publications:
• Gaggioli V, Zeiser E, Rivers D, Bradshaw CR, Ahringer
J and Zegerman P (2014) CDK phosphorylation of
SLD-2 is required for replication initiation and germline
development in C. elegans. J Cell Biol 204, 507-522
Replication initiation must be strictly controlled to occur once, and
only once, in every cell cycle.
• Collart C, Allen GE, Bradshaw CR, Smith JC and
Zegerman P (2013) Titration of four replication factors
is essential for the Xenopus laevis midblastula transition.
Science doi:10.1126/science.1241530
• Mantiero D, Mackenzie A, Donaldson A and Zegerman P
(2011) Limiting factors execute the temporal programme
of origin firing in budding yeast. EMBO J 23, 4805-4814
• Zegerman P and Diffley JF (2010) Checkpoint
dependent inhibition of DNA replication initiation via
phosphorylation of Sld3 and Dbf4. Nature 467, 474-478
• Zegerman P and Diffley JF (2007) Phosphorylation of
Sld2 and Sld3 by cyclin-dependent kinases promotes DNA
replication in budding yeast. Nature 445, 281-285
Xenopus laevis embryos at the Midblastula Transition. Left is a normal embryo,
right is an embryo over-expressing limiting replication factors.
The sequence of eukaryotic replication initiation
Phospho-peptide array analysis of replication initiation factors.
38
THE GURDON INSTITUTE
THE GURDON INSTITUTE
39
Magdalena Zernicka-Goetz
Developmental plasticity, fate and morphogenesis in the mouse embryo
Relocated: The Zernicka-Goetz Group are now located in the Department of Physiology, Development and Neuroscience, University of
Cambridge.
We investigate mechanisms underlying the specification of
cell fate and patterning using mouse embryos as our major
model because this allows us to combine cell biological
and molecular genetic approaches with live imaging in a
system that is close to human development.
Plasticity and Cell Fate acquisition: Embryonic cells
in mouse and human are flexible and how their fate
becomes restricted is unclear. To determine the molecular
steps that mediate the transition from the egg totipotency
towards either differentiation or pluripotency, we have
isolated a number of regulatory genes essential for lineage
determination and follow the interplay between cell
polarity, position and developmental history of cells on fate
specification.
Asymmetric and Symmetric divisions: Development
begins with the asymmetric divisions of the oocyte,
following fertilisation cells divide symmetrically until the
8-cell stage when division asymmetry is again important.
To understand the processes that break symmetry, we
study the events that lead to cell polarisation and spindle
orientation.
Maternal to Zygotic Transition: To understand the
factors essential for the correct development, we have
established a non-invasive method to forecast already
at fertilisation which eggs have the highest chance of
development to birth. We collaborate with IVF clinics to
select with this approach the best quality eggs for transfer
to would-be-mothers.
Self-organisation of pattern: We wish to understand how
embryo integrates the development of different cell types
into an organism. To address this, we have developed
in vitro system to culture and image development at
implantation stages outside the mother and to mimic
several of the key morphogenetic steps using ES cells.
40
THE GURDON INSTITUTE
Selected publications:
• Graham SJ, Wicher KB, Jedrusik A, Guo G, Herath W,
Robson P and Zernicka-Goetz M (2014) BMP signalling
regulates the pre-implantation development of extraembryonic cell lineages in the mouse embryo. Nature
Comm 5: 5667
• Bedzhov I, Leung CY, Bialecka M and Zernicka-Goetz M.
(2014) In vitro culture of mouse blastocysts beyond the
implantation stages. Nature Protocols 9(12): 2732-9
• Bedzhov I and Zernicka-Goetz M (2014) Selforganizing properties of mouse pluripotent cells initiate
morphogenesis upon implantation. Cell 156(5):1032-44.
• Coelho PA, Bury L, Sharif B, Riparbelli MG, Callaini
G, Glover DM and Zernicka-Goetz M (2013) Spindle
formation in the mouse embryo requires plk4 in the
absence of centrioles. Dev Cell 27(5): 586-97
• Leung CY and Zernicka-Goetz M (2013) Angiomotin
prevents pluripotent lineage differentiation in mouse
embryos via Hippo pathway-dependent and -independent
mechanisms. Nature Comm 2013, 4: 2251
Mouse embryo before implantation - trophectoderm in blue and pluripotent
cells in different colours. (Image: CY Leung)
Mouse embryo rosette at the time of implantation”. Nuclei
in blue, membranes in red and apical marker in yellow.
THE GURDON INSTITUTE
41
CATEGORIES OF APPOINTMENT / SENIOR GROUP LEADERS
CATEGORIES OF APPOINTMENT
SENIOR GROUP LEADER
Professor, Director of Research or Reader
GROUP LEADER
5-year grant-funded appointment (maximum 10 years)
CAREER DEVELOPMENT FELLOW
4-year grant-funded appointment
SENIOR RESEARCH ASSOCIATE
Grant-funded appointment within individual groups
RESEARCH ASSOCIATE/FELLOW
Postdoctoral Fellow within individual groups,
appointed or invited by group leader
RESEARCH ASSISTANT
Postgraduate within individual groups, mainly grantfunded
GRADUATE STUDENT
3 or 4 year studentship within individual groups, mainly
grant-funded
RESEARCH TECHNICIAN
Within individual groups, mainly grant-funded
LABORATORY ASSISTANT / TECHNICIAN
Within individual groups or part of core support,
grant-funded
ITALICS: LEAVERS DURING 2014
POSTGRADUATE OPPORTUNITIES
As part of the University of Cambridge, the Institute
welcomes enquiries from prospective graduate
students. We have a thriving population of graduates
who contribute greatly, not only to the stimulating
research environment, but also to the life of the
Institute as a whole. Additionally, graduates become
members of the biological or medical sciences
department to which their group is affiliated. Graduate
studentships are supported mainly by the Wellcome
Trust or Cancer Research UK but additional
sponsorship may be solicited from a variety of sources,
including government research councils. Applicants
should write, in the first instance, to the leader of the
group they wish to join.
42
THE GURDON INSTITUTE
DANIEL ST JOHNSTON PhD FRS
FMedSci, Director
Wellcome Trust Principal Research Fellow
Professor of Developmental Genetics
Member, European Molecular Biology Organization
Director, Company of Biologists
(Member of the Department of Genetics)
DAN BERGSTRALH PhD
Wellcome Trust Research Associate
CATIA ALEXANDRA CARVALHO MENDES
PhD Student
JIA CHEN PhD
Royal Society KC Wong Fellow
NICOLE DAWNEY BSc
Wellcome Trust Research Assistant
HÉLÈNE DOERFLINGER PhD
Wellcome Trust Research Associate/Institute Outreach
Officer
ARTUR FERNANDES MPhil
School of Biological Sciences PhD Student
WERONIKA FIC PhD
Wellcome Trust Research Associate
TIMM HAACK PhD
Wellcome Trust Developmental Biology PhD Student
JACKIE HALL MSc
Wellcome Trust Senior Research Technician
HOLLY LOVEGROVE MSc
Herchel Smith PhD Student
NICK LOWE PhD
Wellcome Trust Research Associate
AVIK MUKHERJEE MSc
CISS PhD Student
DMITRY NASHCHEKIN PhD
Wellcome Trust Research Associate
JENNIFER RICHENS PhD
Wellcome Trust Research Associate
ARAM SAYADIAN MPhil Wellcome Trust PhD
Student (Formerly Wellcome Trust Developmental Biology
PhD Student)
GEORGE SIRINAKIS PhD
Wellcome Trust Senior Research Associate
VANESSA STEFANAK PhD
Administration Manager for the Office of the Director
SENIOR GROUP LEADERS
VITOR TROVISCO PhD
Wellcome Trust Research Associate
HELEN ZENNER-BRANCO PhD
Wellcome Trust Research Associate
JULIE AHRINGER PhD FMedSci
Professor of Genetics and Genomics
Wellcome Trust Senior Research Fellow
Member, European Molecular Biology Organization
(Member of the Department of Genetics)
ALEX APPERT PhD
Wellcome Trust Research Associate
DARYA AUSIANNIKAVA BSc
Darwin Trust PhD Student
FANÉLIE BAUER PhD
Herchel Smith Fellow
RON CHEN PhD
Wellcome Trust Senior Research Associate
MIKE CHESNEY PhD
Wellcome Trust Research Associate
YAN DONG MSc
Wellcome Trust Research Associate
KENNETH EVANS PhD
Wellcome Trust Research Associate
CSENGE GAL PhD
Wellcome Trust Research Associate
CAROLINA GEMMA PhD
Wellcome Trust Research Associate
MORITZ HERRMANN BSc
BBSRC PhD Student
JÜRGEN JÄNES MSc
Wellcome Trust Mathematical Biology PhD Student
DJEM KISSIOV BA
Wellcome Trust Research Assistant
ALICIA McMURCHY PhD
CIHR Research Associate
JOSANA RODRIGUEZ PhD
Wellcome Trust Research Associate
WEI QIANG SEOW BSc
A*STAR PhD Student
PRZEMYSLAW STEMPOR PhD
Wellcome Trust Research Associate
ANNETTE STEWARD BSc
Wellcome Trust Research Assistant
CHRISTINE TURNER
PA/Secretary
CARSON WOODBURY BA
Thouron Scholarship MPhil Student
BRIAN WYSOLMERSKI BA
Biology MPhil Student
EVA ZEISER BSc
Wellcome Trust Research Assistant
ANDREA BRAND PhD FRS FMedSci
Herchel Smith Professor of Molecular Biology
Head of Wellcome Trust Labs
Wellcome Trust Senior Investigator
Member of Council, The Royal Society
Member, European Molecular Biology Organization
(Member of the Department of Physiology,
Development and Neuroscience)
JANINA ANDER BSc
BBSRC PhD Student
JOSEPHINE BAGERITZ PhD
Wellcome Trust Research Associate
ELIZABETH CAYGILL PhD
BBSRC Research Associate
SETH CHEETHAM BSc
Herchel Smith PhD Student
ESTEBAN CONTRERAS SEPULVEDA PhD
Wellcome Trust Developmental Biology PhD Student
MELANIE CRANSTON BA
BBSRC Research Assistant
ABHIJIT DAS PhD
Wellcome Trust Research Associate
CATHERINE DAVIDSON BSc
BBSRC Research Associate
PAUL FOX PhD
Wellcome Trust Research Associate
KATRINA GOLD PhD
Wellcome Trust Research Associate (Formerly Wellcome
Trust Developmental Biology PhD Student)
ANNA HAKES MPhil
Wellcome Trust Developmental Biology PhD Student
JUN LIU BA
Dr Herchel Smith Graduate Fellow
OWEN MARSHALL PhD
BBSRC Research Associate
LEO OTSUKI MPhil
Wellcome Trust Developmental Biology PhD Student
CHLOE SHARD BSc
Australia Trust/McCrum PhD Student
TONY SOUTHALL PhD
Wellcome Trust Research Associate
PAULINE SPÉDER PhD
Wellcome Trust Research Associate
CHRISTINE TURNER
PA/Secretary
JELLE VAN DEN AMEELE MD PhD
Wellcome Trust Research Associate
MO ZHAO BSc
Chinese Scholarship Council PhD Student
NICK BROWN PhD
Professor of Cell Biology
Member, European Molecular Biology Organization
(Member of the Department of Physiology,
Development and Neuroscience)
NATALIA BULGAKOVA PhD
BBSRC Research Associate
JUAN MANUEL GOMEZ PhD
BBSRC Reseach Associate
HANNAH GREEN MPhil
Wellcome Trust Developmental Biology PhD Student
ANNABEL GRIFFITHS BA
MRC PhD Student
SVEN HUELSMANN PhD
University of Jyväskulä Research Associate
BENJAMIN KLAPHOLZ PhD
BBSRC Research Associate
TARUN KUMAR BSc
Commonwealth PhD Student
MIRANDA LANDGRAF MA
PA/Secretary
AIDAN MAARTENS PhD
BBSRC Research Associate
JOHN OVERTON HNC
Wellcome Trust/BBSRC Chief Research Technician
PAULA RODRIGUEZ SANCHEZ MSc
Wellcome Trust Research Assistant
PEERAPAT THONGNUEK MRes
Thai Government PhD Student
JOHN GURDON Kt DPhil DSc FRS
Distinguished Group Leader
Nobel Laureate in Physiology or Medicine 2012
Wellcome Trust Senior Investigator
Foreign Associate, US National Academy of Sciences
Foreign Associate, US National Academy of Sciences
Institute of Medicine
Foreign Associate, French National Academy of
Sciences
Member, European Molecular Biology Organization
Member, Academia Europaea
Honorary Fellow, Royal College of Physicians
Honorary Member of American Anatomical Society
Honorary Member of Anatomical Society of Great
Britain
Honorary Fellow UK Academy of Medical Sciences
(Member of the Department of Zoology)
DILLY BRADFORD
PA/Secretary
NIGEL GARRETT HNC
Wellcome Trust Research Assistant
RICHARD HALLEY-STOTT PhD
MRC Research Associate
EVA HÖRMANSEDER PhD
EMBO Research Fellow
JEROME JULLIEN PhD
Wellcome Trust Research Associate
MAGDALENA KOZIOL PhD
Isaac Newton Trust/Wellcome Trust Research
Associate
KEI MIYAMOTO PhD
Herchel Smith Research Associate
MAMI OIKAWA PhD
MRC Research Associate
ANGELA SIMEONE PhD
Wellcome Trust/MRC Research Associate
(Bioinformatics)
MARTA TEPEREK-TKACZ MSc
Wellcome Trust Developmental Biology PhD Student/MRC
Research Assistant
MUNENDER VODNALA PhD
Swedish Society for Medical Research (SSMF) Fellow
STAN WANG BS
NIH/Gates MD-PhD Student
MING-HSUAN WEN MSc
Taiwan Government PhD Student
THE GURDON INSTITUTE
43
SENIOR GROUP LEADERS
STEVE JACKSON PhD FRS FMedSci
Frederick James Quick Professor of Biology
Head of Cancer Research UK Labs
Member, European Molecular Biology Organization
ERC Advanced Researcher
Associate Faculty Member of the Wellcome Trust
Sanger Institute
(Member of the Department of Biochemistry)
PALLAVI AGARWAL PhD
Cancer Research UK Research Associate
GABRIEL BALMUS DVM PhD
Cancer Research UK Research Associate
LINDA BASKCOMB MSc
Cancer Research UK Senior Chief Research
Laboratory Technician
RIMMA BELOTSERKOVSKAYA PhD
Cancer Research UK Research Associate
ANDREW BLACKFORD PhD
Cancer Research UK Research Associate
JESSICA BROWN MB Bchir
Wellcome Trust Clinical Fellow/PhD Student
TING-WEI (Will) CHIANG MSc
Cambridge Overseas Trust PhD Student
JULIA COATES MA
Cancer Research UK Research Assistant
MATTHEW CORNWELL MChem
Dept Chemistry/Cambridge Cancer Centre/School of
Physical Sciences PhD Student
MUKERREM DEMIR BSc
ERC Senior Research Technician
KATE DRY PhD
Cancer Research UK Information Specialist
JOSEP FORMENT PhD
A-T Society/Cancer Research UK Research Associate
YARON GALANTY PhD
ERC Senior Research Associate
NICOLA GEISLER BSc
Wellcome Trust Chief Research Technician
MAREIKE HERZOG BA
Wellcome Trust PhD Student (joint with Sanger
Institute)
SATPAL JHUJH MSc
ERC Research Technician
DELPHINE LARRIEU PhD
Cancer Research UK/MRC Research Associate
CARLOS LE SAGE PhD
ERC Research Associate
44
THE GURDON INSTITUTE
SENIOR GROUP LEADERS
NATALIA LUKASHCHUK PhD
Cancer Research UK Research Associate
FRANCISCO MUNOZ MARTINEZ PhD
Marie Curie Research Intra-European Fellow
RYOTARO NISHI PhD
Cancer Research UK Research Associate
FABIO PUDDU PhD
EMBO Fellow/EU Research Associate
HELEN REED
PA/Secretary
ISRAEL SALGUERO CORBACHO PhD
Wellcome Trust Research Associate
CHRISTINE SCHMIDT PhD
ERC Research Associate
MATYLDA SCZANIECKA-CLIFT PhD
ERC Research Associate
ROHAN SIVAPALAN BSc
EU Research Assistant/BBSRC/Horizon PhD Student
JON TRAVERS PhD
EU Research Associate
DAVID WEISMANN PhD
EMBO Research Fellow
PAUL WIJNHOVEN BSc
Cancer Research UK Research Assistant/PhD Student
TONY KOUZARIDES PhD FRS
FMedSci
Deputy Director
Professor of Cancer Biology
Cancer Research UK Gibb Fellow
Member, European Molecular Biology Organization
ERC Advanced Researcher
(Member of the Department of Pathology)
ANDREJ ALENDAR MSc
Cancer Research UK Research Assistant
PAULO AMARAL PhD
ERC Research Associate
ANDREW BANNISTER PhD
Cancer Research UK Senior Research Associate,
Senior Radiation Protection Supervisor
ISAIA BARBIERI PhD
Leukaemia & Lymphoma Research Research Associate
ESTER CANNIZZARO MSc
Cancer Research UK PhD Student
KA HING CHE PhD
Cancer Research UK Research Associate
MARIA CHRISTOPHOROU PhD
Cancer Research UK Research Associate
ALISTAIR COOK GIBiol
ERC Chief Research Technician
CHUN YEW FONG MBBS
BMedSci Visiting Leukaemia Foundation of Australia/
Haematology Society of Australia & New Zealand/Royal
Australasian College of Physicians Clinical Fellow
NAMSHIK HAN PhD
ERC Research Associate (Bioinformatics)
SRI LESTARI MSc
Cancer Research UK Senior Research Laboratory
Technician
NIKKI MANN BA
PA/Secretary
VALENTINA MIGLIORI PhD
EMBO Fellow/King’s College Junior Research Fellow
GONZALO MILLAN ZAMBRANO PhD
BBSRC Research Associate
JESSICA MORISON PhD
Visiting Researcher
SAM ROBSON PhD
ERC Research Associate (Bioinformatics)
HELENA SANTOS ROSA PhD
Cancer Research UK Senior Research Associate
PETER TESSARZ PhD
Cancer Research UK/BBSRC Research Associate
EMMANUELLE VIRÉ PhD
ERC/Cancer Research UK Research Associate
MEIKE WIESE MSc
Cancer Research UK PhD Student
BEATA WYSPIANSKA MSc
BBSRC Case PhD Student
RICK LIVESEY MB BChir PhD
Wellcome Trust Senior Investigator
University Reader in Molecular Neuroscience
(Member of the Department of Biochemistry)
SIAN ALEXANDER DPhil
MRCP Visiting Academic Clinical Fellow
HOZEFA AMIJEE PhD
MRC Research Associate
PHILIPP BERG MRes
Wellcome Trust PhD Student
LAURA BRIGHTMAN BA
Stem Cell Centre Seed Funding Research Assistant
PHILIP BROWNJOHN PhD
Alzheimer’s Research UK Research Associate
TATYANA DIAS PhD
Alzheimer’s Research UK Research Associate
LEWIS EVANS PhD
Wellcome Trust Research Associate
JAYNE FISHER
PA/Secretary
ALBERTO FRANGINI PhD
Wellcome Trust Research Associate
PETER KIRWAN PhD
MRC Research Associate (Formerly Wellcome Trust
Development Biology PhD Student)
TERESA KRIEGER MSci
EPSRC PhD Student (Joint with Ben Simons)
AMELIA McGLADE BSc
Innovative Medicines Initiative Research Assistant
AYIBA MOMOH MSc
Wellcome Trust Research Assistant
STEVEN MOORE PhD
Wellcome Trust Research Associate
TOMOKI OTANI MPhil
Wellcome Trust Developmental Biology PhD Student
MANUEL PETER PhD
Innovative Medicines Initiative Research Associate
NATHALIE SAURAT BSc
Woolf Fisher PhD Student/ARUK Research Associate
JAMES SMITH BSc
Innovative Medicines Initiative Research Associate
VICTORIA STUBBS PhD
Innovative Medicines Initiative Research Assistant
ERIC MISKA PhD
Herchel Smith Professor of Molecular Genetics
Wellcome Trust Senior Investigator
Cancer Research UK Senior Research Fellow
Wellcome Trust Sanger Institute Affiliated Faculty
Member
Member, European Molecular Biology Organization
ERC Independent Starting Researcher
(Member of the Department of Genetics)
ALPER AKAY PhD
Cancer Research UK Research Associate
ALYSON ASHE PhD
Herchel Smith Postdoctoral Fellowship
FABIAN BRAUKMANN MSc
ERC Research Assistant/PhD Student
AMY CORDING BSc
Cancer Research UK Research Assistant
TOMAS di DOMENICO PhD
ERC Research Associate (Bioinformatics)
TANAY GHOSH PhD
ERC/Cancer Research UK Research Associate
SABRINA HUBER MSc
Cambridge PhD Training Programme in Chemical
Biology and Molecular Medicine
JOANNA KOSALKA MPhil
Wellcome Trust Developmental Biology PhD Student
MIRANDA LANDGRAF MA
PA/Secretary
JÉREMIE LE PEN MPhil
Wellcome Trust Developmental Biology/ERC PhD
Student
MILAN MALINSKY MPhil
Wellcome Trust Mathematical Biology PhD Student
EYAL MAORI PhD
Herchel Smith Research Associate
RAGINI MEDHI BSTech
MPhil Student
SYLVIANE MOSS PhD
Cancer Research UK Research Associate/Lab Manager
KENNETH MURFITT MPhil
ERC PhD Student (Formerly Wellcome Trust Development
Biology PhD Student)
MARC RIDYARD PhD
Cancer Research UK Lab Manager
ALEXANDRA SAPETSCHNIG PhD
ERC Senior Research Associate (formerly
PETER SARKIES PhD
Gonville and Caius Research Fellow/CRUK Research
Associate
MÉLANIE TANGUY PhD
Cancer Research UK Research Associate
EVA-MARIA WEICK PhD
ERC PhD Student/Research Associate
JONATHON PINES PhD FMEDSci
Director of Research in Cell Division
Cancer Research UK Senior Research Fellow
Member, European Molecular Biology Organization
(Member of the Department of Zoology)
PHILIPPE COLLIN PhD
BBSRC Research Associate
BARBARA DI FIORE PhD
Cancer Research UK Research Associate
ANJA HAGTING PhD
Cancer Research UK Research Associate, Biological
Safety Officer
ANDREW HARRISON PhD
MRC Research Associate
DAISUKE IZAWA PhD
Cancer Research UK/MRC Research Associate
MARK JACKMAN PhD
Cancer Research UK Research Associate, Chemical
Safety Officer
AGATA LICHAWSKA PhD
Herchel Smith PhD Student
CHIARA MARCOZZI MSc
Boehringer Ingelheim/BBSRC PhD Student
TAKAHIRO MATSUSAKA PhD
Cancer Research UK Research Associate
OXANA NASHCHEKINA MSc
Cancer Research UK Chief Research Technician
BERNHARD STRAUSS PhD
MRC Research Associate
JILL TEMPLE MSc
MRC Research Assistant
SAMUEL WIESER MSc
Liechtenstein Government PhD
Student/ MRC Research Assistant
CLAUDIA WURZENBERGER PhD
Marie Cure Research Intra-European Fellow
KEIKO YATA PhD
MRC Research Associate
THE GURDON INSTITUTE
45
SENIOR GROUP LEADERS / GROUP LEADERS
AZIM SURANI PhD CBE FRS
FMEDSci
Director of Germline and Epigenomics Research
Wellcome Trust Senior Investigator
Member, European Molecular Biology Organization
Member Academia Europaea
Associate Fellow, Third World Academy of Sciences
(Member of the Department of Physiology,
Development and Neuroscience)
DELPHINE COUGOT PhD
Wellcome Trust Research Associate
DANG VINH DO PhD
Wellcome Trust Research Associate
LYNN FROGGETT
PA/Secretary
WOLFRAM GRUHN PhD
Wellcome Trust Research Associate/EMBO Research
Fellow
UFUK GÜNESDOGAN PhD
Marie Curie Intra-European Fellow
JAMIE HACKETT PhD
HFSP Research Associate
YUN HUANG BA
MB-PhD Student
NAOKO IRIE PhD
BIRAX Research Associate
ELENA ITSKOVICH MSc
Wellcome Trust PhD Student
SHINSEOG KIM PhD
Wellcome Trust Research Associate
TOSHIHIRO KOBAYASHI PhD
JSPS Fellow
CAROLINE LEE ONC
Wellcome Trust Chief Research Technician, Radiation
Protection Supervisor
CAROL READHEAD PhD
Visiting Academic
ROOPSHA SENGUPTA PhD
Wellcome Trust Research Associate
WALFRED TANG MPhil
Croucher Cambridge International PhD Student
THOR THEUNISSEN PhD
Sir Henry Wellcome Postdoctoral Fellow (Based at
Whitehead Institute, Boston, US)
46
THE GURDON INSTITUTE
JULIA TISCHLER PhD
APART Research Fellow
JAN ZYLICZ MSc
Wellcome Trust PhD Student
JENNIFER GALLOP PhD
Wellcome Trust Research Career Development Fellow
ERC Independent Starting Researcher
(Member of the Department of Biochemistry)
GUILHERME CORREIA MSc
Wellcome Trust PhD Student
HELEN FOX MPhil
Wellcome Trust Developmental Biology PhD Student
LYNN FROGGETT
PA/Secretary
YOSHIKO INOUE PhD
Wellcome Trust/ERC Research Associate
IRIS JARSCH PhD
Wellcome Trust Research Associate
JULIA MASON BSc
ERC Research Assistant
DANIEL SAXTON BSc
ERC Research Assistant
HANAE SHIMO MSc
Funai Foundation PhD Student
ASTRID WALRANT PhD
ERC Research Associate
MERITXELL HUCH PhD
Wellcome Trust Sir Henry Dale Fellow
Beit Prize Fellow
(Member of the Department of Physiology,
Development and Neuroscience)
LUIGI ALOIA PhD
Wellcome Trust Research Associate
JOHN CRANG DPhil
PA/Secretary
CHRISTOPHER HINDLEY PhD
SCI Seed Funding Research Associate
GIANMARCO MASTROGIOVANNI MSc
Marie Curie Initial Training Network PhD Student
MIKEL McKIE MSci
Wellcome Trust Research Assistant
ALESSANDRA MERENDA MSc
Marie Curie Initial Training Network PhD Student
GROUP LEADERS / VISITING STUDENTS & RESEARCHERS
EUGENIA PIDDINI PhD
Royal Society Research Fellow
(Member of the Department of Zoology)
MICHAEL DINAN MPhil
Wellcome Trust Developmental Biology PhD Student
MAJA GOSCHORSKA MPhil
Cambridge Cancer Centre PhD Student
LEA HAMPTON-O’NEIL BSc
MPhil Student
GOLNAR KOLAHGAR PhD
Cancer Research UK Research Associate
KASIA KOZYRSKA MSc
PhD Student
IWO KUCINSKI MPhil
Wellcome Trust Developmental Biology PhD Student
KATHY OSWALD MA
PA/Secretary
SASKIA SUIJKERBUIJK PhD
Rubicon Research Associate
SILVIA VIVARELLI PhD
Cancer Research UK Research Associate
LAURA WAGSTAFF PhD
Cancer Research UK Research Associate
EMMA RAWLINS PhD
MRC Research Fellow
(Member of the Department of Pathology)
GAYAN BALASOORIYA MSc
MRC Research Technician/PhD Student
CHRISTOPH BUDJAN MPhil
Wellcome Trust Developmental Biology PhD Student
ANGELENE HUFFMAN
PA/Secretary
JO-ANNE JOHNSON MB ChB MRCPCH
Wellcome Trust Clinical Fellow/PhD Student
USUA LARESGOITI GARAY PhD
Wellcome Trust Research Associate
MARKO NIKOLIC MA MB BChir MRCP
Wellcome Trust Clinical Fellow/PhD Student
CHANDRIKA RAO MSc
March of Dimes/Wellcome Trust Chief Research
Technician
PHILIP ZEGERMAN PhD
Worldwide Cancer Research International Research
Fellow
(Member of the Department of Biochemistry)
GEYLANI CAN MSc
Turkish Government/Sackler PhD Student
CLARA COLLART PhD
Collaborating Researcher from Professor Jim Smith’s
lab, MRC NIMR, London
VINCENT GAGGIOLI PhD
Worldwide Cancer Research Research Associate
CHRISTINE HÄNNI MSc
Cambridge Cancer Centre PhD Student
ANGELENE HUFFMAN
PA/Secretary
MARK JOHNSON PhD
Worldwide Cancer Research Research Associate
OLEG KOVALEVSKIY PhD
Cancer Research UK Research Associate
BARBARA SCHÖPF PhD
Worldwide Cancer Research Research Associate
MAGDALENA ZERNICKA-GOETZ PhD
Wellcome Trust Senior Research Fellow
Professor of Developmental Biology
Member, European Molecular Biology Organization
(Member of the Department of Physiology, Development
and Neuroscience)
FRANCESCO ANTONICA PhD
Wellcome Trust Research Associate
PAULA ALMEIDA COELHO PhD
Visiting Academic
IVAN BEDZHOV PhD
Wellcome Trust Research Associate
MONIKA BIALECKA PhD
Wellcome Trust Research Associate
LEAH BURY Diplom Mol Med
Cambridge Cancer Centre PhD Student
ANDY COX PhD
Wellcome Trust Research Assistant
JOHN CRANG DPhil
PA/SECRETARY
MOHAMMED GOOLAM MSc
St John’s College Mary Gray PhD Student
SARAH GRAHAM MSc
PhD Student
AGNIESZKA JEDRUSIK PhD
Wellcome Trust Research Associate (Formerly Wellcome
Trust Developmental Biology PhD Student)
CHUEN YAN LEUNG BSc
PhD Student
MARYNA PANAMAROVA BSc
Darwin Trust PhD Student
GAELLE RECHER PhD
Wellcome Trust Research Associate
MARTA SHAHBAZI ALONSO PhD
Wellcome Trust Research Associate
BEN SIMONS PhD
Herchel Smith Professor of Physics
Cavendish Laboratory
EDOUARD HANNEZO PhD
Postdoctoral Researcher
TERESA KRIEGER BA MSci
EPSRC PhD Student (Joint with Rick Livesey)
STEFFEN RULANDS PhD
Postdoctoral Researcher
VISITING STUDENTS/RESEARCHERS
LYNN ASANTE-ASARE
Biochemical Society Vacation Student, University of
Warwick (Jackson Lab)
ROBERTA AZZARELLI
Collaborating Researcher, Hutchison MRC Research
Centre (Huch Lab)
ADAM BENABID BSc
Visiting Master’s Student, École Normale Supérieure, Paris,
France (Piddini Lab)
LUKE BIBBY
University of Cambridge Vacation Student (Pines Lab)
GINA BLAKE
Wellcome Trust Developmental Biology PhD Rotation
Student (Rawlins Lab)
DANIEL BROOK
University of Cambridge Part III Student (Gallop Lab)
CALIN CAUACEAN
University of Cambridge Part II Student (Pines Lab)
AMY DANSEN
University of Cambridge Part II Student (Livesey Lab)
ALFONSA DIAZ TORRES
Visiting PhD Student, Universidad Pablo Olavide, Seville,
Spain (Brand Lab)
ANA DOMINGUEZ PÉREZ
Vacation Student from University of Navarra, Spain
(Brown Lab)
EMILY DUDGEN
University of Cambridge Part II Student (Piddini Lab)
EMILY FALDON
University of Cambridge Part II Student (Brown Lab)
KIRSTY FERGUSON
University of Cambridge Part III Student (Livesey Lab)
HELENA FRANCIS
University of Cambridge Part III Student (St Johnston
Lab)
MARCOS GALLEGO LLORENTE
BBSRC Rotation PhD Student (Brand Lab)
ANA GASOL GARCIA
Visiting Master’s Student, Vrije University, Amsterdam,
Netherlands (Piddini Lab)
ZAMIRA GUERRA SOARES
Visiting PhD Student, UFMG, Brazil (Miska Lab)
ROSANNA HANCOCK
University of Cambridge Part II Student (Zernicka-Goetz
Lab)
OLIVIA HARRIS
Wellcome Trust Rotation PhD Student (Stem Cell,
Huch Lab)
SARAH HERBERG BSc
Visiting Master’s Erasmus Student, University of Bayreuth
(Gurdon Lab)
EVA HIGGINBOTHAM
Wellcome Trust Developmental Biology Rotation PhD
Student (Surani Lab)
ISABEL JOHNSON
Visiting Student, University of Wisconsin, USA (Ahringer
Lab)
BARANSEL KAMAZ
Dr Hadwen Trust Vacation Student, Hacettepe University,
Ankara, Turkey (Huch Lab)
CHRISTINE KLEINERT
Visiting Erasmus Master’s Student, Potsdam University,
Germany (Zegerman Lab)
ANNA KONONEN
Visiting Erasmus Administrator, University of Jyväskylä,
FInland (Administration)
BART KRAMER BSc
Visiting Master’s Erasmus Student, University of
Utrecht, Netherlands (Huch Lab)
THE GURDON INSTITUTE
47
VISITING STUDENTS & RESEARCHERS / SUPPORT STAFF
RIINA LAMPELA
Wellcome Trust Rotation PhD Student (Livesey Lab)
ELODIE LE CLECH
Vacation Student, Sophia Antipolis University, Nice, France
(Rawlins Lab)
KATHERINE LEE
University of Cambridge Vacation Student (Brand Lab)
SANG EUN LEE
Visiting Student, Imperial College, London (Huch Lab)
JONATHAN LIANG
MPhil Student, Computational Biology, University of
Cambridge (Ahringer Lab)
PING MA PhD
Visiting Researcher, University of Georgia, Athens USA
(Miska Lab)
ALISE MOLOTOVA
Amgen Scholar (Gurdon Lab)
MATE NASZAI
University of Cambridge Part II Student (Rawlins Lab)
JOSEPH NELSON
University of Cambridge Part II Student (Ahringer Lab)
JAMES PICKLES
Visiting PhD Student, Northern Institute for Cancer
Research, Newcastle University, UK (Jackson Lab)
ZOE PILLIDGE
BBSRC DTP Rotation Student (Piddini Lab)
SARA PRECIADO PhD
Visiting Marie Curie Fellow, QMUL, London (Jackson
Lab)
CONOR REID
University of Cambridge Part II Student (St Johnston Lab)
TIMO REY
University of Cambridge Part II Student (Zegerman Lab)
ANDREW RUMBOL
University of Cambridge Part III Student (Pines Lab)
THOMAS SANFORD
University of Cambridge Part III Student (Jackson Lab)
LISA SCHUBERT
Visiting Master’s Student, University of Heidelberg,
Germany (Zegerman Lab)
AISHWARYA SIVAKUMAR
Vacation Student, University of Pune, India (Jackson Lab)
LIOR SODAY
University of Cambridge Part III Student (Miska Lab)
TERESA TOUDAL KNUDSEN
Part II Student (Brown Lab)
48
THE GURDON INSTITUTE
DENIS TORRE
Visiting Undergraduate Student, Trieste University, Italy
(Livesey Lab)
BENITA TURNER-BRIDGER
Wellcome Trust Developmental Biology Rotation PhD
Student (Livesey Lab)
ANTOINETTE van OUWERKERK BSc
Visiting Master’s Student, University of Leiden, The
Netherlands (Surani Lab)
GREGOIRE VERNAZ
Visiting Master’s Student, University of Zurich, Switzerland
(Miska Lab)
LOUISE VERON
Visiting Master’s Student, ENS Cachan, France (Miska
Lab)
SAMUEL WATTRUS
Vacation Student, Harvard Stem Cell Institute, Mass, USA
(Gurdon Lab)
STEFANIE WEISS
Visiting Master’s Student, Ludwig-Maximilians University,
Munich (Miska Lab)
LOUISE WHITELEY
University of Cambridge Part II Student (Gallop Lab)
EMMA WICTOME
University of Cambridge Part II Student (Brown Lab)
BEVERLEY WILSON
University of Cambridge Part II Student (Jackson Lab)
NANCY ZHENG
University of Cambridge Part II Student (Brand Lab
SUPPORT STAFF
ADMINISTRATION
BIOINFORMATICS
TECHNICAL SUPPORT
ANN CARTWRIGHT MPhil
Institute Administrator
SUZANNE CAMPBELL BSc
HR/Grants Manager
JANE COURSE
Accounts Manager
DIANE FOSTER
Deputy Administrator
KATHY HILTON DipMgm
CBSG Manager
ANGELENE HUFFMAN
Receptionist
CEZARY KUCEWICZ MA
Accounts/Clerical Assistant
LYNDA LOCKEY
Office Manager
JANET MOORE PhD
Receptionist
SYLVIANE MOSS PhD
Specialised Facilities Manager - Biological Safety Officer
CHARLES BRADSHAW PhD
Bioinformatician
GEORGE ALLEN PhD
Bioinformatician
POLLY ATTLESEY
Unit Manager
RYAN ASBY MELODIE AVAKIAN BSc
LUKE ATTLESEY
CAROLINE BLAKE
ELEANOR DALE
NICOLA EVANS-BAILEY
SOPHIE GARNHAM
MARK GILLILAND
SAMANTHA HANNA BSc
RICHARD HARPER
JACK HARRIS
GILLIAN HYNES
BEN JAGGS
WEN JIN
SHANE JOHNSON THERESE JONES-GREEN BSc
URSZULA KOKOT
ADAM LAW
GRACE LUCAS
ASHLEIGH MATTHEWS
FALLON MILLER
LORRAINE MILLER
ZOE MUMFORD
RACHEL MURFITT
DOMINIC OSBORNE
NIGEL PECK
JASON RISEBOROUGH
HANNAH RULE
DAVID SIMPSON
DEAN SWINDEN
DANIEL WATTS
COMPUTING
ALASTAIR DOWNIE
Computer Systems Manager
RICHARD BUTLER PhD
Research Associate (Imaging)
NICOLA LAWRENCE PhD
Computer Imaging Associate, Laser Safety Officer
NIGEL SMITH
Computer Associate
ALEX SOSSICK BSc
Computer Imaging Associate, Laser Safety Officer
PETER WILLIAMSON BSc
Computer Associate
ACCOUNTS/PURCHASING/STORES
IAN FLEMING
Stores/Purchasing Manager
SIMON ALDIS
Purchasing/Accounts Assistant
DAVID COOPER
Stores Technician
RICHARD ETTERIDGE MA
Purchasing/Accounts Assistant
ANDY VINCENT
Senior Stores Technician
MICK WOODROOFE
Purchasing/Accounts Assistant
COMBINED BUILDINGS SERVICES
GROUP (CBSG)
CLIVE BENNETT
ALAN RIX
JOEL SHUBROOKS
SIMON WILSON
KATHERINE BENNETT
STEPHEN SALT
PAUL TURRELL
MEDIA/GLASS WASHING
JUANITA BAKER-HAY
Media/Glass Washing Manager
SUE HUBBARD
Deputy Media/Glass Washing Manager
JANIS ABBOTT
LISA BAKER
KAZUKO COLLINS
VINCE DAMS
SANDRA HUMAN
MARK JOHNS
TRACY MITCHELL
ZEST CATERING
AMANDA HARRIS
MELISSA PLOWDEN ROBERTS
THE GURDON INSTITUTE
49
INSTITUTE PUBLICATIONS
The following is a list of articles by members of the Institute that were either
published or accepted for publication, since the date of publication of our last
Annual Report.
INSTITUTE PUBLICATIONS
8
Baker AM, Cereser B, Melton S, Fletcher AG, Rodriguez-Justo M, Tadrous
PJ, Humphries A, Elia G, McDonald SA, Wright NA, Simons BD, Jansen M
and Graham TA (2014) Quantification of crypt and stem cell evolution in
the normal and neoplastic human colon. Cell Rep 8, 4: 940 – 947
26
Di Domenico T, Potenza E, Walsh I, Parra RG, Giollo M, Minervini
G, Piovesan D, Ihsan A, Ferrari C, Kajava AV and Tosatto SC (2014)
RepeatsDB: a database of tandem repeat protein structures. Nucleic Acids Res 42, Database issue: D352 – D357 [Miska Group]
27
Forment JV, Flipphi M, Ventura L, González R, Ramón D and Maccabe AP
(2014) High-affinity glucose transport in aspergillus nidulans is mediated
by the products of two related but differentially expressed genes. PLoS One 9, 4: e94662 [Jackson Group]
28
Frey A, Listovsky T, Guilbaud G, Sarkies P and Sale JE (2014) Histone H3.3
is required to maintain replication fork progression after UV damage.
Curr Biol 24, 18: 2195 – 2201 [Miska Group]
1
Ajduk A, Biswas Shivhare S and Zernicka-Goetz M (2014) The basal
position of nuclei is one pre-requisite for asymmetric cell divisions in the
early mouse embryo. Dev Biol 392, 2: 133 – 140
9
2
Alcolea MP, Greulich P, Wabik A, Frede J, Simons BD and Jones PH (2014)
Differentiation imbalance in single oesophageal progenitor cells causes
clonal immortalization and field change. Nat Cell Biol 16, 6: 615 – 622
Balmus G and McIntyre RE (2014) Genetic screens in mice for genome
integrity maintenance and cancer predisposition. Current Opinion in
Genetics and Development 24, 1: 1 – 7 [Jackson Group]
10
3
Ali FR, Cheng K, Kirwan P, Metcalfe S, Livesey FJ, Barker RA and Philpott
A (2014) The phosphorylation status of Ascl1 is a key determinant of
neuronal differentiation and maturation in vivo and in vitro. Development 141, 11: 2216 – 2224
Barbera M, di Pietro M, Walker E, Brierley C, Macrae S, Simons BD, Jones
PH, Stingl J and Fitzgerald RC (2014) The human squamous oesophagus
has widespread capacity for clonal expansion from cells at diverse stages
of differentiation. Gut 64, 1: 11 – 9
11
Barbieri I and Bannister AJ (2014) Evolution of cancer cell resistance
versus intelligent design of epigenetic drugs. Drug Discovery Today:
Disease Models DOI: 10.1016/j.ddmod [Kouzarides Group]
29
Gaggioli V, Zeiser E, Rivers D, Bradshaw CR, Ahringer J and Zegerman P
(2014) CDK phosphorylation of SLD-2 is required for replication initiation
and germline development in C. elegans. J Cell Biol 204, 6: 1075
12
Bartfeld S, Bayram T, van de Wetering M, Huch M, Begthel H, Kujala P,
Vries R, Peters PJ and Clevers H (2014) In vitro expansion of human
gastric epithelial stem cells and their responses to bacterial infection.
Gastroenterology 148, 1: 126 – 136
30
13
Bedzhov I, Graham SJ, Leung CY and Zernicka-Goetz M (2014)
Developmental plasticity, cell fate specification and morphogenesis in the
early mouse embryo. Philos Trans R Soc Lond B Biol Sci 369, 1657 DOI: 10.1098/rstb
Gao P, Postiglione MP, Krieger TG, Hernandez L, Wang C, Han Z, Streicher
C, Papusheva E, Insolera R, Chugh K, Kodish O, Huang K, Simons BD, Luo
L, Hippenmeyer S and Shi SH (2014) Deterministic progenitor behavior
and unitary production of neurons in the neocortex. Cell 159, 4: 775 – 788
31
14
Bedzhov I, Leung CY, Bialecka M and Zernicka-Goetz M (2014) In vitro
culture of mouse blastocysts beyond the implantation stages. Nat Protoc 9, 12: 2732 – 2739
Gapp K, Jawaid A, Sarkies P, Bohacek J, Pelczar P, Prados J, Farinelli L, Miska
E and Mansuy IM (2014) Implication of sperm RNAs in transgenerational
inheritance of the effects of early trauma in mice. Nat Neurosci 17, 5: 667 – 669
32
15
Bedzhov I and Zernicka-Goetz M (2014) Self-organizing properties of
mouse pluripotent cells initiate morphogenesis upon implantation. Cell 156, 5: 1032 – 1044
Gardiol A and St Johnston D (2014) Staufen targets coracle mRNA
to Drosophila neuromuscular junctions and regulates GluRIIA synaptic
accumulation and bouton number. Dev Biol 392, 2: 153 – 167
33
Gold KS and Brand AH (2014) Optix defines a neuroepithelial
compartment in the optic lobe of the Drosophila brain. Neural Dev 9: 18
16
Behjati S, Huch M, van Boxtel R, Karthaus W, Wedge DC, Tamuri AU,
Martincorena I, Petljak M, Alexandrov LB, Gundem G, Tarpey PS, Roerink
S, Blokker J, Maddison M, Mudie L, Robinson B, Nik-Zainal S, Campbell
P, Goldman N, van de Wetering M, Cuppen E, Clevers H and Stratton
MR (2014) Genome sequencing of normal cells reveals developmental
lineages and mutational processes. Nature 513, 7518: 422 – 425
34
Gover O, Peretz Y, Mozes-Koch R, Maori E, Rabinowitch HD and Sela I
(2014) Only minimal regions of tomato yellow leaf curl virus (TYLCV) are
required for replication, expression and movement. Arch Virol 159, 9: 2263 – 2274 [Miska Group]
35
Grallert A, Boke E, Hagting A, Hodgson B, Connolly Y, Griffiths JR, Smith
DL, Pines J and Hagan IM (2014) A PP1-PP2A phosphatase relay controls
mitotic progression. Nature 517, 7532: 94 – 98
36
Graml V, Studera X, Lawson JL, Chessel A, Geymonat M, Bortfeld-Miller
M, Walter T, Wagstaff L, Piddini E and Carazo-Salas RE (2014) A genomic
multiprocess survey of machineries that control and link cell shape,
microtubule organization, and cell-cycle progression. Dev Cell 31, 2: 227 – 239
37
Günesdogan U, Jäckle H and Herzig A (2014) Histone supply regulates S
phase timing and cell cycle progression. Elife 3, e02443 [Surani Group]
38
Günesdogan U, Magnúsdóttir E and Surani MA (2014) Primoridal germ
cell specification: a context-dependent cellular differentiation event. Philos Trans R Soc Lond B Biol Sci 369, 1657
4
Amaral PP and Bannister AJ (2014) Re-place your BETs: the dynamics of
super enhancers. Mol Cell 56, 2: 187 – 189 [Kouzarides Group]
5
Amoyel M, Simons BD and Bach EA (2014) Neutral competition of stem
cells is skewed by proliferative changes downstream of Hh and Hpo.
EMBO J 33, 20: 2295 – 2313
6
Atkin J, Halova L, Ferguson J, Hitchin JR, Lichawska-Cieslar A, Jordan AM,
Pines J, Wellbrock C and Petersen J (2014) Torin1-mediated TOR kinase
inhibition reduces Wee1 levels and advances mitotic commitment in
fission yeast and HeLa cells. J Cell Sci 127, Pt 6: 1346 – 1356
7
Aymard F, Bugler B, Schmidt CK, Guillou E, Caron P, Briois S, Iacovoni JS,
Daburon V, Miller KM, Jackson SP and Legube G (2014) Transcriptionally
active chromatin recruits homologous recombination at DNA doublestrand breaks. Nat Struct Mol Biol 21, 4: 366 – 374
Branching E11.5 mouse embryonic lung stained for ubiquitous lung marker, Nkx2.1
(yellow) & Hoechst (blue). (Christoph Budjan, Rawlins Group)
50
THE GURDON INSTITUTE
17
Belotserkovskaya R and Jackson SP (2014) Keeping 53BP1 out of focus in
mitosis. Cell Res 24, 7: 781 – 782
18
Bergstralh DT and St Johnston D (2014) Spindle orientation: what if it
goes wrong? Semin Cell Dev Biol 34, 140 – 145
19
Blasius M, Wagner SA, Choudhary C, Bartek J and Jackson SP (2014) A
quantitative 14-3-3 interaction screen connects the nuclear exosome
targeting complex to the DNA damage response. Genes Dev 28, 18: 1977 – 1982
20
Brownjohn PW and Ashton JC (2014) What can be concluded from
blocking peptide controls? Appl Immunohistochem Mol Morphol 22, 8: 634 [Livesey Group]
21
Brownjohn PW, Reynolds JN, Matheson N, Fox J and Shemmell JB (2014)
The effects of individualized theta burst stimulation on the excitability of
the human motor system. Brain Stimul 7, 2: 260 – 268 [Livesey Group]
Beta-tubulin staining for microtubules with DAPI counterstain, on cells which were
cold-treated to induce microtubule depolymerisation and UV-treated to induce cell
stress. (Laura Wagstaff, Piddini Group, in collaboration with Rafael Carazo-Salas.)
22
Centanin L, Ander JJ, Hoeckendorf B, Lust K, Kellner T, Kraemer I, Urbany
C, Hasel E, Harris WA, Simons BD and Wittbrodt J (2014) Exclusive
multipotency and preferential asymmetric divisions in post-embryonic
neural stem cells of the fish retina. Development 141, 18: 3472 – 3482
23
Chen RA, Stempor P, Down TA, Zeiser E, Feuer SK and Ahringer J (2014)
Extreme HOT regions are CpG-dense promoters in C. elegans and
humans. Genome Res 24, 7: 1138 – 1146
24
Christophorou MA, Castelo-Branco G, Halley-Stott RP, Oliveira CS, Loos
R, Radzisheuskaya A, Mowen KA, Bertone P, Silva JC, Zernicka-Goetz M,
Nielsen ML, Gurdon JB and Kouzarides T (2014) Citrullination regulates
pluripotency and histone H1 binding to chromatin. Nature 507, 7490: 104 – 108
25
Di Cerbo V, Mohn F, Ryan DP, Montellier E, Kacem S, Tropberger P, Kallis
E, Holzner M, Hoerner L, Feldmann A, Richter FM, Bannister AJ, Mittler
G, Michaelis J, Khochbin S, Feil R, Schuebeler D, Owen-Hughes T, Daujat S
and Schneider R (2014) Acetylation of histone H3 at lysine 64 regulates
nucleosome dynamics and facilitates transcription. Elife 3:e01632 [Kouzarides Group]
THE GURDON INSTITUTE
51
INSTITUTE PUBLICATIONS
INSTITUTE PUBLICATIONS
39
Hackett JA and Surani MA (2014) Regulatory principles of pluripotency:
from the ground state up. Cell Stem Cell 15, 4: 416 – 430
60
Livesey FJ (2014) Human stem cell models of dementia. Hum Mol Genet 23, R1: R35 – R39
40
Halley-Stott RP, Jullien J, Pasque V and Gurdon J (2014) Mitosis gives a
brief window of opportunity for a change in gene transcription. PLoS Biol 12, 7
61
41
Hara K, Nakagawa T, Enomoto H, Suzuki M, Yamamoto M, Simons BD and
Yoshida S (2014) Mouse spermatogenic stem cells continually interconvert
between equipotent singly isolated and syncytial states. Cell Stem Cell 14, 5: 658 – 672
Lowe N, Rees JS, Roote J, Ryder E, Armean IM, Johnson G, Drummond
E, Spriggs H, Drummond J, Magbanua JP, Naylor H, Sanson B, Bastock R,
Huelsmann S, Trovisco V, Landgraf M, Knowles-Barley S, Armstrong JD,
White-Cooper H, Hansen C, Phillips RG, UK Drosophila Protein Trap
Screening Consortium, Lilley KS, Russell S and St Johnston D (2014)
Analysis of the expression patterns, subcellular localisations and interaction
partners of Drosophila proteins using a pigP protein trap library.
Development 141, 20: 3994 – 4005
42
43
Herzig B, Yakulov TA, Klinge K, Günesdogan U, Jäckle H and Herzig A
(2014) Bällchen is required for self-renewal of germline stem cells in
Drosophila melanogaster. Biol Open 3, 6: 510 – 521 [Surani Group]
62
Hindley CJ, Mastrogiovanni G and Huch M (2014) The plastic liver:
differentiated cells, stem cells, every cell? J Clin Invest 124, 12: 5099 – 5102
44
Ho JW, Chen RA, Ahringer J et al. (2014) Comparative analysis of
metazoan chromatin organization. Nature 512, 7515: 449 – 452
45
Huch M, Gehart H, van Boxtel R, Hamer K, Blokzijl F, Verstegen MM, Ellis
E, van Wenum M, Fuchs SA, de Ligt J, van de Wetering M, Sasaki N, Boers
SJ, Kemperman H, de Jonge J, Ijzermans JN, Nieuwenhuis EE, Hoekstra R,
Strom S, Vries RR, van der Laan LJ, Cuppen E and Clevers H (2014) Longterm culture of genome-stable bipotent stem cells from adult human liver.
Cell 160, 1: 299 – 312
3D reconstruction of Drosophila adult posterior midgut. The oxygen-supplying trachea
is shown in yellow and nuclei in of the gut epithelium in blue. (Saskia Suijkerbuijk,
Piddini Group)
53
Koch B, Sanchez S, Schmidt CK, Swiersy A, Jackson SP and Schmidt OG
(2014) Confinement and deformation of single cells and their nuclei inside
size-adapted microtubes. Adv Healthc Mater 3, 11: 1753 – 1758
Korzelius J, Naumann SK, Loza-Coll MA, Chan JS, Dutta D, Oberheim J,
Gläßer C, Southall TD, Brand AH, Jones DL and Edgar BA (2014) Escargot
maintains stemness and suppresses differentiation in Drosophila intestinal
stem cells. EMBO J 33, 24: 2967 – 2982
46
Irie N, Tang WW, and Surani MA (2014) Germ cell specification and
pluripotency in mammals: a perspective from early embryogenesis.
Reprod Med Biol 13, 4: 203 – 215
54
47
Ismail HM, Hurd PJ, Khalil MI, Kouzarides T, Bannister A and Gout I (2014)
S6 kinase 2 is bound to chromatin-nuclear matrix cellular fractions and is
able to phosphorylate histone H3 at threonine 45 in vitro and in vivo. J Cell Biochem 115, 6: 1048 – 1062
55
48
Izawa D and Pines J (2014) The mitotic checkpoint complex binds a
second CDC20 to inhibit active APC/C. Nature doi: 10.1038/nature13911
Larrieu D, Britton S, Demir M, Rodriguez R and Jackson SP (2014)
Chemical inhibition of NAT10 corrects defects of laminopathic cells.
Science 344, 6183: 527 – 532
56
49
Jullien J, Miyamoto K, Pasque V, Allen GE, Bradshaw CR, Garrett NJ,
Halley-Stott RP, Kimura H, Ohsumi K and Gurdon JB (2014) Hierarchical
molecular events driven by oocyte-specific factors lead to rapid and
extensive reprogramming. Mol Cell 55, 4: 524 – 536
Larrieu D, Rodriguez R and Britton S (2014) Chemical inhibition of
NAT10 corrects defects of laminopathic cells. Médecine Sciences 30, 8-9:
745-747 [Jackson Group]
57
Lescroart F, Chabab S, Lin X, Rulands S, Paulissen C, Rodolosse A, Auer H,
Achouri Y, Dubois C, Bondue A, Simons BD and Blanpain C (2014) Early
lineage restriction in temporally distinct populations of Mesp1 progenitors
during mammalian heart development. Nat Cell Biol 16, 9: 829 – 840
58
Liang H, Esposito A, De S, Ber S, Collin P, Surana U and Venkitaraman AR
(2014) Homeostatic control of polo-like kinase-1 engenders non-genetic
heterogeneity in G2 checkpoint fidelity and timing. Nat Commun 5: 4048 [Pines Group]
59
Liu J, Spéder P and Brand AH (2014) Control of brain development and
homeostasis by local and systemic insulin signalling. Diabetes Obes Metab 16 Suppl 1: 16 – 20
50
Keeling J, Bhaseen MJ and Simons BD (2014) Fermionic superradiance in a
transversely pumped optical cavity. Phys Rev Lett 112, 14
51
Kim S, Günesdogan U, Zylicz JJ, Hackett JA, Cougot D, Bao S, Lee C,
Dietmann S, Allen GE, Sengupta R and Surani MA (2014) PRMT5 protects
genomic integrity during global DNA demethylation in primordial germ
cells and preimplantation embryos Mol Cell 56, 4: 564 – 579
52
Knobel PA, Belotserkovskaya R, Galanty Y, Schmidt CK, Jackson SP and
Stracker TH (2014) USP28 is recruited to sites of DNA damage by the
tandem BRCT domains of 53BP1 but plays a minor role in double-strand
break metabolism. Mol Cell Biol 34, 11: 2062 – 2074
52
THE GURDON INSTITUTE
Loza-Coll MA, Southall TD, Sandall SL, Brand AH and Jones DL
(2014) Regulation of Drosophila intestinal stem cell maintenance and
differentiation by the transcription factor Escargot. EMBO J 33, 24: 2983 – 2996
63
Magnúsdóttir E and Surani MA (2014) How to make a primordial germ
cell. Development 141, 2: 245 – 252
64
Malm AV, Harrison AW and Waigh TA (2014) Optical coherence
tomography velocimetry of colloidal suspensions. Soft Matter 10, 41: 8210 – 8215 [Pines Group]
65
Marchalant Y, Brownjohn PW, Bonnet A, Kleffmann T and Ashton JC
(2014) Validating antibodies to the cannabinoid CB2 receptor: antibody
sensitivity is not evidence of antibody specificity. J Histochem Cytochem 62, 6: 395 – 404 [Livesey Group]
66
Matsusaka T, Enquist-Newman M, Morgan DO and Pines J (2014) Coactivator independent differences in how the metaphase and anaphase
APC/C recognise the same substrate. Biol Open 3, 10: 904 – 912
67
Morais-de-Sá E, Mukherjee A, Lowe N and St Johnston D (2014) Slmb
antagonises the aPKC/Par-6 complex to control oocyte and epithelial
polarity. Development 141, 15: 2984 – 2992
68
Neville MC, Nojima T, Ashley E, Parker DJ, Walker J, Southall T, Van
de Sande B, Marques AC, Fischer B, Brand AH, Russell S, Ritchie MG,
Aerts S and Goodwin SF (2014) Male-specific fruitless isoforms target
neurodevelopmental genes to specify a sexually dimorphic nervous
system. Curr Biol 24, 3: 229 – 241
69
Nishi R, Wijnhoven P, le Sage C, Tjeertes J, Galanty Y, Forment JV, Clague
MJ, Urbé S and Jackson SP (2014) Systematic characterization of
deubiquitylating enzymes for roles in maintaining genome integrity. Nat Cell Biol 16, 10: 1016 – 18
70
Okayama S, Kopelovich L, Balmus G, Weiss RS, Herbert BS, Dannenberg
AJ and Subbaramaiah K (2014) p53 protein regulates Hsp90 ATPase
activity and thereby Wnt signaling by modulating Aha1 expression. J Biol Chem 289, 10: 6513 – 6525 [Jackson Group]
71
Olsson B, Legros L, Guilhot F, Strömberg K, Smith J, Livesey FJ, Wilson DH,
Zetterberg H and Blennow K (2014) Imatinib treatment and Aβ42 in
humans. Alzheimer’s and Dementia 10, 5: S374 – S380
72
Otsuki L, Cheetham SW and Brand AH (2014) Freedom of expression:
Cell-type-specific gene profiling. Wiley Interdisciplinary Reviews:
Developmental Biology 10.1002/wdev.149
73
Polato F, Callen E, Wong N, Faryabi R, Bunting S, Chen HT, Kozak M,
Kruhlak MJ, Reczek CR, Lee WH, Ludwig T, Baer R, Feigenbaum L, Jackson
S and Nussenzweig A (2014) CtIP-mediated resection is essential
for viability and can operate independently of BRCA1. Journal of
Experimental Medicine 211, 6: 1027-1036
74
Rawlins EL and Giangreco A (2014) The best laid schemes of airway
repair. Eur Respir J 44, 2: 299 – 301
75
Ritsma L, Ellenbroek SI, Zomer A, Snippert HJ, de Sauvage FJ, Simons BD,
Clevers H and van Rheenen J (2014) Intestinal crypt homeostasis revealed
at single-stem-cell level by in vivo live imaging. Nature 507, 7492: 362 – 365
76
Rodriguez-Bravo V, Maciejowski J, Corona J, Buch HK, Collin P, Kanemaki
MT, Shah JV and Jallepalli PV (2014) Nuclear pores protect genome
integrity by assembling a premitotic and Mad1-dependent anaphase
inhibitor. Cell 156, 5: 1017 – 1031 [Pines Group]
77
Rosenthal N and Zernicka-Goetz M (2014) A tribute to Sir John Gurdon.
Differentiation 88, 1: 1 – 2
Confocal image of human embryonic lung tissue (70 days old from date of
conception), stained for Sox9 in green (marking the crucial multipotent progenitor
population of the lung in the distal tip) and Sox2 in red (marking the proximal stalk)
(Marko Nikolic, Rawlins Group)
THE GURDON INSTITUTE
53
INSTITUTE PUBLICATIONS
78
Roshan A, Simons BD, Murai K and Jones PH (2014) Live imaging of
human keratinocytes reveals two modes of cell proliferation. British Journal of Surgery 101: 5 – 5
79
Sarkies P and Miska EA (2014) Small RNAs break out: the molecular cell
biology of mobile small RNAs. Nat Rev Mol Cell Biol 15, 8: 525 – 535
80
Savage KI, Gorski JJ, Barros EM, Irwin GW, Manti L, Powell AJ, Pellagatti A,
Lukashchuk N, McCance DJ, McCluggage WG, Schettino G, Salto-Tellez M,
Boultwood J, Richard DJ, McDade SS and Harkin DP (2014) Identification
of a BRCA1-mRNA splicing complex required for efficient DNA repair
and maintenance of genomic stability. Molecular Cell 54, 3: 445-459
[Jackson Group]
81
82
83
Schiavone D, Guilbaud G, Murat P, Papadopoulou C, Sarkies P, Prioleau
MN, Balasubramanian S and Sale JE (2014) Determinants of G
quadruplex-induced epigenetic instability in REV1-deficient cells. EMBO J 33, 21: 2507 – 2520 [Miska Group]
Shulman JM, Imboywa S, Giagtzoglou N, Powers MP, Hu Y, Devenport D,
Chipendo P, Chibnik LB, Diamond A, Perrimon N, Brown NH, De Jager
PL and Feany MB (2014) Functional screening in Drosophila identifies
Alzheimer’s disease susceptibility genes and implicates Tau-mediated
mechanisms. Hum Mol Genet 23, 4: 870 – 877
Simmini S, Bialecka M, Huch M, Kester L, van de Wetering M, Sato T, Beck
F, van Oudenaarden A, Clevers H and Deschamps J (2014) Transformation
of intestinal stem cells into gastric stem cells on loss of transcription factor
Cdx2. Nat Commun 5: 5728
84
Simon M, Sarkies P, Ikegami K, Doebley AL, Goldstein LD, Mitchell J,
Sakaguchi A, Miska EA and Ahmed S (2014) Reduced insulin/IGF-1
signaling restores germ cell immortality to Caenorhabditis elegans Piwi
mutants. Cell Rep 7, 3: 762 – 773
85
Singer ZS, Yong J, Tischler J, Hackett JA, Altinok A, Surani MA, Cai L and
Elowitz MB (2014) Dynamic heterogeneity and DNA methylation in
embryonic stem cells. Mol Cell 55, 2: 319 – 331
86
Southall TD, Davidson CM, Miller C, Carr A and Brand AH (2014)
Dedifferentiation of neurons precedes tumor formation in Lola mutants.
Dev Cell 28, 6: 685 – 696
87
Spéder P and Brand AH (2014) Gap junction proteins in the blood-brain
barrier control nutrient-dependent reactivation of Drosophila neural stem
cells. Dev Cell 30, 3: 309 – 321
88
Teperek M, Miyamoto K, Simeone A, Feret R, Deery MJ, Gurdon JB and
Jullien J (2014) Sperm and spermatids contain different proteins and bind
distinct egg factors. Int J Mol Sci 15, 9: 16719 – 16740
89
Tessarz P and Kouzarides T (2014) Histone core modifications regulating
nucleosome structure and dynamics. Nat Rev Mol Cell Biol 15, 11: 703 – 708
90
Tuysuz N, Koole W, Garcia TM, van Til N, Wagemaker G, Huch M, Vries RG
and Clevers H (2014) Gene therapy of Type I mucopolysaccharidosis by
liver organ stem cells in the mouse model. Human Gene Therapy 25, 11: A63 – A63
54
THE GURDON INSTITUTE
TALKS BY INSTITUTE RESEARCHERS
91
Viré E, Curtis C, Davalos V, Git A, Robson S, Villanueva A, Vidal A, Barbieri
I, Aparicio S, Esteller M, Caldas C and Kouzarides T (2014) The breast
cancer oncogene EMSY represses transcription of antimetastatic
microRNA miR-31. Mol Cell 53, 5: 806 – 818
92
Walsh I, Di Domenico T and Tosatto SC (2014) RUBI: rapid proteomicscale prediction of lysine ubiquitination and factors influencing predictor
performance. Amino Acids 46, 4: 853 – 862 [Miska Group]
93
Walsh I, Giollo M, Di Domenico T, Ferrari C, Zimmermann O and Tosatto
SC (2014) Comprehensive large-scale assessment of intrinsic protein
disorder. Bioinformatics 31, 2: 201 – 208 [Miska Group]
94
Weick EM, Sarkies P, Silva N, Chen RA, Moss SM, Cording AC, Ahringer
J, Martinez-Perez E and Miska EA (2014) PRDE-1 is a nuclear factor
essential for the biogenesis of Ruby motif-dependent piRNAs in C. elegans.
Genes Dev 28, 7: 783 – 796
95
Wickramasinghe VO, Andrews R, Ellis P, Langford C, Gurdon JB, Stewart
M, Venkitaraman AR and Laskey RA (2014) Selective nuclear export of
specific classes of mRNA from mammalian nuclei is promoted by GANP.
Nucleic Acids Res 42, 8: 5059 – 5071
96
Wilson MT, Goodwin DP, Brownjohn PW, Shemmell J and Reynolds JN
(2014) Numerical modelling of plasticity induced by transcranial magnetic
stimulation. J Comput Neurosci 36, 3: 499 – 514 [Livesey Group]
97
Wolfram V, Southall TD, Günay C, Prinz AA, Brand AH and Baines RA
(2014) The transcription factors islet and Lim3 combinatorially regulate
ion channel gene expression. J Neurosci 34, 7: 2538 – 2543
98
Xi W, Schmidt CK, Sanchez S, Gracias DH, Carazo-Salas RE, Jackson SP
and Schmidt OG (2014) Rolled-up functionalized nanomembranes as
three-dimensional cavities for single cell studies. Nano Lett 14, 8: 4197 – 4204
99
Xu H, Xian J, Vire E, McKinney S, Wei V, Wong J, Tong R, Kouzarides T,
Caldas C, Aparicio S (2014) Up-regulation of the interferon-related genes
in BRCA2 knockout epithelial cells. J Pathol 234, 3: 386 – 397
100 Yakulov T, Günesdogan U, Jäckle H and Herzig A (2014) Bällchen
participates in proliferation control and prevents the differentiation of
Drosophila melanogaster neuronal stem cells. Biol Open 3, 10: 881 – 886
101 Yata K, Bleuyard J-Y, Nakato R, Ralf C, Katou Y, Schwab R, Niedzwiedz W,
Shirahige K and Esashi F (2014) BRCA2 coordinates the activities of cellcycle kinases to promote genome stability. Cell Reports 7, 5: 1547 – 1559 [Pines Group]
102 Zernicka-Goetz M and Hadjantonakis AK (2014) From pluripotency
to differentiation: laying foundations for the body pattern in the mouse
embryo. Philos Trans R Soc Lond B Biol Sci 369, 1657
JANUARY
JOHN GURDON: Medical Faculty: Annal Medical Fellows/Students, Cambridge,
UK
TONY KOUZARIDES: HHMI/NYU Langone Medical Center, New York, New
York, USA
ERIC MISKA: Genome Dynamics Seminar Series, Montpelier, France
ERIC MISKA: Barbados micRNA Anniversary Meeting, Bridgetown, Barbados
BEN SIMONS: Curie Institute, Paris, France
AZIM SURANI: Insitute for Stem Cell Biology and Regenerative Medicine,
Bangalore, India
PHILIP ZEGERMAN: University of Nottingham, UK
FEBRUARY
ANDREA BRAND: LS2 (Life Sciences Switzerland) Annual Meeting, Lausanne,
Switzerland
ANDREA BRAND: Developmental Biology Society Meeting, Paris, France
ANDREA BRAND: 7th Asian Pacific Organization for Cell Biology Congress,
Singapore
JOHN GURDON: Bangalore Bio: biotech for a better tomorrow, Bangalore,
India
JOHN GURDON: Indian Institute of Science, Bangalore, India
JOHN GURDON: Old Etonian Association, London, UK
JOHN GURDON: Cambridge Enterprise and Technology Club, Cambridge, UK
JOHN GURDON: Kings College, London, UK
MERITXELL HUCH: 3Rs Prize Lecture, London, UK
MERITXELL HUCH: 3-D Cell Cultures & Drug Discovery, Vienna, Austria
MERITXELL HUCH: Webminar ISSCR, Skokie, Illinois, USA
TONY KOUZARIDES: Michael Lund Nielsen, University of Copenhagen,
Denmark
TONY KOUZARIDES: Institute Pasteur, Paris, France
ERIC MISKA: BIG, Norwegian Biochemical Society, Oslo, Norway
ERIC MISKA: RNAi 2014, Oxford, UK
ERIC MISKA: Annual Lausanne Genomic Days, Lausanne, France
JON PINES: 2nd UK-Korea Mitosis Workshop, Seoul, South Korea
MARCH
JULIE AHRINGER: Keystone Chromatin Meeting, Oberstdorf, Germany
ANDREA BRAND: 15th Servier-IGIS Symposium Neural Orchestration of
Metabolism and Islet Function, St Jean Cap Ferrat, France
NICK BROWN: Plenary speaker, International Meeting of the German Society
for Cell Biology, Regensburg, Germany
JOHN GURDON: College de France, Paris, France
JOHN GURDON: Bishops Stortford College, UK
JOHN GURDON: Society of Toxicology, Phoenix, Arizona, USA
MERITXELL HUCH: ILTS Workshop, Rotterdam, Netherlands
STEVE JACKSON: Abcam Meeting, St. Kitts, Saint Kitts And Nevis
STEVE JACKSON: BSCB/BSDB Annual Spring Meeting, Coventry, UK
STEVE JACKSON: Cancer Science Symposium, Cambridge, UK
TONY KOUZARIDES: Keystone Symposia, Oberstdorf, Germany
ERIC MISKA: Tel Aviv Universit, Tel Aviv, Israel
EUGENIA PIDDINI: BSCB/BSDB Annual Spring Meeting, Coventry, UK
EMMA RAWLINS: BSCB/BSDB Annual Spring Meeting, Coventry, UK
DANIEL ST JOHNSTON: BSCB/BSDB Annual Spring Meeting, Coventry, UK
AZIM SURANI: Keystone Symposia: Chromatin mechanisms and cell physiology,
Oberstdorf, Germany
PHILLIP ZEGERMAN: St. Olaves Grammar School, Orpington, UK
PHILLIP ZEGERMAN: University of Dundee, Dundee, UK
APRIL
JULIE AHRINGER: Wellcome Trust Conference Chromatin: From nucleosomes
to chromosomes, Hinxton, UK
ANDREA BRAND: College de France, Paris, France
NICK BROWN: Seminar, Dept of Pharmacology, Yale School of Medicine, New
Haven, Connecticut, USA
NICK BROWN: Seminar, Department of Cell and Systems Biology, Toronto,
Canada
JOHN GURDON: Keystone Symposium, Squaw Creek, California, USA
JOHN GURDON: Penn’s Institute for Regenerative Medicine, Pennsylvania, USA
JOHN GURDON: John Ray Society, Cambridge, UK
STEVEN MOORE: The future of research and clinical applications in
neuroscience, Genoa, Italy
JON PINES: Ramon Areces Foundation, Santander, Spain
JON PINES: Brunel University, Brunel, UK
JON PINES: Newcastle University, Newcastle, UK
JON PINES: DKFZ-ZMBH Alliance Meeting, Heidelberg, Germany
EMMA RAWLINS: ERS Research Seminar ‘The many faces of stem cells in
respiratory diseases’, Barcelona, Spain
EMMA RAWLINS: International Society for Cellular Therapy Meeting: Satellite
session on tracheal engineering, Paris, France
AZIM SURANI: DKFZ-ZMBH, Heidelberg, Germany
EMMANUELLE VIRE: Institute Cochin, Paris, France
MAY
JULIE AHRINGER: Keynote Speaker: European C elegans Meeting, Berlin,
Germany
JULIE AHRINGER: University of Edinburgh, UK
THE GURDON INSTITUTE
55
TALKS BY INSTITUTE RESEARCHERS
TALKS BY INSTITUTE RESEARCHERS
ANDREA BRAND: European Molecular Biology Laboratory (EMBL),
Heidelberg, Germany
ANDREA BRAND: The Stem Cell Niche - Development and Disease
Conference, Copenhagen, Denmark
NICK BROWN: UK Drosophila Developmental Cell Biology Workshop 2014,
Edzell, UK
JOHN GURDON: Hay Festival, Hay-on Wye, UK
JOHN GURDON: School of Biological Sciences Postdoc Retreat, Cambridge,
UK
MERITXELL HUCH: Pint of Science Festival, Cambridge, UK
MERITXELL HUCH: Stem Cell Research mini conference, Imperial College,
London, UK
STEVE JACKSON: Abcam Meeting Mechanisms of Recombinations: 50th
Anniversary Meeting of the Holliday Model, Alicante, Spain
BENJAMIN KLAPHOLZ: 5th French Cell Adhesion Club Symposium, Marseille,
France
ERIC MISKA: Translational Control of Brain Function & Epigenomic Engineering,
London, UK
ERIC MISKA: Wellcome Trust Translation conference, London, UK
ERIC MISKA: MPI Tuebingen, Germany
JON PINES: Dundee University, UK
BEN SIMONS: The Stem Cell Niche - Development and Disease, Copenhagen,
Denmark
DANIEL ST JOHNSTON: ESF/EMBO Conference on Cell Polarity and
Membrane Trafficking, Pultusk, Poland
AZIM SURANI: Reprogramming during development, induced pluripotency and
disease, Paris, France
JUNE
ANDREA BRAND: Lawrence Berkeley National Laboratory, Berkeley, California,
USA
ANDREA BRAND: EMBO International Workshop on Molecular and
Developmental Biology of Drosophila, Crete, Greece
NICK BROWN: EMBO International Workshop on Molecular and
Developmental Biology of Drosophila, Crete, Greece
NICK BROWN: Seminar, Biomedical Research Foundation, Athens, Greece
JOHN GURDON: Rockefeller Symposium + Honorary degree, New York, UK
MERITXELL HUCH: BASL - British Association for the Study of the Liver, Seal
Hayne, Devon, UK
ERIC MISKA: ETH Zurich Seminar, Switzerland
EUGENIA PIDDINI: EMBO, Crete, Greece
BEN SIMONS: Theory of Living Matter Group, Cambridge, UK
AZIM SURANI: ISSCR Annual Meeting, Vancouver, Canada
56
THE GURDON INSTITUTE
JULY
JULIE AHRINGER: University of Washington, Seattle, USA
JOHN GURDON: Chromatin Club/Abcam, London, UK
JOHN GURDON: University of Cambridge, Cambridge, UK
MERITXELL HUCH: FASEB liver meeting, Keystone, Colorado, USA
STEVE JACKSON: Association of Radiation Research Meeting, Sussex, UK
ERIC MISKA: RNA Habitats, Salzburg, Austria
STEVEN MOORE: 2nd Stem BANCC workshop on human cerebral cortex
differentiation, Cambridge, UK
BEN SIMONS: Theory of Living Matter Group, Cambridge, UK
JAMES SMITH: 2nd Stem BANCC workshop on human cerebral cortex
differentiation, Cambridge, UK
DANIEL ST JOHNSTON: Wellcome Fellows Meeting, London, UK
AZIM SURANI: Cell Symposia: Transcriptional regulation in development,
Chicago, Illinois, USA
AUGUST
ANDREA BRAND: Santa Cruz Developmental Biology Meeting, California, USA
YARON GALANTY: Benzon Symposium No.60 Nuclear Regulation by
Ubiquitin, Copenhagen, Denmark
JENNY GALLOP: Xenopus Meeting, California, USA
JOHN GURDON: 15th Xenopus Meeting, Asiloma, California, USA
TONY KOUZARIDES: EMBL, Heidelberg, Germany
JON PINES: 60th Benzon Symposium, Copenhagen, Denmark
AZIM SURANI: Gordon Conference, Mammalian reproduction, translating basic
science into clinical applications, New London, New Hampshire, USA
EVA-MARIA WEICK: CSHL Conference - Regulatory & non-coding RNAs, Long
Island, New York, USA
PHILIP ZEGERMAN: Xenopus Meeting, Pacific Grove, California, UK
SEPTEMBER
JULIE AHRINGER: Chromatin and Epigenetics Conference, Cold Spring Harbor,
USA
PAULO AMARAL: Uppsala University, Uppsala, Sweden
PAULO AMARAL: Stockholm RNA society, Stockholm, Sweden
NICK BROWN: Get Connected 4: Cellular microenvironment, Manchester, UK
NICK BROWN: Shaping Cells and Organisms Symposium, Cologne, Germany
TATYANA DIAS: ARUK Cambridge Network Committee, Cambridge, UK
JENNY GALLOP: EMBO Advanced Course, Heidelberg, Germany
JOHN GURDON: 1st Congress of the Polish Biochemistry, Cell Biology, Biology,
Biophysics & Bioinformatics, BIO 2014, Warsaw, Poland
JOHN GURDON: Victor Chang Cardiac Research Institute and Garvan Institute
of Medical Research, Sydney, Australia
JOHN GURDON: Combio 2014, Canberra, Australia
JOHN GURDON: Monash University, Melbourne, Australia
MERITXELL HUCH: The European Cancer Stem Cell Research Institute
Symposium, Cardiff, UK
STEVE JACKSON: Cancer Pharmacogenomics and Targeted Therapies
Conference, Hinxton, UK
PETER KIRWAN: From Cells to Tissue: Stem Cells, Tissue Repair and Tissue
Engineering for Diabetes, Eye Disease and Neurodengerative Diseases, Dublin,
Republic of Ireland
TONY KOUZARIDES: Cold Spring Harbor Laboratory (CHSL), Cold Spring
Harbor, New York, USA
ERIC MISKA: Fondazione Cariplo, Brescia, Italy
EUGENIA PIDDINI: JEDI Meeting, Marseille, France
DANIEL ST JOHNSTON: Cologne’s International Graduate Programme
in Biology and Maria Leptin’s 60th Birthday, Institute for Genetics, Cologne,
Germany
BEN SIMONS: Japan Neuroscience Society, Yokohama, Japan
BEN SIMONS: Centre for Development Biology, Kobe, Japan
BEN SIMONS: National Institute of Basic Biology, Okazaki, Japan
JULIE WATSON: European Respiratory Society, Munich, Germany
OCTOBER
JULIE AHRINGER: EMBO Conference, Cargese, Corsica, France
ALPER AKAY: Herchel Smith Symposiusm, Cassis, France
ANDREW BANNISTER: UCL, London, UK
ANDREA BRAND: Neurofly 2014, Hersonissos, Crete, Greece
ANDREA BRAND: Developmental Biology Course, Insitut Curie, Paris, France
JOHN GURDON: Abcam, Chromatin & Epigenetics: from Omics to Stem Cells,
Strasbourg, France
JOHN GURDON: Harveian Oration, London, UK
JOHN GURDON: Stem Cells meeting, Provence, France
MERITXELL HUCH: Next generation cell culture models workshop, Hinxton,
UK
TONY KOUZARIDES: BDEBATE PEBC-IDIBELL, Barcelona, Spain
TONY KOUZARIDES: Abcam, Strasbourg, France
TONY KOUZARIDES: Wellcome Trust, Hinxton, Cambridge, UK
ERIC MISKA: MPI Seminar, Cologne, Germany
ERIC MISKA: IMB Conference: Nuclear RNA in Gene Regulating & Chromatin
Structure, Mainz, Germany
ERIC MISKA: Living Matter Talk at Rockefeller University, New York City, New
York, USA
EUGENIA PIDDINI: EMBO, Stockholm, Sweden
JON PINES: CR UK Clare Hall Laboratories, London, UK
AZIM SURANI: Foundation Les Treilles Meeting, Nice, France
AZIM SURANI: State Key Laboratory of Reproductive Biology (SKLRB), Beijing,
China
MÉLANIE TANGUY: Herchel Smith Symposiusm, Cassis, France
NOVEMBER
ALPER AKAY: Churchill College, Cambridge, UK
YARON GALANTY: IRIBHM, Brussels, Belgium
JOHN GURDON: ISD and BSDB, London, UK
JOHN GURDON: Wellcome Trust Researcher Meeting: Development, Ageing
and Regenerative Medicine, Ware, UK
MERITXELL HUCH: Erasmus Liver Day, Rotterdam, Netherlands
MERITXELL HUCH: British Society for Developmental Biology, London, UK
STEVE JACKSON: Molecular mechanisms of cellular surveillance and damage
responses, Heidelberg, Germany
TONY KOUZARIDES: University of Miami, Miller School of Medicine, Miami,
Florida, USA
ERIC MISKA: MCB Seminar, Boston, Massachusetts, USA
ERIC MISKA: MRC Leicester Unit Seminar Series, Leicester, UK
JON PINES: Southampton University, Southampton, UK
JON PINES: EPFL, Lausanne, Switzerland
EMMA RAWLINS: National Institute for Medical Research, London, UK
DANIEL ST JOHNSTON: Distinguished Lecture Max Planck, Martinsried,
Germany
BEN SIMONS: Wellcome Trust Researcher Meeting on Pathogens, Immunity and
Cell Biology , Hertfordshire, UK
AZIM SURANI: International Society for Stem Cells (ISSCR): Global controls in
Stem Cells, Singapore, Singapore
AZIM SURANI: Wellcome Trust Researcher Meeting, Ware, UK
PHILIP ZEGERMAN: 3Rs Meeting, Tokyo, Japan
DECEMBER
ANDREA BRAND: UK/Japan Workshop University College London, UK
JENNY GALLOP: University of Kent, Canterbury, UK
JOHN GURDON: MRC, London, UK
JOHN GURDON: CSAR Churchill College, Cambridge, UK
TONY KOUZARIDES: Max Planck Institute of Immunobiology and Epigenetics,
Freiburg, Germany
TONY KOUZARIDES: RIKEN and UCL, London, UK
ERIC MISKA: Translational Science Update, London, UK
ERIC MISKA: MPI Epigenetics Meeting, Freiburg, Germany
DMITRY NASHCHEKIN: Sainsbury Laboratory, Research in Genetics Day,
Cambridge, UK
EMMA RAWLINS: DanStem, Copenhagen, Denmark
THE GURDON INSTITUTE
57
GURDON INSTITUTE SEMINARS / STAFF AFFILIATIONS
LENT TERM
1) 21 January, Cédric Blanpain, Interdisciplinary Research Institute
(IRIBHM), Université Libre de Bruxelles, Belgium:
“Stem cell dynamics during development, homeostasis and cancer”
2) 28 January, ( The Anne McLaren Lecture) Maria Leptin, EMBO,
Heidelberg, Germany:
“Cell shape and morphogenesis: sub cellular and supra-cellular
mechanisms”
3) 11 February, Pierre Gönczy, EPFL, Lausanne, Switzerland:
“Mechanisms of centriole assembly”
4) 11 March, Detlef Weigel, Max Planck Institute for Developmental
Biology, Tübingen, Germany:
“Origins and consequences of (epi)genetic variation in Arabidopsis thaliana
and its relatives”
5) 25 March, Claude Desplan, Department of Biology, New York
University, USA:
“Generating neuronal diversity: stochastic or deterministic choices”
MICHAELMAS TERM
6) 14 October, Alexander Aulehla, European Molecular Biology,
Laboratory (EMBL), Heidelberg, Germany:
“Self-organization of genetic oscillators during mouse mesoderm
development”
7) 21 October, Péter Lénárt, European Molecular Biology, Laboratory
(EMBL), Heidelberg, Germany:
“Microtubules need actin’s help in large oocytes, to collect chromosomes
and to break the nuclear envelope”
8) 25 November, W. James Nelson, Department of Biology, Stanford
University, USA:
“Evolution of cell-cell adhesion, and new insights into mechanisms in
animals ”
9) 2 December, Iain Hagan, Cancer Research UK, Manchester Institute,
University of Manchester:
“A Protein phosphatase 1/protein phosphatase 2A relay controls mitotic
progression”
58
THE GURDON INSTITUTE
OTHER INFORMATION
STAFF AFFILIATIONS
JULIE AHRINGER is a member of the Scientific Advisory Boards of the
MRC Clinical Sciences Centre and Wormbase.
ANDREA BRAND is a member of Council of The Royal Society, member
of the Wellcome Trust/Royal Society Sir Henry Dale Fellowship Committee
and a Founding Board Member of The Rosalind Franklin Society (USA). She
is on the Board of Directors of the Cambridge Science Centre and is a
Patron of the Cambridge Science Festival.
JOHN GURDON is an honorary member of the British and American
Anatomical Societies, a board member of Diagnostics for the Real World
and a member of the Faculty of 1,000.
STEVE JACKSON is an Associate Faculty Member of the Wellcome Trust
Sanger Institute and is founding Scientist and Chief Scientific Officer of
MISSION Therapeutics Ltd. He is a member of the Scientific Advisory
Boards for the MRC Protein Phosphorylation and Ubiquitylation Unit
(Dundee), the Beatson Institute (Glasgow), the MRC Toxicology Unit
(Leicester), the Radiation Oncology and Biology Institute (Oxford), the
MRC Clinical Sciences Centre (London) and the Netherlands Cancer
Institute (Amsterdam). He is on the Steering Committee for the Cambridge
Cancer Centre, and is a member of the CRUK Science Committee.
MAGDALENA ZERNICKA-GOETZ is a Fellow of The Academy of
Medical Sciences and EMBO
HONOURS AND AWARDS
JULIE AHRINGER – Professorship, University of Cambridge
ANDREA BRAND – Honorary Fellow, Brasenose College, University of
Oxford.
NICK BROWN – Professorship, University of Cambridge
JOHN GURDON – Honorary Degree, Rockefeller University
MERITXELL HUCH – Wellcome-Beit Prize, NC3Rs International Prize
STEVE JACKSON – Fellowship of Imperial College Faculty of Medicine
STEVE JACKSON AND DELPHINE LARRIEU – Winners of GSK
Discovery Fast Track Challenge
BEN SIMONS – Institute of Physics Rosalind Franklin Medal
PHILIP ZEGERMAN – EMBO Young Investigator
AZIM SURANI – Jawaharlal Nehru Science Fellowship (JNSF), ISSCR
McEwen Award for Innovation
AZIM SURANI – Cell; Nature Communications; Cell Stem Cell; BMC
Epigenetics and Chromatin; Epigenome; Epigenomics; Epigenetic Regulators;
Regenerative Medicine; Differentiation; Stem Cell Research and Therapy;
Faculty of 1,000; Cell Research; Cell Discovery
MAGDALENA ZERNICKA-GOETZ – Development; Differentation;
Developmental Dynamics; Cells
INTERNATIONAL SCIENTIFIC ADVISORY BOARD
DR GENEVIEVE ALMOUZNI, Institut Curie, Paris, France
DR ADRIAN BIRD, Wellcome Trust Centre for Cell Biology, University of
Edinburgh
DR STEVE COHEN, Institute of Molecular and Cell Biology, Singapore
DR JUDITH KIMBLE, Department of Biochemistry, University of
Wisconsin-Madison, USA
DR ELISABETH KNUST, Max Planck Institute of Molecular Cell Biology
and Genetics, Dresden, Germany
DR ROBB KRUMLAUF (Chairman), Stowers Institute for Medical
Research, Kansas City, USA
TONY KOUZARIDES is a member of the Cancer Research UK Science
and Strategy Advisory Group, part of the Scientific Advisory Board for
the Centre for Genomic Research (Spain), the Institute of Molecular
Biology (Crete) and the Centre for Epigenetics and Biology (Spain). He
is the founder and director of a Spanish cancer charity Vencer el Cancer
(Conquer Cancer) and a founder of Chroma Therapeutics and Abcam Plc.
THE GURDON INSTITUTE – Athena SWAN Bronze Award
DR MATTHIAS PETER, ETH Zurich
EDITORIAL BOARDS OF JOURNALS
CHAIRMAN OF THE MANAGEMENT COMMITTEE
JULIE AHRINGER – eLife; PLoS Biology; PLoS Genetics; Molecular Systems
Biology
ERIC MISKA is an Honorary Faculty Member of the Wellcome Trust
Sanger Institute
ANDREA BRAND – eLife; Neural Development; Fly; Biology Image Library
PROFESSOR DUNCAN MASKELL, Head of the School of the Biological
Sciences and Marks & Spencer Professor of Farm Animal Health, Food
Science & Food Safety, Department of Veterinary Medicine
JONATHON PINES is a member of the Scientific Advisory Board for
the UMR144, Institut Curie, Paris, France, and the Evaluation Panel of the
Institute of Biochemistry, ETH, Zurich.
DANIEL ST JOHNSTON is a Director of the Wellcome Trust Four-Year
PhD programme in Developmental Biology at the University of Cambridge,
a non-executive Director of the Company of Biologists, and acting Editor of
Disease Models and Mechanisms.
AZIM SURANI is a member of the Steering Committee of the Wellcome
Trust-Medical Research Council Cambridge Stem Cell Institute, and theme
leader of the Pluripotency Programme. He is also a member of the
Cambridge-India Partnership Advisory Group, founder and Chief Scientific
Advisor for CellCentric Ltd, a member of the Steering Committee for the
UK Stem Cell Bank, and a member of the Royal Society Hooke Committee.
He is also a member of the Scientific Advisory Board of the Institute of
Stem Cell Biology and Regenerative Medicine, Bangalore, India.
JOHN GURDON – Current Biology; Development; Faculty of 1,000;
Growth and Differentiation; International Journal of Developmental Biology;
Proceedings of the National Academy of Sciences of the USA
MERITXELL HUCH – Cogent Biology
STEVE JACKSON – Aging; Biomolecules; Carcinogenesis; DNA Repair ;
EMBO Journal ; Genes and Development; The Scientist; Science Signaling
(Board of Reviewing Editors)
RICK LIVESEY – BMC Developmental Biology; Molecular Autism
EMMA RAWLINS – Pediatric Research
JON PINES – EMBO Journal; EMBO Reports; Open Biology; eLife
BEN SIMONS – Development
DANIEL ST JOHNSTON – Development; Faculty of 1,000
THE GURDON INSTITUTE
59
DESTINATIONS OF LEAVERS DURING 2014
GROUP LEADERS
MAGDALENA ZERNICKA-GOETZ: Group Leader, Department of Physiology,
Development & Neuroscience, University of Cambridge
POSTDOCTORAL RESEARCHERS
HOZEFA AMIJEE: Project Manager, Total Scientific, Cambridge, UK (Livesey Lab)
FRANCESCO ANTONICA: Postdoctoral Researcher, Department of
Physiology, Development & Neuroscience, University of Cambridge (ZernickaGoetz Lab)
ALYSON ASHE: Group Leader, University of Sydney, Australia (Leaver 2013)
IVAN BEDZHOV: Postdoctoral Researcher, Department of Physiology,
Development & Neuroscience, University of Cambridge (Zernicka-Goetz Lab)
MONIKA BIALECKA: Postdoctoral Researcher, Department of Physiology,
Development & Neuroscience, University of Cambridge (Zernicka-Goetz Lab)
MIKE CHESNEY: Senior Scientist, Illumina, Cambridge UK (Ahringer Lab)
MARIA CHRISTOPHOROU: Group Leader, MRC Human Genetics Unit, MRC
IGMM, University of Edinburgh, UK (Kouzarides Lab)
PHILIPPE COLLIN: Senior Scientist, Horizon Discovery, Waterbeach,
Cambridgeshire UK (Pines Lab)
SVEN HUELSMANN: Lecturer and Study Coordinator at the University of
Tübingen (Brown Lab)
AGNIESZKA JEDRUSIK: Postdoctoral Researcher, Department of Physiology,
Development & Neuroscience, University of Cambridge (Zernicka-Goetz Lab)
OLEG KOVALEVSKIY: Career Development Fellow, MRC-LMB, Cambridge, UK
(Zegerman Lab)
TAKAHIRO MATSUSAKA: Senior Scientist, Horizon Discovery, Waterbeach,
Cambridgeshire UK (Pines Lab)
GAELLE RECHER: Postdoctoral Researcher, Department of Physiology,
Development & Neuroscience, University of Cambridge (Zernicka-Goetz Lab)
JOSANA RODRIGUEZ: Group Leader, Institute for Cell and Molecular
Biosciences, Medical School, Newcastle University, UK (Ahringer Lab)
PETER SARKIES: Group Leader, Epigenetics Division at the MRC Clinical
Sciences Centre, Imperial College, London (Miska Lab)
MARTA SHAHBAZI ALONSO: Postdoctoral Researcher, Department of
Physiology, Development & Neuroscience, University of Cambridge (ZernickaGoetz Lab)
TONY SOUTHALL: Lecturer, Department of Life Sciences, Imperial College,
London, UK (Brand Lab)
PETER TESSARZ: Group Leader, Max Planck Institute for Biology of Ageing,
Cologne (Kouzarides Lab)
JON TRAVERS: Postdoctoral Fellow, Medimmune, Cambridge, UK (Jackson Lab)
EMMANUELLE VIRÉ: Senior Investigator, MRC Prion Unit, London UK
(Kouzarides Lab)
ASTRID WALRANT: Maitre de Conferences (Lecturer), Universite Pierre et
Marie Curie (UPMC), Chemistry Faculty, Paris, France (Gallop Lab)
60
THE GURDON INSTITUTE
RESEARCH ASSISTANTS/TECHNICIANS
AMY CORDING: Research Assistant, Department of Biology, University of York,
UK (Miska Lab)
ANDY COX: Research Assistant, Department of Physiology, Development &
Neuroscience, University of Cambridge (Zernicka-Goetz Lab)
DJEM KISSIOV: PhD Student, University of California, Berkeley, USA (Ahringer
Lab)
AMELIA McGLADE: Research Assistant, Garvan Institute, Sydney (Livesey Lab)
CHANDRIKA RAO: PhD Student, University of Edinburgh, UK (Rawlins Lab)
Research
PhD/MPhil STUDENTS
DARYA AUSIANNIKAVA: Postdoctoral Associate, Dr Thorsten Allers’ lab,
University of Nottingham, UK (Ahringer Lab)
JESSICA BROWN: SpR Medical Oncology, Addenbrooke’s Hospital NHS Trust,
Cambridge UK (Jackson Lab)
LEAH BURY: PhD, Department of Physiology, Development & Neuroscience,
University of Cambridge (Zernicka-Goetz Lab)
ESTEBAN CONTRERAS SEPULVEDA: Postdoctoral Researcher, University of
Chile, Santiago, Chile (Brand Lab)
KATRINA GOLD: Postdoctoral Researcher, UCSF, San Francisco, USA (Brand
Lab)
MOHAMMED GOOLAM: PhD Student, Department of Physiology,
Development & Neuroscience, University of Cambridge (Zernicka-Goetz Lab)
SARAH GRAHAM: PhD Student, Department of Physiology, Development &
Neuroscience, University of Cambridge (Zernicka-Goetz Lab)
ANNABEL GRIFFITHS: Travelling, Volunteer in Cambodia, Science Consultant
on return to UK (Brown Lab)
TIMM HAACK: Postdoctoral Researcher, Salim Seyfried’s Lab, Hannover Medical
School, Germany (St Johnston Lab)
RICHARD HALLEY-STOTT: Medical Degree, University of Cape Town, South
Africa (Gurdon Lab)
MORITZ HERRMANN: Pharmaceuticals Analyst, GlobalData, London (Ahringer
Lab)
PETER KIRWAN: Senior Postdoctoral Scientist, Talisman Therapeutics,
Cambridge UK (Livesey Lab)
CHUEN YAN LEUNG: PhD Student, Department of Physiology, Development
& Neuroscience, University of Cambridge (Zernicka-Goetz Lab)
AGATA LICHAWSKA: Postdoctoral position, Krakow, Poland (Pines Lab)
KENNETH MURFITT: Senior Scientist, Novartis, Maidstone, Kent (Miska Lab)
MARYNA PANAMAROVA: PhD Student, Department of Physiology,
Development & Neuroscience, University of Cambridge (Zernicka-Goetz Lab)
MARTA TEPEREK-TKACZ: Business Development Consultant, Oxbridge
Biotech Roundtable, London, UK (Gurdon Lab)
STAN WANG: Completing PhD in USA before returning to Medical School in
Columbia (Gurdon Lab)
BEATA WYSPIANSKA: Senior Scientist, GSK, Stevenage, UK (Kouzarides Lab)
The Institute on retreat, October 2014
(photo by James Smith, Livesey Group)
ACKNOWLEDGEMENTS
Prospectus produced in the Wellcome Trust/Cancer Research UK Gurdon Institute.
Edited by Ann Cartwright, production by Alastair Downie
Group photographs by James Smith, Livesey Group.
Print management by H2 Associates, Cambridge
Front cover: False-coloured DIC images of HeLa cells at different stages of mitosis, surrounding the central image
of the double gene knockout celebration cake. The cake itself is also a ‘double knockout’ of sources of allergens;
gluten and almond. Time lapse images captured on Deltavision microscope
(Keiko Yata and Oxana Nashchekina, Pines Group)
Back cover: The Institute tea party, in recognition of one million kWh saved since launching our energy saving
campaign in March 2012
(photo by Paul Holland, Office of External Affairs and Communications, University of Cambridge)
Wellcome Trust/Cancer Research UK Gurdon Institute
The Henry Wellcome Building of Cancer and Developmental Biology
University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, United Kingdom
Telephone: +44 (0)1223 334088
Fax: +44 (0)1223 334089
http://www.gurdon.cam.ac.uk
e-mail: [email protected]