Cambridge Instit ute for Therapeuttic Immunology

& Infectious Disease

Institute Directory CITIID CITIID

www.citiid.cam.ac.uk A PERFECT L O C A T I O N

The Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID) was established by the Department of Medicine to support both fundamental and translationaal research on human disease.

CITIID houses up to 250 scientists working in research groups focussed on understanding the pathogenesis and improving the management of immune‐related disorders, as well as transforming our understanding of how infectious agents interact with humans. Later in this directory you’ll find out more about these studies.

There is also a strong focus on health issuess of global importance, and on antimicrobial resistance. In particular, we work closely with key overseaas partners including universities, agencies and industry to ensure that we can increase our global impaact in a coordinated programme.

CITIID brrings together geographically dispersed groups working on immunity and infection across the Cambridge Biomedical Campus, the largest Biomedical campus in Europe. By encouraging research at the clinical interface, for example in the Cambridge Clinical Research Centre and with industry, CITIID also faciilitates the translation of scientific disscoveries into clinical benefits.

Its location on the Cambridge Biomedical Campus places CITIID at the centrre of the Cambridge Cluster where it benefits from the closeness of several other major health‐related organisations such as the Royal Papworth Hospital, MRC‐Laboratory of Molecular Biology, CRUK Cambridge Institute and Astra Zeneca’s global research and development headquarters.

CITIID transforms immunity and infection research in Cambridge by providiing researchers with advanced facilities that enable them to optimise their work on understandding human immune, inflammatory and infectious diseases. By bringing together its clinical capabilities and key industry partners, CITIID is also well‐placed to drive therapeutic breakthroughs, improove patient outcomes and advance population health both in the UK and abroad.

1 O U R S C I E N C E

Designed to Lead Developments in Autoimmunity

CITIID is already driving therapeutic breakthroughs in immune‐related diseases such as inflammatory bowel disease, type‐1 diabetes, vasculitis annd systemic lupus erythematosus. The incidence of such disease is rising worldwide at a truly alarming rate and collectively they represent a major global public health issue. We study what drives these diseases – how they develop and evolve, and how they cause harm to humans – and use this understanding to develop new therappeutic strategies that can be translated quickly and safely to patiennts worldwide.

A key strength of CITIID is that immune‐mediated disease is studied alongside innfectious disease. The human immune system has evolved to defend us against the dangerous microorganisms that cause disease; autoimmunity and inflammatory diseases are caused when this defence mechanism malfunctions. Crucially, a dysfunctional immune system uses the same molecular pathways and mechanisms as a healthy immune system uses to fight infection, which means vital insights emerge from tackling infectious disease and autoimmunity together.

Over the last four years scientists at CITIID have established a whole genome sequencing‐based programme to investigate undiagnosed primary immunodeficiency. This has provided over three hundred diagnostic reports, uncovered many genes associated with primary immunodeficiency, and allowed the genetic variation in the development of these diseases to be explored.

2 O U R S C I E N C E

Designed to Combat Multi‐Drug Resistance

Antimicrobial resistance is one of the biggesst threats to global health and internaational food security. Scientists at CITIID develop novel approaches to overcome this challenge.

Considering how human genetics responds to infection is a key component in understanding the role of the microbiome in health and disease. By using cutting‐edge scientific metthods to monitor the spread oof antimicrobial resistance, our scieentists can even use historical data to understand the spread of multi‐drug resistance long after an outbreak. Such studies have an eenormous impact on our ability to manage the continued spread of a disease and allow us to anticipate future threats.

A Centre for Global Answers

Infectious disease, antimicrobial resistance and autoimmunity are three of hummanity’s deadliest foes and are major global challenges. Cambridge is one of the world’s leading research universities, working across disciplines with international partners to find solutions to manage and eradicate these threats.

CITIID is at the centre of efforts to understand the spread of disease. We have researchers working in countries across the globe, meaning we have a very real understanding of the iimpact diseases have on patients. Many of our projects are in third world countries where socio‐ecoonomic factors make the impacts of these diseases even greater.

3

3 5

CITIID Lead CI CITIID Biolog Molec footpri Cambri Biome T

y u d n i dge HE . s lar ing ical

t

TI primarily

in

Immuno

MRC Stem

Centre

Adde Dis

C L L c n c n I

I , e e logy located

ABORAT brookes’s overy ll where

Institute D

Unit

in we

th

a O a O H H

t t

a at and he ospital nd RY share

is

newly

embed the OF

Inn

stat

to

Mi M

c o o e e l f l O f O d d onstructed ner acilitate

ed vation of LECULA

within Therap the

AD

ar cli

t t R e t R e n n a DENBRO he

utics ical nd

research BIOLOG

MRC

purpos

transl Instit

La

O b O l b a a u u e a Y e a Y oratory tion, te. boratories ‐ KE’S

built

CITIID

as

HO Jeffr

of w

a S e M S e M

e e lso ll with PITAL y olecular

as

Cheah 4 has

the the

a

The Jeffrey Cheah Biomedical Cenntre

A New Site of Research Excellence

The Jeffrey Cheah Biomedical Centre was established in 2016 with funding ffrom the Higher Education Funding Council for England (HEFCE) UK Research Partnership Infrastructure Fund (UKRPIF), the Univversity of Cambridge Capital Fund, and philanthropic donations. The majority of our researchers are located in this state‐of‐the‐ art facility on the Cambridge Biomedical Campus.

FROM CONCEPT IN 2016

TO COMPLETION IN 2019

5 6 CITIID CITIID

OUR SCIENTISTS

Steve Baker Studies the microbiology, genetics, and treatment of enteric infections in 10 developing countries.

Allan Bradley Is strongly focused on using genetics to discover genes involved in various biological 11 systems.

Elisabeth Busch‐Nentwich Explores molecular phenotypes through the transcript sequencing of whole vertebrate embryos and tissues. 1 2 Sarah Caddy Studies how rotavirus is targeted by antibodies, focusing on vaccination and maternal 13 immunity.

Menna Clatworthy Is interested in the humoral immunity in humans, how this goes wrong in disease and 1 4 how this can be manipulated therapeutically using experimental medicine studies.

Gordon Dougan Undertakes research focused on the genetic analysis of host/pathogen interactions 15 during infection, particularly those involving enteric bacteria.

Ravindra Gupta Focuses on drug‐pathogen and host‐pathogen interactions to inform successful global HIV 1 6 control/cure strategies.

Christoph Hess Studies the molecular basis of how cellular metabolism is regulated, and itself 1 7 regulates, immune‐function in healthy and disease states.

David Jayne Conducts clinical trials and associated biomarker work in primary systemic vasculitis and 1 8 systemic lupus erythematosus.

Nicole Kaneider Investigates GPR35’s function, and in particular the communication of immune cells 19 expressing this receptor, and their environment.

Arthur Kaser Investigates the mechanisms that underlie inflammation at mucosal surfaces. 2 0

James Lee Studies the genetic associations in immune‐mediated disease by combining genetics, 21 , molecular biology and immunology‐based approaches.

Paul Lehner Works on functional genetic and proteomic technologies to identify novel genes and 2 2 therapeutic approaches in virus:host interaction.

6

Florian Marks Operates a network of field sites capable of conducting multi‐center epidemiological field 23 studies for basic research and clinical trials

Nick Matheson Studies host‐virus interactions to provide insights into viral pathogenesis, cell biology, 24 and novel therapeutic approaches.

Eoin McKinney Studies aberrant immune response to indicate novel tests and/or interventions that can 25 help clinicians treating immune pathology.

Yorgo Modis Our overarching goal is to gain a mechanistic understanding at the molecular level of how 26 the cell detects cytosolic viral RNA and how it silences viral gene expression.

Ankur Mutreja Uses advanced bacterial genomics and metagenomics to better understand the 27 evolution and spread of pathogens.

James Nathan Gains novel insights into oxygen and metabolite sensing pathways, providing potential 28 new therapeutic targets for inflammatory disease and cancers.

Virginia Pedicord Seeks mechanistic insights into the influence of commensal microbes on local and 29 systemic immune responses.

Lalita Ramakrishnan Researches the pathogenesis of tuberculosis, and tries to understand both host and 30 pathogen strategies that can interact to result in clearance or disease.

John Sinclair Studies the molecular basis of viral latency and reactivation and how the host immune 31 response combats viral disease.

Ken Smith Combines genomics, immunology and clinical medicine to analyse the regulatory 32 mechanisms underpinning immune‐mediated disease and develop diagnostic tools.

Alex Taylor Is evolving and engineering synthetic genetic polymers, xeno nucleic acids (XNA), as novel 33 tools for reprogramming biology and precision medicine.

Chris Wallace Develops statistical methods and uses integrative statistical analysis of genomic data to 34 improve diagnosis and treatment of immune mediated diseases.

Mark Wills Is interested in the immunobiology of HCMV in lytic and during latent infection, and how 35 the pathogen persists for the lifetime of its human host. 7 OUR IMPACT

Much of the research undertaken in CITIID has a focus on real‐ challenges. As such, it has a significant positive impact on the people’s lives.

The stories below describe some examples of the impact that our research has on a daily basis.

From genetic analysis to personalised medicine in inflammatory bowel disease. Our research found a transcriptional signature that is detectable within peripheral 36 blood and correlates with long term clinical outcome in IBD.

New treatments for Tuberculosis. Our researchers revealed that the actively‐growing bacteria induce bacterial efflux 38 pumps that could be therapeutic targets using existing drugs.

The immunity of inflammatory bowel disease is exposed. Our researchers developed experimental methods for examining the 40 environmental stresses and microbes that could be responsible for triggering IBD.

Improving sanitation in rural Africa. Our researchers are providing evidence that simple behavioural changes can improve 42 the health of rural communities in the third world.

Cholera: a global challenge. Our researchers are using information locked within the genomes of historic cholera 44 samples to predict the spread of past cholera pandemics throughout Africa.

8 CORE FACILITIES

CITIID offers a range of core facilties to it’s researchers and the wider scientific community. Some of these are listed below, and our facility contacts are more than happy to discuss your specific needs.

NIHR Cambridge BRC Phenotyping Hub Equipped with state‐of the art equipment including high speed cell sorters, bench top 46 analysers, microscopes and high content/high throughput equipment.

Imaging Offering a variety of instruments and analysis techniques. 47

Computing High‐performance computing (HPC) cluster and 200TB network storage that is 47 configured similar to the HPC

Immuno‐metabolism Offering cutting‐edge, high‐resolution liquid chromatography mass spectrometry (LC‐MS) 48

Bio‐Safety Level 3 CL‐3 containment facilities for working with non‐attenuated clinical organisms 48

9

Steve Baker Sanger Universi work we Previous antimicr developi resistant antimicr and replace phyloge Our dysenter income We

ran focu

hos are

m I w o a n t o o a s c n l p

y y n y as graphy stitute. ent

ountries bials bial s pathogens. g

ital

on

(Shigella), our

internati

of of

new

how of closely

access

surveil

resist lab resea

Ca gast of

sp w

e m m i e n o n o a a v v a a

e e nteric rch rointestinal lance. terventions nally bridge nt olve as s and cific This

linked located

pathoge

well

over

treat bacter

and recog

work bacte

Mor

as an

a m m t t s

r h n o h n n t o

o d n t o p d e p e

ria, ia will ent

pathogens e s.

ised ther

read the for

broadly that the the

next

looking

on

mainta

Wellco

progr diarrh

with

treat

caus

the sg 5

Q C T c p Q D A fi T D M i I e N 2 d I N P T D P r T P n n

n n

n n a a

o o e e h h 0 o a 0 h o e a h e

n i n y h h y n h h y O O O n y O O O r a i e r m a g i e m g c c o ew ontyphoidal in uantifying UBLICATI a ather ng mme

n vasive fants: rk i 18; gut ur

b47 sistant oviding emotherapy drews phoid mmunity ears losely Vietnam. ediatric ht e e

terica

Alwis I: I: I: eal

Quynh

infectious e

specific 10.1016/S1 10.1128/m 10.1038/s4 SE

an Role phylogeo

endemic

DOI: Variant Africa against

key

and microb ,

AE, diseas

R, Pham

JR, intern of inclu

A

@ conj Dise

Sero at Nhi

10.1

of typh Tu

Phu

Pro

Birt Bak are dia re

e .

A M L M s p

L, a

D f v d T e 0 v d u o a 2 e a e s u o a 2 e o a B o s

c B

s c r v 1 o e i r v t 1 o n 4 e i t g n 4 O

rrhoeal g as P h disease

93/infdis/jiy50 T… oid earch ational olutionary 018; ng ota de aternally e s ntimicrobia se ia tting gate eventing settings Salmonella io.01056 ective f 467 r ar 73 cus raphy

timicrobial

am.ac …

S…Lo Cohort Multidrug NS

such

TLT

program Alwis ‐

Baker our in

Typhimu Baker 3099(18)3 of

‐ fever

DOI: 018 and

on vaccin

Immu in

NC;

on

focu

Imm

and

R as

‐ dise Bake con

0737

Asia S; foc in 18

10.1 S;

ty

th St th

in

Ac

La

e

N

Jo

c h B p

B a p a n i u 7 s o c u a a n u e s n

m r u

t a e n m r i e t u h n n

i 0 e r u h a l ‐ . n n 0 0 r s a l ‐ .uk 093/jac/dky23 umans sub 1 in rium aker 0 quired urnal of Infect of urnal ncidence .

ough inued hoid ture access daptation

teric s

dy.

Resistant ocomprom cet 350

s: olera nity resistance. S; se

fections ‐ e

are generatio z

Urban

and

MBio a

in

in

‐ Infectiou S;

Sahara Commun 5

new

Associ Agains

Journa

Vietn infecti fever,

an the

coho

and Anti and in will

wor 2018

ur Vi

to of

lo

S

a

i a c n

e n e n n r k b b r k b b s m s a m a a r a r u u l i

o w

1 i o w 1

lead i ; t i nd ; t ious Disease ious ause ons. Antimicrobial t m Disease

sed multi cations tnamese otavirus.

ody an lmonella

ted bacillary

sage Africa.

l pact ‐ of

on studies

in middle

where

clonal

10

Patient

drug the with

in This into

the for drug

by 2018

of

2018

s

We are sstrongly focused on using genetic manipulation to discover the genes invvolved in various biological systems, using somatic mutagenesis in mice to discover cancer genes and genetic screens in ES cells in culture

Allan is also a serial entrepreneur and has established several biotechnology companies. While in the USA he founded Inc. that is publicly traded, and Spectral Genomics Inc. that was later acquired. In the UK he founded Kymab Ltd.. in 2010, and Petmedix Ltd. in 2017. PUBLICATIONS

Revealing hhidden complexities of genomic rearrangements generated with Cas9. Boroviak K, Fu B … Bradley A; Scientific Reports 2017; DOI: 10.1038/s41598‐017‐12740‐6

Bradley Comprehensive annotation and evolutionary Bradley insights into the canine (Canis lupus familiaris) antigen receptor loci. Martin J, Ponstingl H … Bradley A; Immunogenetics 20188; DOI: 10.1007/s00251‐017‐1028‐0

Molecular synergy underlies the co‐occurrence patterns and phenotype of NPM1‐mutant acutte myeloid leukemia. Dovey OM, Bradley A … Vassiliou GS; Blood 2017; 6 DOI: 10.1182/blood‐2017‐01‐760595

Enhancing the genome editing toolbox: genome wide CRISPR arrayed libraries. Metzakopian E, Strong A … Bradley A; Scientific Reports 2017; DOI: 10.1038/s41598‐017‐01766‐5

ab2592@c am.ac.uk

11 Allan Allan Gene mutations or environmental factors such as infection or drugs can have genome‐

wide effects on gene expression levels across many different pathways. However, it is

h not always clear which of these perturbations can be buffered by the system and which lead to detrimental outcomes as reflected in morphological changes or reduced health. c

i We inve stigate in vivo gene regulatory responses to challenges such as genetic mutations, chromatin disruption and infection. Specifically, we use our zebrafish mutant w archive of ~43,000 alleles covering 60% of protein‐coding genes, CRISPR/Cas9 t tih mutagenesis and genome‐wide sequencing approaches to understand the relationship of chromatin structure, transcriptional changes and phenotypic outcomes. For example, partial loss of gene function can have substantial effects on genome‐wide gene

en expression levels without gross morphological defects. This provides an opportunity to dissect gene regulatory networkks and distinguish network‐specific, but tolerated gene

N N expression changes, from those that affect organismal development and health. ‐

h PUBLICATIONS

The gene regulatory basis of genetic compensation during neurral crest induction. Dooley CM, Wali N … Busch‐Nentwich EM; bioRxiv 2018; doi.org/10.1101/314534

usc Placentation defects are highly prevalent in embryonic lethal mouse mutants. Perez‐Garcia V … Busch‐Nentwicch EM … Hemberger M;

B Bh 2018; doii: 10.1038/nature26002

A high‐resolution mRNA expression time course of embryonic development in zebrafish. White RJ, Collins JE … Busch‐Nentwich; Elife 2017; DOI: 10.7554/eLife.30860

th The age of heterozygous telomerase mutant parents influences the adult phenotype of their e offspring irrespective of geenotype in zebrafish. Scahill CM, Digby Z … Busch‐Nentwich; Wellcome Open Research 2017; DOI: 10.12688/wwellcomeopenres.12530.2 b bth

Loss of the chromatin modifier Kdm2aa causes BrafV600E‐independent sppontaneous melanoma in zebrafish. sa Scahill CM, Digby Z … Busch‐Nenntwich; PLoS Genetics 2017; DOI: 10.1371/journal.pgen.1006959

li li e m [email protected]

E E 12 We are a newly established research group that focuses on the antibody response to rotavirus infection and vaccination.

Protective rotavirus antibodies: Rotavirus is a common cause of gastroenteritis in childrenn, especially in low‐income countries. An estimated 215,000 children under five years olld die from rotavirus infection every year. Fortunately rotavirus vaccines have recently become available, but the exact mechanisms by which these vaccines work are poorly understood. This means that predictinng vaccine efficacy is a challenge.

We elucidate the mechanisms of antibody‐mediated protection to rotaviruus using molecular techniques. In collaboration with clinical partners, this enables the

development of accurate tests to predict rotavirus vaccine effectiveness, and help to develop the next generation of rotavirus vaccines.

Maternal antibodies: Rotavirus vaccines are negatively affected by the presence of maternal antibodies that are transferred from the mother to infant and protect the infant from infection. It is not known how maternal antibodies prevent vaccines from inducing protective immune responses in infants. We are using different models for rotavirus infection, combined with genetic approaches and deep sequencing to unravel the mechanisms of maternal antibody vaccine blockade. It is anticipated that understanding the activity of PUBLICATIONS maternal antibodies will lead to Complement C4 prevents viral infection through rational design of improved nneonatal capsid inactivation. Cell Host Microbe 2019 vaccines.

Caddy Characterization of innate immune viral sensors Caddy in patients following allogeneic hematopoietic

stem cell transplantation. Caddy SL et aal; Innate Immunity 2018; DOI: 10.1177/1753425918757898

Genogroup IV and VI canine noroviruses bind to histo‐blood groups antigens. Caddy S et all; Journal of Virology 2014; DOI: 10.1128/JVI.01008‐14

Norovirus‐Mediated Modification of the Translational Landscape via Virus and Host‐ Induced Cleavage of Translation Initiation Factors. Emmott E … Caddy S … Goodfellow; Molecular Cell Proteomics 2017; DOI: 10.1074/mcp.M116.062448

s lc50@c am.ac.uk

13 Sarah Sarah We research humoral immunity in humans, how this goes wrong in disease and how it can be manipulated therapeutically using experimental medicine studies. In collaboration with Glaxo‐Smith‐Kline we recently investigated how belimumab affects the B cell compartment in renal transplannt recipients,. We found it suppressed new antibody formation and skewed residual B cells to an IL10‐producing, regulatory phenotype.

Autoantibodies are associated with a number of kidney diseases, and antibody‐mediated rejection has a negative impact on transplant outcomes. We use human kidney tissue to profile resident immune cells using flow cytometry, mass cytometry, aand RNA sequencing, and complementary mouse models to understand function. We have shown how the intrarenal sodium gradient can influence mononuclear phagocyte position and function to optimise defence against ascending infection.

Although IgA is the dominant antibody isotype found at mucosal suurfaces during health, genetic variation in FcgRs can influence susceptibility to inflammatory bowel disease (IBD), suggesting that IgG may be important during inflammationn. We are currently investigating how the induction of IgG in the gastrointestinal tract can activate gut‐ resident cells, how this is regulated, and whether this is important in IBD.

Increasing evidence shows immune cells change their metabolism in response to activation signals and that some of PUBLICATIONS these changes may be required for

their function. We investigate how IgG Single‐cell transcriptomess from human kidneys reveal the cellular identity of renal tumors. immune complexes affect macrophage Clatworthy Clatworthy Young MD … Clatworthy MR … Behjati S; 2018; DOI: metabolism and influence IgG‐ 10.1126/science.aat1699

associated tissue inflammation. PI3Kδ hyper‐activation promotes development of B cells that exacerbate Streptococcus pneumoniae infection in an antibody‐independent manner. Stark AK … Clatworthy MR … Okkenhaug K; Nature Communications 2018; DOI: 10.1038/s41467‐018‐05674‐8

Belimumab in kidney transsplantation: an experimental medicine, randomised, placebo‐ controlled phase 2 trial. Banham GD, Flint SM … Clatworrthy MR; Lancet 2018; DOI: 10.1016/S0140‐6736(18)30984‐‐X

nna nna Renal Sodium Gradient Orchestrates a Dynamic Antibacterial Defense Zonne. Berry MR, Mathews RJ … Clatworthy MR; Cell 2017; DOI: 10.1016/j.cell.2017.07.022

m r [email protected]

Me Me 14 We use complementary genomics and phenotypic approaches to explore host/pathogen interactiions, and are interested in how pathogens colonise and cross mucosal surfaces and then spread and survive in the deeper tissues of the body. We use the analysis of microbe populations to identify candidate evolutionary signatures that we can then

analyse to determine their contribution to phenotype, if any.

In one aspect of our work we perform clinical studies that enable us to develop mmethods to analyse the pathogen, microbiota and host response simultaneously, and to bring genomics into clinical practice. We have interests in antibiotic resistancce, how pathogens evade therapies, and in vacciness. Our focused research topic for maany years has been typhoid, or enteric fever, and the Salmonella bacteria that cause it.

Our clinical work has enabled clinical teams to track infections by sequencing pathogens as they enter the hospital. We also look at environmental microbial contaminatiion using genomics and informatics approaches, to link specific environmental conteexts with patient isolates. This allows us to monitor antibiotic resistance, with the ambbition of promotiing the appropriate use of antibiotics.

Our work on typhoid has led to the creation of the global whole genome database for Salmonella Typhi which houses over 5,000 isolates from around the world. This database monitors the spread of typhoid and identifies novel antibiotic resistant poppulations. With othher groups we investigate typhoid pathogenesis using microbial geneticcs linked to field studies, and human challenge models using stem cells. The value oof these models lies in our ability to challenge PUBLICATIONS them with pathogens and ccompare Dougan Phylogeographical analysis of the dominant functional readouts to clinical Dougan multidrug‐resistant H58 clade of Salmonella outcomes. Typhi identifies inter‐ and intracontinental transmission events. Wong V, Baker S… Dougan G; Nature Genetics 2015; doi: 10.1038/ng.3281

Identification of enterotoxigenic Escherichia coli (ETEC) clades with long‐term global distribution. von Mentzer A, Connor TR … Dougan G; Nature Genetics 2014; DOI: 10.1038/ng.3145

Emergence of host‐adapted Salmonella enteritidis through rapid evoluton in an immunocompromised host. Klemm E … Dougan G … Kingsley RA; Nature Microbiology 2016; DOI: 10.1038/nmicrobiol.2015.23

Interleukin‐22 promotes phagolysosomal fusion to induce protection against Salmonella enterica Typhimurium in human epithelial cells. Forbester JL, Lees EA … Dougan G; Proceedings of the National Academy of Sciences 2018; DOI: 10.1073/pnas.1811866115 g d312@c am.ac.uk

15 Gordon

Ravindra Gupta control/ barrier lineage We resistan We

stud als

o t c c c o y

ells e ure

seek cure.

drug in

that both strate

and path

for

t

r r g g o o understandi m eated ies. gen

a

reservo

and This

and

h

i o o u u n n ir ng st ntreated cludes

of ‐

pathoge of

HIV

how

rk ide in

p w W M M m P 2 S w C W D T r f f G D M r A D M m a D D G h H P

n t n a a o a o a e e e e u u e e 0 0 n c n c O O O O O O O O u a u a

he NA IV lobal g20@c UBLICATI lcochova lcochova ith HIV tients ilure

acrophages therland noRes 16 r trospective striction ide tivates G1–like Isolates pta tification ematopoie I: I: I: I: interaction

10.1016/S1 10.15252/e 10.15252/e 10.1038/s4 ; 1

central

adult in

DOI: damage

RK

variatio remissi

is

of epide vitro Study

et infec

10.1 able

P, P,

SA

KA, W al

sta in

to

Su Ca HI

;

n

H H M G M G N 0 r N

o 0

m o a m a

t o

m 1 t o t t m 1

4 s . t e e t mbj.20169602 mbj.20179688

4 s ONS s n Collier te V–1 induced 38 roup; cohort O ature ed protease herland

f ic well 586 73 n to HD1

plication rvous iology

m.ac. to acrophage

in

/

stem novel recomm following ‐ sre

allows

3099(15)0 with

infect inform 019 SJ suscep

Lancet I Lancet

2019; p infecti

and DA … 38153

KA syst

stud ‐ ‐

102

of

Gup cell

by div

… d

H e in e b g

n d n e e

e e 7 b d n e e 7

u t f u t n

f t n t 5 G I 0 s y e 0 t

t 5 G IV–1 0 s o y

erse 0536 on Gupta s. ‐ hibitors terminants ibility ficiency. nded locks m rug on a transplanta opoisomer CCR5 fectious Dise fectious uccessful

4 . upta

k

RK;

and ‐

dividing ‐

resista 8

HIV EMBO

to RK;

Δ RK; first HIV of

32/ a

is

byp EM

an 1 HI th Sci ‐

m

g 1 Δ 2 n B l n B s s

a d a a d V a V e e i

0 i l 0 l a a t

t entific Report 32 ulti– ubtypes. ne O obal se ce se

of 18; ion. myeloid refore – infection ss

associatio

2017;

2016;

1

inhibitor followin regimen 16 SAMHD

drug

subtyp HIV centr

a

o n 1 e e g s s s f

The metabolic repertoire of immune cells, which encompasses metabolic enzymes/pathways, the available nutrient sensors and metabolic checkpoint kinases, and the epigeenetic programming of metabolic geenes, enables and modulates specific immune functions.

Our goal is to delineate the molecular basis of how cellular metabolism is regulated, and itself regulates, immune‐function in healtth and disease states, and to deffine how environmental cues are integrated at the cellular level by immune cells to shape cellular metabolism and function. Hess Hess

PUBLICATIONS

The spectrum of T cell metabolism in health and disease. Bantug GR, Galluzzi L … Hess C; Nature Reviews Immunology 2018; DOI: 10.1038/nri.2017.99

Mitochondria–ER contact sites are immunometabolic hubs that orchestrate the rapid recalll response of memory CD8+ T cells. Bantug GR, Fischer M … Hess C; Immunity 2018; DOI: 10.1016/j.immuni.2018.02.012

Memory CD8+ T Cells Require Increased Concentrations of Acetate Induced by Stress for Optimal Function. Balmer ML, Ma EH … Hess C; Immunity 2016; DOI: 10.1016/j.immuni.2016.03.016

Complement Regulates Nutrient Influx and Metabolic Reprogramming during Th1 Cell Responses.. Kolev M … Hess C … Kemper C; Immunity 2015; DOI: 10.1016/j.immuni.2015.05.024

Rapid effector function of memory CD8+ T cellss requires an immediate‐early glycolytic switch. Gubser PM, Bantug GR … Hess C; Nature Immunology 2013; DOI: 10.1038/ni.2687

c h [email protected]

17 Christoph

David Jayne the autoim training Europe and David have support Throug microbi rituxima and licensed and We at Our industry

opti

futu

con syst bio beli lup

ch

J m h m a a m o

a d e u r mumab, emic

b n

yne and e. nged me une d ising for uct developme s, arker ‐

courses Vasculitis

induced

genomic at

other

leads

in acade clinical lupus

diseas our

curre Adde

studi gran

alo

u

a m m i e e

n n n a n a n

n n n n n e u e e

S s and nt

rythematos trials clinical imed derstandin ia. dications. econdary t ociety. brooke’s e

s lomatosis

physician

have or therapie of

transcri

in

an and

defi at co ser

Thr int

m

m H

H a A v A s v g s s g s w o e w o e n n immunodefi us. ptomic ssociated the ospital, bination,

ice

rnational ugh ed of ith and of focus

It

these in

the

polyan

the has

first sever

stud has role Ca

dj

so

le

a c

bi w M P D Ja C A A E 2 K w p N R G v o E R M c B P c o n n m m

d d i c c 0 u g 0 r u g r a h h o a h h o f f f f O O o s o n o s nto o

ies

NCA zathioprine been e ciency, nditions. omarker asal yne UBLICATI d cClure 106@c onbichler ith cell iety of ficacy fect 18; iitis eumatology eumatology eudinterm sculitis. rrent paluni bridge llaborative global I:

ntrolled

long autoimm the

10.1002/ar

ANCA

in polyangi D DOI:

disease therap carriag he

of

Associ and

et

M B ANCA

‐ devel

S, and and term

al dise

clini 10.1 has

cell

Flos and A

an

;

in

w

St

ur Ar Ja …

e u o y d S u o 0 a 2 2 e

0

a e

o e

y s e a o

u y s a

c t u

a

c t i t c a i f

t i c J i f i J merging ONS outcomes afety 018; 018; targeted 93/rheumatol mann research

vasculitis, nitiated .40802 ne ne ne rk ted hritis &Rheu dius ayne tis. or al se in pment linical dy. studies is of m.ac. we

D

trials ANCA

in Mainten activity

the diseases Staphyl

proteo

DOI: DOI: Vasculit

D

in

O of

prima

Jones

are

prac …

pati curr of

10.1 10.1 Beli in Harr

a

tre ‐

Jay to th as ne

s j m i l

m A

A e o o n

e a o o n e e a m R t e t e m r n R t e e o u a n e s a e r o u a 0 0 e e s a r 0 0 e , , ison mumab twork is: ise repurpose NCA t int d

e coccus ; ries e y

nts gy/key317 ociated tment rapy 02/art.40776 38/s41584 xploring nce atology ANCA

nt

especially Nature Revi

three

k D;

by in systemic A

and working

EM; Preside

Arthritis with

of

Rando of vasculi

lupus Ken

with our

asso inte

opti Rheu Re and 2019

the

and vas

gr ‐ S

0 m

t m g m

1

m r 1

m a a e r e r o n

o n n h n h t &

t

c & c n ; n ; t m c t v v six national roup

ission between 8 ituximab

ws herapies nulomatosi is iated ns. ephritis. t e ith, ulitis: asculitis ature asculitis

ized ‐ atology

0065

of aimed months 18

nasal

that

has the

in

x of

s

Disease aassociation of genetic variation may pinpoint pathophysiologic mechaniisms and can give insight into protein function, paarticularly for receptors whose ligands are unknown or controversial. Coding variants often affect pathways of wider biological importance.

The orphan G‐protein coupled receptor Gpr35 is one such gene, where a coding variant has been associated with an increased risk for inflammatory bowel disease, andd primary sclerosing cholangitis that has an increased cancer risk. We investigate GPR35’s ffunction, and in particular the communication of immune cells expressing this receptor and their environment.

We are also interested in GPR35’s molecular interactions with other cell membrane proteins and its implications on signalling events and cell function.

PUBLICATIONS

GPR35 promotes glycolysis, proliferation, and oncogenic signaling by engaging with the sodium potassium pump. Schneditz G, Elias JE … Kaneider NC; Science Signaling 2019; DOI: 10.1126/scisignal.aau9048

Reversal of murine alcoholic steatohepatitis by Kaneider pepducin‐based functional blockade of Kaneider interleukin‐8 receptors. Wieser V, Adolph TE; Gut 2017; DOI: 10.1136/gutjnl‐2015‐310344

A matrix metalloprotease‐PAR1 system regulates vascular integrity, systemic inflammation and death in sepsis. Tressel SL, Kaneider NC; EMBO Molecular Medicine 2011; DOI: 10.1002/emmm.201100145

'Role reversal' for the receptor PAR1 in sepsis‐ induced vascular damage. Kaneider N et al; Nature Immunology 2007; DOI: 10.1038/ni1525

Reversing ssystemic inflammatory response syndrome with chemokine receptor pepducinss. Kaneider N et al; Nature Medicine 2005; DOI: 10.1038/nm1245

n k 428@c am.ac. uk

19 15 Nicole

Arthur Kaser determi we defining mediato Followin increase disease This biologic Using diseases betwee majority A habitat

single

hav app

a

n e o a

n

v rs

roach g in layer a

in Crohn's l f es ariety of

host

more

this

the mechanis key

incidenc genetical

of risk the

micro of path path, open and the

of for rec

ho dis int

t

w w e e C m C m e s e e e u e u s e e s e

m m b b ly t's we ntly

rohn's

chniques, ay stinal nfolded ase iota and up icrobiota s

suscepti

of that bona

have

a and

discov preval from

Crohn

wind

epith dise

are

ulc re fid pr

t i i b o d b o a e a p e n s p n s e h e

h e e o otein elial 's e le w

se, red affected orted cluding nce e rative thought

disease immune sterile to individu

leprosy,

cells of an

explor

resp

mec

colit

thes

ak ent co

ho

b s t p M P D C i C E T d D D Ki d A M H N s I D K T P o

o o e o m m h h y m y m a n a n a a t x s t x s a a a a i i h h i O O O

e e o i O O O e e o r i 13orf31 i c

m efective st parates KG2D

UBLICATI nse s. edicine and churtschentha ls, ress rely anisms sease. perimental M testinal 729@c role ser der thogenesis. ives somi ells. plex

be

e munometa I: I: I: risk diseases

the

WM,

tissue 10.1038/ni. 10.1038/n 10.1126/sci road

A

and

MZ,

at

system

promo

for Crohn' lead

& novel S genes enviro ‐ geneti A

medi

Kaser … 2017;

Blum the

Boro

to

of

(FA whi the los epi Kas onc AT of

a

C h t

m m

G e b G s e b

b t a A t

o M t t a A t o r M s t i i v r s

D e v a D c e rohn's nmental a o 3 bolic ler ostatin ONS ence.aao4158

c c

erg mmunomet r of he helial es complex ound Further

.4338 asis iak ted 16L1

dicine

h 532 OI: & disease

of w A IN) juvenile m.ac. models,

inflamma

M

Blumberg

MULT1 inflamm RS;

are gut,

K hypomo 10.1084/j

the

inflam

Blumber … … is of

functi ‐

medi the cell

Scienc 2017;

Kaser disea Kaser

a

M the

whi

the

‐ foll mu

fac ce lik

an

w e id in

w u m w

m

o

n a o

R n

n a c D R a e

e n c D A a e t e A e o g u A t o t A e s o g t uk a orld. t

se. rphic tualistic abolic iopathic

d cause p inflammato S; n. ted e h ory tral

inflammat ors tory OI: doplasmic m.20162041

ation.

; 2017; wing R; … ileitis.

Nature Imm densely regulatio Nature Me

itself explore

Adolph Journal of 10.1084/j

remova that regulat

bowel

autop bowel

this

for path

har

art rel

m TE;

p p t o

t o

n b n a b a d

r e d r e a h a h w h h o w h h o h h r o o

aradigm, l unology

Experimental ry eticulum y Journal of

m.20160791 ours icine

y tionship disease, disease. ritis. agy e r e and of pulated ay

trigger

of bowel bowel major

steep 20

IRE1

that

2017;

and

the

2016; α

Genetic studies have successfully identified many regions of the human genome that alter an individual’s risk of autoimmune or inflammatory disease, but the molecular mechaniisms that underpin these associatioons – and hence the reason they increase disease susceptibility – remain largely unknown.

We uncover the biology responsible for genetic associations in immune‐mediated disease by combining genetics, genomics, molecular biology, and immunologgy‐based approaches. We have a particular focus on the non‐coding genome, including ennhancers and non‐coding RNA, and the role that theyy might play in regulating immune responses. Our goal is to not only shed new light on pathways involved in disease pathogenesis, but to also provide novel therapeutic opportunities. Other interests include better understanding the genetic and biological ddeterminants of prognosis in autoimmune, inflammatory and infectious diseases ‐ as distinct from those pathways that drive disease development.

James currently holds a Intermediate Clinical Fellowship, and has

recently spent 2 years at Harvard as PUBLIC ATIONS part of this. He is also a member of

Genome ‐wide association study identifies the UK and International IBD Genetics distinct genetic contributions to prognosis and Consortia. susceptibillity in Crohn's disease. Lee JC et al; Nature Genetics 2017; DOI: 10.1038/ng.3755

Genome‐wide association study implicates immune activation of multiple integrin genes in inflammatoory bowel disease. Lee de Lange KMM … Lee JC … Barrett JC; Nature Genetics 2017; Lee DOI: 10.1038/ng.3760

Genome‐wide association studies in Crohn's disease: past, present and future. Verstockt B, SSmith KG & Lee JC; Clinical &Translational Immunology 2018; DOI: 10.1002/cti2.1001

Human SNP links differential outcomes in inflammatoory and infectious disease to a FOXO3‐regulated pathway. Lee JC et al; Cell 2013; DOI: 10.1016/j.cell.2013.08.034

Gene‐expression profiling of CD8 T‐cells predicts prognosis iin patients with Crohn disease and ulcerative colitis. Lee JC et al; Journal of Clinical Investigation 2011; DOI: 10.1172/JCI59255

j c [email protected]

21 18 James

Paul Lehner pathwa We eradicat silences (Human manipul Our and chromat innate extrachr chromo

ma stud use

n y s

a i o i inisation.

s on. ies omal dates mmune HIV Silencing ting

mosomal functional

appropri

have therape immun viruses machin As

and

chro lifelo

DNA

the

pr

In s m

H e H o s o y y

n e n e e e

a a ilencing. ndogenous utic HIV, stem

atin vided manipulate ub),

g allowing ry ted genetic ultimate

pathways HIV

to

targets.

it

silenci

a

by treat enabl uniqu

is activ nov

All

th

a u v

n m n e m i e

n n n

e n , i e , e i a e ation, ntracellular e cells ruses. d retrotransp

g cellular integrated, l

silenced ent.

replicatio insight

informs epigeneti pathway proteom

defend

and

Ret pj

int

pr

in m 2 R r T D B r C 2 T C H D T c m E P e e e e n c c n c c 0 0 e 0 0 e o u

o u u u h p

h p o O O o O O M h M h r r o t s o t s y y UBLICATI osons,

l30@ca ic hasovnikarova hasovnikarova c

ediates 18; 17; press gulates bbez rr oviruses, lls.

egrated s , e igenetic cesses their I: I: arcot per parasites,

and

poorly by chromatin

TM6

ML 10.1038/na 10.1126/sci transcript and HUSH

technolo

DOI: DOI: about

‐ ‐

Masson activat

avoid silenci ‐ youn geno

main Mari

Lehne

10.11 10.10 viral

pro

posi anti

pro

co HI sil ex

v V m v g m V m e v g m

e

p e l p e t i L t v O e

t i L t v O e ‐ 0 3 ike

iral elimination. g ional ng r tion ides on

IA, IA, viruses ure23643 regulation

ncing ains

tumour ‐ ressed

m.ac.u nce.aaa7227 retrotrans cures. ides 1/gr.228171. 8/ng.3878 … provirus e plex ies P Tooth NS

Lehner Timms Timms against

of

incomi

Dawson effect evasio HIV,

the

to

a coope

HUSH uniqu repre by

dise Our

pot

ret im

RT RT ex tar P

ide th

th

v

p i

e p

n n v a m p n n v … … a m e p … … 1 M r e n s e 1 M r

n s e

e g

i

a e g iral rates ase at ntify osons ariegation Rowe complex nd oviruses g 17 ression sor

Understan

k ntial tegrate discovery et

Lehner Lehner HUSH unity. A;

and insights

defies

Nature opportu viruses invasion

mutatio their

compl

HM; with

novel rout and

PJ; PJ; com offe

of

er Ge fu

20

N Sc t in

T

P e P o n e o n i e i e d h d h a a n a n a n n o r n n o r n n n R 1 n R n i 1 p i

to

D

ew ence ture Genetics x ing ost s dication through through ction ome Researc ome 7;

to f ities IM28

lex cellular

human

to in

‐ escape

L1 which HUSH

novel

22

genes.

MORC2 host host

how 2015;

HIV

cell and

for by

to

h .

Vaccines are one of the most powerful tools to prevent infectious diseases. New vaccine s are currently planned or already in the development pipeline. Epidemiological and clinical studies are necessary to determine the burden of existing diseases, the level of vaccine protection and identification of target populations. They can improve

existing vaccination schemes and quantify the add‐on benefits such as herd protection and vaccine effects pertinent to the reduction of antimicrobial resistance.

Jointly with the International Vaccine Institute in Seoul, Republic of Korea, and other

institutions, we operate field sites in 14 African and 12 Asian countries capable of conducting multi‐center epidemiological field studies that provide well‐characterized sampless for basic research and as a platform for the conduct of clinical trials.

Our research focuses on late‐stage support of generating clinical data for a novel typhoid conjugate vaccine through a cluster‐randomized Phase III trial in Ghanaa and an effectiveness trial in the Democratic Republic of the Congo, thereby generatinng data urgently required by the World PUBLIC ATIONS Health Organization and Gavi, the What is the best immunization strategy for Vaccine Alliance. protecting African children against invasive Salmonella disease? Jeon HJ, Pak GD … Marks F; Clinical Infectious Diseases This well‐established field network 2018; DOI: 10.1093/cid/ciy386 can be leveraged as a platform for scientists in CITIID and beyond to Updates estimates of typhoid burden in sub‐ Marks Marks Saharan Africa. add‐on study activities and obtain Kim J‐H, Mogasale V … Marks F; Lancet Global Health 2017; samples in high quality to execute

DOI: 10.1016/S2214‐109X(17)30328‐5 basic sciences research.

Incidence of invasive Salmonella disease in sub‐ Saharan Africa: a multicenter population‐based surveillance study. Marks F et al; Lancet Global Health 2017; DOI: 10.1016/S2214‐109X(17)30022‐0

Effectiveness of the Viet Nam produced, mouse brain‐derived, inactivated Japanese encephalittis vaccine in Northern Viet Nam. Marks F et al; PLoS Neglected Tropical Diseases 2012; DOI: 10.1371/journal.pntd.0001952

Sm‐p80‐based schistosomiasis vaccine: double‐ blind preclinical trial in baboons demonstrates comprehensive prophylactic and parasite transmission‐blocking efficacy. Zhang W … Marks F … Siddiqui AA; Annals of the New York Academy of Science 2018; DOI: 10.1111/nnyas.13942 fm471@ca m.ac.u k 23 Florian Florian

Nick Matheson cancer. includi metab collabo primar As pathog Studyi Evoluti host regulat our Amino acid dysreg causes Human infecti proteo by To

well disc Ami

cu

tr fa n o a o

r n u

y m o c

o e e

rate

no rent acid nsporters. g n. lism

as g tors lation approxima Immunode n nesis ver d

ics

human

different

by host amino has Acids

to proces target resear metab in

HIV and

of on

‐ char oth pro vir

hu

in

a

C f c u

e e t t e c e c s c s o c o o v o d v d

f f e acterise D4+ ely ndreds s

d es h iciency id proteomics r Cell iral r lism uced ll

by

related

therefore viral

biology, interacti

transpor and

1

infectio

the Cultur

T

is milli

an

path path of

Vir incr cell

cha vir

s p

‘ e o h h o u u e a r o e a o e a e r a u t r u s t s

ns easingly nges

ways ogenesis n s oteins nd ttings, , ns

rms , aims s.

(SILAC) we (HIV)

AIDS

and and and

suggest

theref

have in

race’ target to,

in

‐ intr infe

rela nj

re

an HI a 1)

M M M P D M V r C D M a M D G b r T D N p F P

b b o o n n c c s s e e e e a a u u c V e c V e O O O O O O O O reenwood

y rimary a ell y ed cts pu between ted d ognised road re d m25@c UBLICATI atheson atheson understand veals veals novel anipulation cellular cessories amati

mporal nctional I: I: I: I: ‐

lentiviral infected

Tandem T

10.1016 10.1016/j.c 10.7554/eL 10.7554/eL

surface by and

almost provide cell deaths

expert

A, viruse

antag remo

thera

hum et NJ Nef.

Willi

im EJ, pr an to

/j

al pr et

vi

t

.c p V T

V o

; m d m 4 o M d 4 o M M

a M d

a d s a o r s s a al i o o h o i i r s C roteomic a i o o

peutic s, ONS teomic if fe.18296 fe.41431 am.ac. se cells, 0 of every uses

mson n hape elling the om.2015.09. viro.2016.06. atheson ell Host Micr teomic unobiolog the nism cell ;

ass

variants. Current Opi Current we key CD4+

million

immuno in

import crime?

surfac

Tag JC

manip use

includ

and of the of appr

year

anal insi

NJ T atla

am

th

m

M

St c (T

p … m

i p T y e g a 0 y e g a 0 e m o t e a m a o e e m o t e a m 0 o e i . a

y

0 . a a y able MT) nions in Virol in nions ing ulating ino s , Lehner 03 he hts uk theson

lls. be

p

14 aches. ople

sis nce

host proteins mune etabolism We and of

of

2015;

cell

acid of ‐ cells

HIV

Isotop based

HIV

into of

world phos have 2) PJ; NJ; HIV

and surf

res the

me

inf

eLi id

inf eLi

th

d i

e p e b o p

b o p e p e n e n e

f f

a a u f u t f f w t f e w e by ey e analysing gy ntify pathways identified hoproteom oth e

onse ction fection unctional abolism ce ring Labelling

ction

2016; ide, 2019;

2016; HIV

infect. amino 24

viral new viral in

and

and

by

e

The immune response is a powerful and complex system that has evolved to protect us from pathogens and from tumours. Loosing control of that system results in immunopathology, manifest in a broad rangge of diseases including autoimmunity (such as T1D, lupus, multiple sclerosis), autoinflammmatory disease (such as Crohn’s disease,

Ulcerative colitis), infection and malignancy.

We use a systems immunology approach – modelling a broad range of quantitative traits as a me ans of discovering previously unsuspected relationships between them – to better understand the dysfunctional immune response in human disease. Our goal is to better understand what shapes an aberrant response and to use the information to indicate novel tests and/or interventions to help clinicians treating immune pathology.

Examples of the approach include, 1) the association of T‐cell exhaustion withh clinical outcome in numerous autoinflammatory/autoimmune diseases, 2) the use of this observation to develop a clinical assay for patient stratification, being brought to the clinic through a University start‐up (PredictImmune), 3) the identification of a natural killer cell subset associated with clinical outcome in multiple sclerosis, and 4) the identification of molecular signatures predicting clinical onset of T1D in at risk chiildren.

Ongoing projects include predicting and understanding clinical progression at various stages in T1D (in collaboration with the TEDDDY consortium, TrialNet and JDRF), multiple sclerosis (in collabo ration with the Immune Tolerance Network), and in human ageing (in collab oration with the SardiNIA consortium) in addition to a Wellcome‐funded study investigating novel ways of PUBLICATIONS modulating T cell exhaustion. Metabolic exhaustion in cancer, infection and autoimmunity. McKinney EFF & Smith KGC; Nature Immunology 2018. DOI: 10.1038/s41590‐018‐0045‐y

T cell exhaustion and immune‐mediated disease – the potential for therapeutic exhaustion. McKinney McKinney McKinney EFF & Smith KG; Current Opinion in Immunology 2016; DOI: 10.1016/j.coi.2016.09.005

T cell exhaustion: understanding the interface of chronic viral and autoinflammatory diseases. McKinney EFF & Smith KG; Immunology & Cell Biology 20116; DOI: 10.1038/icb.2016.81

T cell exhaustion, costimulation and clinical outcome in autoimmunity and infection. McKinney EFF, Lee JC … Smith KG; Nature 2015; DOI: 10.10388/nature14468 [email protected] 25 Eoin Eoin

Yorgo Modis functio cells respons non Our health level resoluti set it underst silencin sensing cell’s genomi must Viruses

silenc

of

res pat

re pr d

t b

e a h n

c

c g i e i o a e e hogenic tail. ncipal ophysical, nd depend erefore s ognise

cytosolic arch nding n s

DNA

against of

of viral

structur disease.

endoge This

integr is

into

innat

at

gen

and foc on

be

en

cy wi th

b v t e a e a u u

d d a a h

h t e i t e i e iochemical nised ll host

l ral

ssed osolic delivered silence ogenous ted

e

expression

antiviral molecular informati help

nucleus,

RNA.

cell viral on, viral

viral esta

the

m

1)

d A n

s o

o a

o o a D D b e b a e e

a . w

e n u . w

n d ucleic double

nd level how r n fence chinery lish expression quences. NA. In

here

additiona generate to

pursuit cell

basic cells

of obtai

Our

it acids is ‐

biol stra

ho can an to

d w w p p n n

o o o o n d n d o

o

, l , l istinguish y inflammato y express

rinciples

of f gical ded verarching

be

the detailed defence 2) in 10.1073/pn Fenwick SAM Structural 2018; Douse silencing ATPase Neuropat DOI: Yu different CryoEM DOI: Hwang interferon mitochon MAVS PUBLIC National Ac modis this

integrat

integrat

Q,

how the

RNA cell

appro 10.101 10.111

goal

Qu DOI:

enzy CH1,

MS,

and olig

MK

de cy

K

of di

p is t st

m

m m

m & w h b 1 A & w h b e

A s e a a h r

d a a h r m a 6 1 d a t B e m a 6 1 a t m B e

B

m

t B t 0 omers ructures tages

… studies y generated e osol. s.1705402114 y

d @mrc thogenic demy of Scien demy of echanistic /j.molcel.201 /febs.14772 how signaling. goal ects echanism oulanger ic ches, rial e d loor Modis .1038/s4146 TIONS replicate

erization

e.

response

Modis multiple

are viral into inflamma mutation

membr S

cytoso

viruse

… of Some is

Y; with apply

small

the

Y Modi

DN

of

Mol J

AT of …

to …

vi

vi t

d E s

A

A a a h 7 P n a 8 M h h i 7 P a

8 M h i M r t a e l a M r t s i i a t

l e l a

s s a t lic s ing s ynamics s nsights er tory ce perin, ‐ nd .10.012 al , hat ost

ructural

eir cular Cell

lick 018

ne

gain odis

hydrolysis. viruses mb.ca focus Y; in DA5 and and

the

2017; viral than

nucleic

Nature Co

amplifie

a

SE; ‐ remode MORC2 cell genes. is

03045

Y;

innate compl

cells activ

an dsRN a 4) trans FEBS J Proce

on

RNA

DOI:

80

gen wit

an 201

m als m an ‐ th

x u

a

n n e a e

d

d e h e h V e V e c s e d c s e

d m m m A e c

m

c o A l e c 8 c o 8 o

o tiviral perturb ted m

chanisms. ing and

chanistic dings of the ing oexist

riptional mentary ids ; iral immune , urnal

epigenetic me.

munications

atomic filaments cellular

3) deliver .ac.u induce

26

and

upon high

from RNA how how

The

2018; radic

in

GH ‐ ‐

a K

a k l L t

We use advanced bacterial genomics and metagenomics to better understand the evolution and spread of pathogens locallly, nationally and globally. Realising the complexity of big data and its limited understanding in most developiing and underdeveloped countries, we mine genomic data for robust, risk‐informative diagnostic markers. I use these markers and design smart detection probes as field‐compatible technoloogies. This technology‐driven global health approach makes my research accessible to the communities who need it most.

PUBLICATIONS

Defining endemic cholera at three levels of spatiotemporal resolution within Bangladesh. Domman D … Mutreja A … Thomson NR; Nature Genetics Mutreja 2018; DOI: 10.1038/s41588‐018‐0150‐8 Mutreja

Integrated view of Vibrio cholerae in the Americas. Domman D … Mutreja A … Thomson NR; Science 2017; DOI: 10.1126/science.aao2136

Genomic history of the seventh pandemic of cholera in Africa. Weill FX … Mutreja A … Thomson NR; Science 2017; DOI: 10.1126/science.aad5901

Retrospective Analysis of Serotype Switching of Vibrio cholerae O1 in a Cholera Endemic Region Shows It Is a Non‐random Process. Karlsson SL … Mutreja A … Lebens M; PLoS Neglected Tropical Diseases 2016; DOI: 10.1371/journal.pntd.0005044

am872@ca m.ac.u k

27 Ankur

J ames NhN ath an focusin 2 chroma dioxyge the relates 2 oxoglut and sensing A metazo The and ‐ ‐

OG OG

funda

sta can nu abi

d d

b t g a

a t e ependent nases lity to cers. mental rient pathways, ility

in. ns. rate pendent on

the

We to

HIF of

Central

(2 envir

have

sens

prol Hyp

‐ req aim sign OG

d d o o u

p u y p o y o e e , ,

ioxygenases diverse alling,

ioxygenases nment.

to l irement roviding xia

also to and hydroxyla

(i)

Inducibl this

know uncov

respo mito

fun

Our

for pr po c n s c r n o c o c s s e t

er tential

. e nd

, hondrial tions,

es cess

cell goal

as The transcript and

new

α‐ to (PHDs)

surviva

is

are ketog

most chan inclu (ii)

new

met to j

fu

a M M D G p h T 1 B H o M D M c H f T 2 St c M P a

g a a n n g g 0 0 e y 0 0 e y

u w u w g g h h d h h i O O

d O O r o e a r x o e a x

IF1 IF1alpha lutarate), l ain efanovic UBLICATI n33@c iles therapeutic on ctors, ice ain .1016/j.cmet. 18; itochondria ARCH6 ction tosolic

rr llular es group oglutarate bolic e e ing terotypic well I: I: lyubiquitin is hich

SP, 10.1016/j.c 10.7554/eLi vacuolar proteaso

α

GL1, AL, Factors

DOI: the in

insights novel alterin

stabilit

Costa

and iron descri Burr path sense TME

oxyg

Lobb

abili 10.15 Barre and of and

pr

a

S A c a i

l w M w e M o n n o

g P ‐ n n e

e g h 2 e e ( S

h 2 a m S y t n a m d e

y t

n d I e

A O t t l

A t t HIFs). 252/embr.201 nzymes fe.22693 bed chains. T TRC8

lyl targets 016.09.015 y sights d n

vels. t … ays romatin lrep.2015.06.

ail … d ehydrogen intracellula 199 protein into

cell in

TPase e

… m.ac. S,

NS

to Nathan iron

Nathan

homotyp availabili

hydroxyl distingui Nathan anchore Dickson aerobic

that

respon functio

and pheno

facilit

their Howe

in

for

com for

tha

JA; lip

JA; re inf

C ox JA;

A C c

o e

C

x a m

o f a m u

t 0 o S u c a t 0 i e S a d s i c a 7 t i ; e a d s C a ic d 7 t t Cell Metabolis plex atalytic d luence t r ver, ygen n DC115, biological nflammato y te

61 ypes ylation Cell Reports onditions. Life hes se 45603

tion

odelling.

proteins. k oxygen, to lysine require

is

of Nathan

the comple

2017;

2 and

betwe the

conser

by and ‐ these

by

OG

quali

the

con acti and

2 2 reg

11

loc

as JA;

re

m m d o

c

x r e e s 2 2 r r a a m

m l v t o v v u t e o v

u e a x a x t m

e t

inked embly EMBO Report y 015; n etabolite the rol ctivity controls enzymes y ed ity ygen, pendent ntrolling l lating levance,

odelling

2016 disease oxygen control

28

– in 2

the ;

all DOI: of 2 ‐

o

f s

The human gastrointestinal tract houses a large number of microbes, collectively known as the microbiota that supports host metabolism, immune development and pathogen resistancce. Intestinal dysbiosis, disruption oof bacterial diversity in the gut, can result from antibiotic use or pathogenic infection and has been strongly linked to increased disease susceptibility. This dysbiosis‐induced susceptibility indicates that normal commensal bacteria are capable of preventing disease; however, mechanisms involved in key host‐commensal interactions are still largely unknown.

We seek mechanistic insights into the influence of commensal microbes oon both intestinal infections and inflammation and systemic immune responsess. These fundamental insights will give us a bettter understanding of infectious diseases, autoimmune disorders and even cancer, and will enable the development of new approaches to combat these diseases. Using in vivo models, cellular immuunology, transcriptomics and proteomics and working closely with the Wellcome Trust Sanger Institute, we characterise the complex inteeractions between the commensal microbial community, intestinal epithelium and adaptive immune cells. Pedicord Pedicord PUBLICATIONS

Intestinal epithelial and intraepithelial T cell crosstalk mediates a dynamic response to infection. Hoytema van Konijnenburg DP … Pedicord VA … Mucida DD; Cell 2017; DOI: 10.1016/j.cell.2017.08.046

Exploiting a host‐commensal interaction to promote intestinal barrier function and enteric pathogen tolerance. Pedicord VA et al; Science Immunology 2016; DOI: 10.1126/sciimmunol.aai7732

A secreted bacterial peptidoglycan hydrolase enhances tolerance to enteric pathogens. Rangan KJ, Peedicord VA … Hang HC; Science 2016; DOI: 10.1126/science.aaf3552

Absence of MHC class II on cDCs results in microbial‐dependent intestinal inflammation. Loschko J … Pedicord VA … Nussenzweig MC; Journal of Experimental Medicine 2016; DOI: 10.1084/jem.20160062

[email protected] am.ac. uk

29 Virginia Virginia

Lalita Ramakrishnan pathoge lead In understa strategie Findings immune mutants. monitor tubercul tubercul zebrafis mechani specific Howeve that Tubercul In We

bacte

the

are

to are

d h

i r r n i

h

o o s mmunity. t ous rug s nterested composed nd

ia, ,

from he ms

ost

Our cell sis. sis

model strategies bacteria for

we the

infectio toleranc

infectio

caused of

We

resear we

chem

inter the

ident path

gran to

ex

se

T

i z s c n v o

n v o o o p

n p o n o c t n c t n o o e t e t uloma a ify ek e. by ention.

taxis, f hat loit h

udy

genesis brafish n the

process understan results

differenti

is and persist a

the

result

the

sheddi immun patho close

adhe form

char

Co opt in ba

of ha an

i

w n n g w n a t n s g a s n t n l v d g v d g d a s d acterise sion ation he eprosy. ical ity is enesis

ted sequently, e g enetic

ithin clearance

the

modulate

light

borne formatio of to

transpa macrop

and mech

and tuber granu

vastl

of

on

rela ag vir

o t

M m D T i t H D B M T L D C m e P D M l p A P

a i

T T n e u n e u u a u o o h h u t u o o y

y y a y e a e u u s O O O O t s O O O O t h h h h g c g c

n rency ost it we UBLICATI s hages, B adigan lomas prosy. nistic fections. berculosis lence acrophages macrophag r bin mbier ive igration. e rg sosomal

t 973@. cape ulosis. gregation, berculosis I: I: I: enolic I: enolic

of disease. role using

different pathogen

inflamma 10.1016/j.c 10.1016/j.c 10.1016/j.i 10.1016/j.c in RD,

DM, genotyp

of are

granulo

huma

CJ, from Levitte

CA1,

basis and facto

in

desp

M. lym Roca Glyc glyc

Zeb gen

O'Le

als

dis tu

C

g

t p

n

a n m m o m r a r m

o e b o r r

o e c b e e b u c e e e t e e o o e b u F e e e . t i e o o S F e . O i S a mas, orders hocytes s mbier afish nd susceptibili as te

ory lipid tically ll.2011.12.02 ll.2016.02.03 ll.2017.07.03 J y

sis, muni.2017.08 erculosis, ry

berculosis, lipid f netic

icrobicidal …

response am.ac directed populatio

that using NS comprom

mycobact

SM Ramakrish

the underst Ramakrish

well

respons funda

compl initiat CJ

are facilit

subv

tract defin drive

… dev

Ram

this an

as Ra na

t

m m t n n d e a h w n d 3 4 0 e h e s a h 8 w e a s 3 4 0 e e

t e a

t e a m a a e t n

m a a n o . o t .uk

tissue ising

turally nan erial .003 e ed ies

s ental erapies tes s rt the nding krishnan lopment x an susceptibili bility immune e other

akrishnan

zebrafish

Mycobact

to nerve

immune and

host

have L;

host

L;

mycoba

to

mycob agent Cell

macro persist

Cell

mech

of

resid

both

are susc imm

def L;

this da and can 201

de

20

ze

L; Im o

r

m m

1 e s e s v v f a e o f a e o C e C e p e b 6 e e p b 6 e i m i

a m a une t

t ence, cterial munity

2; tructures nforming optimize eloped nces host ell cterial gulation. ; bacterial f y age nt ptible rium hage rafish nisms antigen

disease.

odel

human to

2017; 30

cells.

in

and and

the

lepra

2017;

to to to

of

a ‐

e

Human cytomegalo virus (HCMV), the largest known human herpesvirus, is a major cause of disease in individuals whose immune systems are compromised or immaturee and is the

commonest infective cause of congenital damage to the central nervous systemm. Similarly,

it can also be life‐threatening in the context of organ or bone marrow trannsplant or advanced HIV infection.

Unlike many other viruses, HCMV is never cleared after primary infection but persists for the lifetime of the host. In part, this is due to the profound ability of HCMV to avoid the host’s immune response.

However, this lifelong viral persistence is allso underpinned by a biological property of all

herpesviruses; the ability to undergo latent infection where viral genomes are carried

silently in the absence of detection of infectious virions. It is now clear that that

reactivation of virus from these latent poolls is also a major cause of congenital infection

as well as causing much of the HCMV‐mediated disease obsserved in

immunocompromised transplant patients and patients with AIDS.

We seek the molecular basis of viral latency and reactivation of this persisteent human virus and how the host immune response combats virus disease. Such an understanding of how latent virus infection alters cellular functions to optimise the cell for latent carriage allows us to identify potential PUBLICATIONS “Achilles heels” of the virus to enable Defining the transcriptional landscape during us to target the latent viral reservoir in Cytomegalovirus latency with single‐cell RNA Sequencing. patient populations. Shnayder M, Nachshon A … Stern‐Ginossar N; MBio 2018; DOI: 10.11288/mBio.00013‐18

DeSUMOylase activity is required for Sinclair Cytomegalovirus reactivation from latency. Sinclair

Poole EL, Kew VG … Reeves MB; Cell Reports 2018; DOI: 10.10166/j.celrep.2018.06.048

Targeting the latent cytomegalovirus reservoir with an antiviral fusion toxin protein. Krishna BA, Spiess K … Sinclair. Nature Communications 2017; DOI: 10.10388/ncomms14321

Latency‐Associated Expression of Human Cytomegalovirus US28 attenuates cell signaling pathways to maintain latent infection. Krishna BA, Poole E … Sinclair JH; MBio 2017; DOI: 10.11288/mBio.01754‐17

[email protected]

31 John John Ken Smith kg M 1 T fl H D M o T 1 L s g G D M a M P e e u a u a 0 0 0 0 e e u u

O O O O u a u a ow enome UBLICATI cKinney cKinney cell e rlinton, .1038/ni.3209 .1038/ng.375 etabolic sceptibility netic cs2 toimmunit re? I: I: tcome

JC

10.1038/na 10.1038/s4

many

, exhaust

Biasci,

contri

@ DM ‐ EF, EF

wid in

ex

&

m

c Le

& a D

S e h 5 e h y u 5 y u

e

e 1 e a

e 1 a t i O t S in mith … butions

on,

ure14468 . austion

590 toimmunit mories mith association JC

m.ac.

Smith NS Crohn's

costimu KGC; 018

Smith KGC.

KG; ‐

0045 to doe

in Nat Na

K

N d p c p n y G y G u u a i u t u a i t a s ancer, s ‐ lation rognosis

sease.

ure Immunol ; y ture Genetics ture re Immunolo study and

it Nature

k

take

infecti

and

infec ident

2015 to

an

c m m o o o d g o t d g t l l ;

ifies inical 2017; gy ion

y n.

ake

2018;

2015;

32

and

distinct

an DOI:

DOI:

SLE

Synthetic biology seeks to engineer biological processes. “Xenobiology” goes further by manipulating biological processes using artiificial chemistries. For example, fuunctional oligos have huge potential for precision mediccine as they can be made to target tumours or pathoggens with remarkable specificity, and are easily prepared on a standard desktop oligo synthesiser.

Unfortunately, natural processes present some serious hurdles: nucleases severrely limit their stability in vivo, and their chemical unifformity impacts the type of interactions or mechanisms they can participate in. Taking lessons learnt from the medicinal chemistry of antisense therapeutics, artificial oligo chemistries – or “xeno nucleic acids” (XNAs) ‐ offer properties beyond those of DNA and RNA, such as reduced immunogenicity and nuclease‐ resistance in serum.

We have established methods to evolve biostable XNAs into high affinity aptamers for specific targets, into a variety of catalysts or “XNAzymes”, and self‐assembling nanoscale polyhedra. We are exploring the developpment of functional XNAs as a platform technology to direct or manipulate biologgy, for example using assemblies of XNA aptamers to target pathogens and tumor cells precisely modulate immune effector mechanisms in response to disease. XNAs hhave the potential to offer the molecular precision and function of bbiologics PUBLICATIONS but the advantages of short oligos, A synthetic genetic polymer with an uncharged including the ability to turn backbone cchemistry based on alkyl‐phosphonate nucleic acids. activities on & offf with Arangundy‐Franklin S … Taylor AI … Holliger P; Nature complementary strands as Chemistry 2019 antidotes. Beyond DNA and RNA: the expanding toolbox of synthetic genetics , RNA Worlds. Taylor AI, Hooulihan G. and Holliger P; 2019; CSH Press

Taylor Nanostructures from synthetic genetic polymers. Taylor Mutschler H,, Taylor AI, … Holliger P; ChemBioChem 2016;

DOI: 10.7554/eLife.43022

Catalysts from synthetic genetic polymers. Taylor AI, Pinheiro VB … Holliger P; Nature 2015; DOI: 10.1038/nature13982

Synthetic genetic polymers capable of heredity and evolution. Pinheiro VB, Taylor AI … Holliger P; Science 2012; DOI: 10.1126/science.1217622

a [email protected]

33 Alex Alex We elucidate the aetiology of human immune‐mediated diseases, to improve diagnosis

and treatment. We use integrative statistical analysis of genomic data and develop statistical methods to improve inference, or adapt to new biological teechniques: Genetic association discovery and fine mapping. This focuses on increasing power through optimal study design, borrowing information between related diseases using the conditional False Discovery Rate, and distinguishing the variant(s) driving the association from their correlated neighbours. Comparative genetic studies of immune‐mediated diseases. We usse co‐localisation to

integrate over the fine mapped posterior distributions of causal variants of different diseases to determine which diseases share which genetic risk factors Integrative analysis of genetic and omic data. Co‐localisation methods to enable us to understand disease mechanisms by mapping disease associated genetic variants to the genes they regulate. We also develop techniques to integrate genetic association data with maps of promoter‐enhancer chromatin contacts in the cell nucleeus. Dissection of heterogeneity within diseases. Human diseases have been classified according to clinical presentation, but there is increasing evidence that clinical symptoms

may not align to aetiological causse. PUBLICATIONS This is motivating a transition towards Chromosome contacts in activated T cells identify alternative aetiological, molecular, or autoimmune disease candiidate genes. treatment‐responsive, classifications Burren OS, Rubio García A … Walllace C; Genome Biology 2017; DOI: 10.1186/s13059‐017‐‐1285‐0 of diseases and their subtypes. Our comparative studies of diseases can A method for identifying genetic heterogeneity within phenotypically‐defined disease subgroups. inform treatment decisions using Liley J, Todd JA & Wallace C; Natture Genetics 2017; DOI: 10.1038/ng.3751 stratified medicine approaches. A pleiotropy‐informed Bayyesian false discovery Wallace Wallace rate adapted to a shared coontrol design finds new disease associations from GWAS summary

statistics. Liley J & Wallace C; PLoS Genetics 2015; DOI: 10.1371/journal.pgen.100449926

Lineage‐specific genome arrchitecture links disease variants to target genes. Javierre BM … Wallace C … Fraser P; Cell 2016; DOI: 10.1016/j.cell.2016.09.037

Statistical colocalization off genetic risk variants for related autoimmune diseasses in the context of common controls. Fortune MD, Guo H … Wallace C;; Nature Genetics 2015; DOI: 10.1038/ng.3330 cew 54@ca m.ac.uuk

Chris Chris 34 Infection by human cytomegalovirus (HCMV) of immunocompetent indiviiduals is generally asymptomatic or mild, and is self‐limiting. However, HCMV is never cleared after primary infection, but persists for the lifetime of the host as a latent infecttion that avoids immune surveillance. While HCMV is a paradigm for viral immune evasion strategies, the resolution of primary infection is dependent on effective CD4 and CD8+ T cell ressponses. In contrast, infection of individuals whose immune systems are compromised (such as HIV/AIDS patients and transplant patients) or immature (such as a foetus) often leads to wide spread viral replication and dissemination and this can often be life threatening. Similarly, reactivvation of the virus in individuals wwho are immunosuppressed also leads to significant morbidity and mortality.

We address a number of important questions concerning the immunobiology of HCMV both in lytic and during latent infection, with a particular focus on the generation, maintenance and function of memory CD4+ and CD8+ T cells and the immune evasion

mechanism employed by the virus to modulate Natural Killer cell function. We eestablish how this human pathogen persists for the liifetime of the host in the presence of a fully competent and potent cell mediated immune response. Our view is that this knnowledge PUBLIC ATIONS will allow us to develop credible strategies to make latently infected HCMV Specific CD4+ T Cells are poly‐functional and can respond to HCMV Infected Dendritic cells immunological targets and Cells in vitro. thus lead to the elimination of Jackson SE, Sedikides GX … Wills MR; Journal of Virology 2017; DOI: 10.1128/JVI.02128‐16 latent HCMV reservoirs in vivo and

in solid organ donor tissue, which Diverse speecificities, phenotypes, and antiviral activities of cytomegalovirus‐specific CD8+ T would have a significantt clinical cells. Jackson SE, Sedikides GX … Wills MR; Journal of Virology benefit for transplant patients. Wills 2014; DOI: 10.1128/JVI.01477‐14 Wills

Human cytomegalovirus latency‐associated proteins elicit immune‐suppressive IL‐10 producing CD4+ T cells. Mason GM, Jackson … Wills MR; Plos Pathogens 2013; DOI: 10.1371/journal.ppat.1003635

Human cytomegalovirus latency alters the cellular secretome, inducing cluster of differentiation (CD)4+ T‐cell migration and suppression of effector function. Mason GM, Wills M … Sinclair JH; Proceedings of the National Academy of Sciences 2017; DOI: 10.1073/pnas.1204836109 mrw1 [email protected]

35 Mark Mark IMPACT From genetic analysis to personaliised medicine in IBD

Professor Ken Smith

One in every three‐hundred people has inflammatory bowel disease (IBD, comprising Crohn’s disease & ulcerative colitis) in the developed world and its incidence is rising fast in developing countries. In the UK alone there are around 620,000 sufferers with an annual cost to the NHS in excess of £1bn. Around half of patients have an aggressive, relapsing disease course, while in others the disease is more quiescent.

Unfortunately, prognostic markers that can rreliably identify these patients are not available in clinical practice. This hinders disease management because patients with aggressive disease will be undertreated by conventional escalating dose therapy, while those with quiescent disease can be exposed to the risks and side‐effects of unnecessary immunosuppression.

Research in the Professor Smith’s lab found a transcriptional signature that is detectable within peripheral blood CD8 T‐cells (a type of white blood cell) at diagnosis and which correlates with long term clinical outcome in IBD. The signature reflects a process called exhaustion, in which T‐cells progressively lose the ability to sustain an effective immune response. Paatients following an aggressive disease course have less exhaustion than those with a quiescent course. However, translating these findings to the clinic originally presented a technical challenge as blood cell populations needed separating to see a signal, something that is not practical in a routine clinical setting. To meet this challenge the researchers utilised novel approaches too find a whole blood biomarker that could provide the same results without the need for cell separatiion.

The answer was to measure gene expressioon using microarrays and then use machine learning to identify the suitable whole‐blood biomarker. Ultimately this led to the identiffication of a 15 gene assay that could identify different disease courses by stratifying patients into aggressive and quiescent types of IBD, and an independent, prospective multicentre study validaated these findings.

Having established these principles, a spin‐out company (PredictImmune Ltd) wwas created through Cambridge Enterprise to translate this work into the clinic. The prognostic tests it provides for immune‐mediated conditions can aid disease management and improve patient outcomes. As differing IBD patients may require different treatment regimens, the test ensuures that they receive the most appropriate course of treatment and avoid excessive side effects resulting from over‐ treating the quiescent group, or excessive morbidity from under‐treating the aggressive group. With funding from the Wellcome Trust, Predictimmune is also running a biomarker‐stratified trial that is a first of its kind for inflammatory disease and will determine whether the biiomarker can deliver personalised care.

The test uses a small blood sample to accurately predict each patients’ outccome at the point of diagnosis, and is available as a laboratory testing service and soon also as a kit. The group is also developing predictive tests for other immune‐mediated diseases that pose long‐term burdens on patients as well as imposing a financial burden society. They include tests for lupus, multiple sclerosis, diabetes mellitus and rheumatoid aarthritis.

[email protected]

36 PATH WAY ANALYSIS I M PLICATE S REGUL ATION O F IMMUN E RESPO NSES

AND MONON UCLEAR PHAGOCYTES IN CROHN’S DISEASE PROGNO SIS

(a) Horizontal bar plot of –log10 (P values) for the top 40 most enriched pathwaays in an analysis of 29 LD‐pruned prognosis‐associated SNPs (meta P < 1 x 10‐5) across 1,751 pathways annotated by Gene Ontology. (b) Bar plot of –log10 (P values) for 155 specific cell‐types and conditions. Significant enrichment was observed in “monocyte‐derived macrophages stimulated by M‐CSF” and “monocyte‐derived macrophages stimulated by M‐CSF and interferon‐gamma”. Other non‐

immune cell‐types included neural, skin, lung, liver, stem cells, smooth muscle,, stromal cells, bone marrow progenitors, endothelial and epithelial cells. Dotted lines represent Bonferroni‐corrected significance thresholds. Analyses performed using SNPsea.

S EE THE WORK HERE

d oi.org/10.1186/ISRCTN118 08228 h ttps://www.p redictimmune.com/ 37 IMPACT New trreatments for Tuberculosis

Professor Ramakrishnan

Tuberculosis (TB) is an infectious disease thhat affects over 10 million people worldwide, of whom 10% die if the disease is untreated. Globally it is one of the top 10 diseases that causes death, and over 95% of these deaths occur in low and middle‐income countries. Ending the TB epidemic by 2030 is one of the health targets of the United Nations Sustainable Development Goals, but multidrug‐ resistant TB (MDR‐TB) is a continuing public health crisis and a health security thhreat.

Eradication of TB is impeded by the need for long complex treatment ‐ six months with four drugs ‐ for duraable cure. Furthermore, this need for long treatment could be the catalyst for the development of antibiotic‐tolerant TB. Reseearchers in Professor Ramakrishnan’s lab have found that the actively‐growing bacteria become antibiotic‐tolerant upon entry into host macrophages (part of our immune system, and the bacteria’s site of growth) by inducing bacterial effflux pumps. With this knowledge, the researchers are testing the therapeutic value of a several drugs that are known to inhibit these efflux pumps and are already approved for other purposes. For example, a clinical trial with verapamil (commonly used as an antiarrhythmic) as a treatment‐shortenning agent has been approved and funded by the Government of India and is entering Phase 2. Linked to this trial, studies to better understand bacterial efflux pump‐mediated drug tolerance mechanisms are in progress through a multi‐investigator collaborative grant from the British MRC and the Inndian Department of Biotechnology.

Other clinical trials are also underway for studies on meningitis TB, the most severe and lethal form of this disease. In this study, Professor Ramakrishnan and her colleagues found that TB susceptibility is heightened in conditions of inadequate or excessive inflammation, and is regulated by the balanced production of steroid called eicosanoids. The balance of these steroids is maintained by an enzyme that is referred to as leukotriene A4 hydrolase (LTA4H), and it is variantts of this enzyme that control the severity of TB meningitis. In collaboration with colleagues from thhe Oxford University Wellcome Trust Research Unit (OUCRU) in Vietnam, researchers from the Rammakrishnan lab have found that because of this variation in the enzyme, the standard therapy for meningitis TB is only effective in patients with a high inflammatory state, while patients with the low inflammatory state patients may be harmed by it.

Based on this finding, the OUCRU investigators have begun a randomised clinicaal trial where patients are genotyped and those with the low‐LTA4H genotype are given steroid treatment or placebo. If the previous findings are confirmed, then in the future steroid treatment will be restricted to the excessive inflammation subset, improving the outcome of both groups. To address this, in the meantime, the zebrafish is being used to reveal mechanistic details of the LTA4H‐mediated low and high inflammatory states. New targets for trreatments are being revealed and drugs that have already been approved for other purposes have been identified. The use of personaliseed genomics will have consequences for guiding clinical treatmentts, where patients from the two LTA44H genotypes will be treated with different therapies.

ag97 [email protected] .ac.uk

38

C E REBROSPINAL FLUID (CSF) LEVELS OF CYTO KINE EXPPRESSION IN

H UMAN I M MUNODEFICIENCY VIRUS (HIV)–INFECTED (( I) AND H IV‐ UNINFEC TED (UI) PATIENTTS WITH TUBERCU LOUS M EENINGITIS

Concentrations are in picograms/milliliter for all cytokines except interleukiin 6 (IL‐6), for which

concentrations are in nanograms/milliliteer. Statistical comparisons between HIV‐infected and HIV‐ uninfected were made by the Mann–Whitney test corrected for multiple testing across the 10 cytokines. Only P values of ≤ .05 are shown. Abbreviations: IFN‐γ, interferon γ; TNF‐α, tumor necrosis factor α.

SEE THE F ULL A R TICLE AT

DOI: 10.10 93/infdis/jix050

39 IMPACT The immunity of inflammatory bowwel disease is exposed

Professor Arthur Kaser

The incidence of Inflammatory Bowel Disease (IBD) is doubling every ten years. The increase in the number of children diagnosed with the dissease is of particular concern. Resiistance to drugs and patients who do not respond to treatment is also a challenge.

However, the picture is not all bad. While there was no available treatment 30 years ago, now at least, thhere are several therapeutic interventions that can be used. For example, anti‐TNF treatment transformed the lives of many following its approval in 1998, and antibodies against integrin and interleukin‐23 have been more recently been employed.

Studies by Professor Kaser’s lab, have been partly responsible for some of these successes. These have enabled the development of experimental methods for examining the ennvironmental stresses and microbes that could be responsible for triggering IBD. Simultaneously they have probed the genetics that are associated with the disease. They have found that of the over‐‐200 genetic loci that are associated with IBD, variations in one protein particularly stand out. Proofessor Kaser and his researchers have used state‐of‐the‐art methods to study it.

Unusually, the protein they identified was of unknown function and lacked any similarity to other known proteins. This in itself was unusuaal, yet it was also associated with an increased risk of juvenile idiopathic arthritis and leprosy. At the time the link between these diifferent diseases was striking, but slowly over the last two years a number of other genetic changes have been identified that unite them, something that suggests a common cause.

The scientists named the unknown protein FAMIN, a term that is short for Fattty Acid Metabolism– Immunity Nexus. This name reflected their finding that FAMIN was highly expressed in macrophages, where it interacts with fatty acid sysnthase and is involved in the metabolism of cellular lipids. A change in FAMIN had a negative impact oon the energy stores of these essential immune cells. Consequently, macrophages that contained mutations in FAMIN were less able tto kill bacteria. What is more, these effects were seen in the cells of mice and humans, a consistenccy across species that provided welcoming confirmation of the causal genetic risk to IBD.

Metabolic pathways in immune cells are eemerging as important determinants of immunological function and the work of Professor Kaser and his colleagues’ shows that a core metabolic regulator of immune cell function is affected by genetic variation that results in a predisposition to inflammatory diseases and infection. Such findings have profound implications for the treatment of IBD and pave the way for further studies of gene‐environmental interactions. Cambridge has consequently been the epicentre for clinical trials which are ongoing throughout the UK. Importantly, the studies have been accelerated by the unique collaborative environment that is provided by Cambridge, involving researchers in other University departments, as well as other Cambridge centres such as the Wellcome Sanger Institute.

[email protected]

40 FAMIN: A CAU SAL GENETIC RISK FOR INFLAMMATORY BOWEL DISE ASE

(a) Immunoblot analysis (IB) of FAMIN and FASN (right margin) in primary human macrophages

(MΦ) derived from peripheral blood mononuclear cells and in U937 and THP‐1 macrophages

differentiated with PMA (below blots), asssessed after immunoprecipitation (IP) with antibody to FAMIN (anti‐FAMIN), mouse immunoglobulin G (IgG; control) or anti‐FASN (above lanes) or without immunoprecipitation (Input; far riight). Left margin; molecular size, in kilodaltons (kDa). (b) PLA of FAMIN and FASN (yellow) in THP‐1 macroph ages also stained with the DNA‐binding dye DAPI (blue throughout). (c) Immunoblot analysis of FAMIN or GAPDH (loading coontrol) in lysates of HEK293 cells transfected to express humann FAMIN(p.2 54I) or FAMIN(p.254V) or with vector alone

(above lanes), assessed with (top blot) or without (bottom two blots) immunooprecipitation with

anti‐FAASN. (d) Immunofluorescence analysis of the co‐localization of FAMIN (red) with PMP70 (green) in primary human macrophages (enlarged from areas outlined in Supplementary Fig. 2a); far right, enlargement of the area outlined at left. (e) PLA of FAMIN and PMMP70 (red) in THP‐1 macrophages. (f) Immunofluorescence analysis of the co‐localization of FASN (red) with catalase (green) in mFamin+/+ and mFamin−/− M1 macrophag es (M1Φ) and M2 macrophages (M2Φ). (g)

Quantification of the results in f. (h) PLA of FASN and catalase (yellow) iin mFamin+/+ and

mFamin−/− M1 and M2 macrophages. (i) Quanficaon of the results in h. Scale bars (b,d–f,h), 5

μm. Data are representative of three independent experiments (a–e) or one experiment with three mice and six cells (f,g) or ten cells (h,i) imaged per sample (f–i; mean + s.e.m.).

SEE THE F ULL A R TICLE AT

D OI: 10.1038/ni.3532

41 IMPACT Improving sanitation in rural Africa

Dr Ankur Mutreja

Changes in behaviour can provide some of the most successful means of reducing the spread of disease.

In Ghana and Uganda there are often no esstablished infrastructures for sanitatiion and it is common for rural communities to relieve themselves in the open environment. Researchers in Dr Mutreja’s lab are providing evidence that the use of pit‐latrines can improve the health off these communities. They are taking samples from pit‐latrines and individuals within the communities, to provide evidence that pathogen burdens are decreased by following simple sanitation measures. The researchers later return to the communities to explain their findings and coordinate efforts with local community leaders and government officials to promote sanitation and reduce the spread of disease.

In one arm of these studies, samples are taken from the faeces of volunteers, and from pit‐latrines within village communities. By using whole genome sequencing (WGS) off these samples our researchers can examine their genetic components, to establish a highly detailed portrait of the pathogens contained within them, and to probe mechanisms of disease transmission.

Pit‐latrines are a low‐cost solution for reducing disease transmission and require minimal design and materials. By examining their microbial composition our researchers can determine whether the environment of the latrines kills harmful pathogens and limits the spread of disease. By collecting samples from different regions of the pit‐latrines, our teams can also establish the optimal depth at which they should be dug and provide communities and local authorities with practical advice.

In a second arm of these studies the Mutreeja lab trains local community healtth workers to create culturally sensitive digital media that that promotes WASH (clean Water, Sannitation and Hygiene) interventions. This enables these workers to share content with their commmunities using mobile communication platforms such as tablets and mobile phones. This approach provides a flexible solution that can incorporate new knowledge, beliefs and practices, and rapidly communicate these to local communities.

To enable this work, the researchers received an Impact Acceleration Award from the Economic and Social Research Council to foster relations between civil society, academia, NGOs and policy makers. Using a participatory approach they have joined with the community as equal partners, and have used sciientific evidence to promote the use of WASH in areas of Africa wheere clean water and sewage systems are absent. Taking this approach means their research is more responsive to community needs, overcomes many of the cultural barriers that exist within the communities, and engages stakeholders at both local and national levels.

am87 [email protected]

42 SANITA TION IN LOW AND MIDDLE INCOME COUNTRIES

S EE THE WORK HERE

DOI: 10.1 080/17 441692.2018.1536156 43 IMPACT Cholera: a global challenge

Professor Dougan

Infections are caused when disease‐causing agents enter the body and multiply. Cholera is one of these, for which it was estimated that globally there were 1.3 – 4 million cases in 2017 that resulted in 143,000 deaths. This is the consequence of the seventh cholera pandemic that originated in South Asia in 1961 and spread throughout the globe over the course of the next 50 years. Such information can help to determine the factors that are important in the spread of a disease and allows us to gauge the impact of future interventions. Hoowever, little was known about its propagation routes in Africa, the country most affected by this pandemic.

Global‐health researchers in Professor Dougan’s lab have reconstructed the spread of cholera across the African continent by analysing the genomes of 1070 cholera isolates. By mapping this data onto historical records of cholera outbreaks they found that resurgence of cholera in the 1970’s was the result of two strains, one in West Africa, and another in East Africa. The Wesst African strain first appeared between July and September 1970 and was likely to have come fromm Asia. Countries such as Angola saw infection despite being over 1000 km from the closest country with cholera at the time and this then led to an outbreak in Mozambique in 1973. Subsequent reintroductions led to new outbreaks extending over many years up to 2003. The East African strain also appeared in 1970, but was more closely related to strains from the Middle East, suggesting that it may have distinct origins. This strain was associated with several outbreaks over the next 40 yearrs, leading to 125,000 cases of cholera in South Africa in 2001‐2002, and 98,000 cases in Zimbaabwe in 2008‐2009. Originally, even these strains had originated in Asia, but some had circulated in the then endemic Middle East before moving into the East African region. Many of the outbreaks could be linked to large migrations resulting from war or from religious practices such as pilgrimage.

This work has also provided information about antibiotic resistance. Professor Dougan and his colleagues found that African isolates became increasingly resistant over time, and while the earliest of these were recovered in the early 1980’s, only resistant forms have been collected since 2000. This finding reveals that the spread of resistance is likely to have occurred by determinants that were already in these disease‐causing organisms as they spread, rather than independent local acquisitions of resistance. In other words, the bacteria already contained thee genetic information they needed to overcome antibiotics and its emergence was likely to have reesulted from human interventions such as the poorly coordinated use of antibiotics.

These high‐resolution genomic studies show how it is possible to use modern sscientific methods to monitor the spread of diseases, even long after outbreak events. The genomic ddata were consistent with events that were controlled by human activities, and demonstrated that human transmission chains are more important than climatic or environmental speculations. As such, these studies highlight that the long‐term spread and maintenance of cholera in Africa is largeely a consequence of direct, human to human transfer, or indirect infection by water polluted with human faeces. Such large‐scale scientific interpretations over long time scales have an enormous impact on our ability to govern the continued spread of this disease and anticipate future outbreaks.

gd312@cam .ac.uk 44 PHYLO GENY O F THE SEV ENTH P A NDEMIC V. CHOLERAE EL TTOR ISOL ATES

Maximum likelihood phylogeny of the

1070 genomes studied, including M66 as an outgroup. Branches are color‐ coded according to their geographic location, inferred by stochastic mapping of the geographic origin of each isolate onto the tree. (B)

Maximum clade credibility tree for a subset of 228 representative isolates. Twelve introduction events (T) are shown, along with three previously described waves (3) as coloured arrows.

Inferred propagation routes of the seventh pandemic V. cholera, showing twelve introduction events (T) with median date ranges for the most reccent common ancestor in

years. The first numbber indicates the median

MRCA of the African iisolates and their closest relative from the source location, and the second number indicattes the median MRCA of the African isolates. Introductions and inferred secondary transmission chains are indicated by thick and dashed arrows, respectively. Secondary transmissioon chains for West Africa in 1970 to 1971 are bassed on published records. The geographic presence of the various lineages

is indicated by a circle, triangle, and diamond for waves 1, 2, and 3, respectively, and coloured according to the inferred transmission events. The size of the shapes is proportional to the number of genomes analysed.

SEE THE F ULL A R TICLE AT

DOI: 10.1126/scien ce.aad 5901 45 NIH R Cam bridg e BRC P hen otypi ng H u b

CITIID also offers a range of scientific services. The NIHR Cambridge BRC Cell Phenotyping Hub provides state‐of‐the‐art flow cytometry, cell sorting and single‐cell technology facilities for the handling of unscreened human samples. It was established in 2011, to service the immunology community’s need foor human cell sorting and flow phenotyping. In 2012, 700 hours of cell‐sorting time was used. By 2018 this had increased to 3300 hours. This platform provides rapid access flow services to both fundamental and patient‐‐based research from 8.00am ‐ 11.00pm. In future the hub will incorporate a broader range of immunophenotyping and immunoometabolic analysis, coupled to a strreamlined process for analysing vaccine responses (to exxperimental or standard vaccination),, immunomodulatory and immunosuppressive clinical trials and studies that use geneticallly‐defined individuals recruited from the Cambridge BioResource.

The Hub offers a wide range of flow‐based approaches, including single cell sorting, multicolour phenotyping (16‐18 colours), multi‐way sorting, conccurrent sorting and ies ies phenotyping, apoptosis and proliferation assays, cell viability monitoring, assay development et cetera.

t t The following equipment is availabble as a service:

i i Attune NxT flow acoustic cytometer BD Arialll cell sorter BD Fortessa fow cytometer (four): 3 laser, 18 colour high speed flow analyser BD FACSantoll flow cytometer: 3 laser, 8 colour analyser BD Influx cell sorters BD FACSaria Fusion cell sorters with 16+ fluorescent channels BD FACSJazz cell sorter BD FACSCalibur cell analyser BD FACSMelody cell sorter Cellomics Arrayscan XTI automated cellular imaging and analysis Coulter Multisizer 3 flow cytometeer for cell size & concentration Leica Sp5 confocal Leica LMD6 laser capture microdissection microscope Leica SPE Confocal Leica LM Cellomics Arrayscan XTI high‐throughput screening microscope Opera Phoenix (Perkin Elmer) high content confocal Facil Facil SeaHorse XF96 measure OCR and ECAR of live cells in a 96‐well plate format Miltenyi AutoMACS Pro cell separator Symphony analyser (5 lasers, 28 detectors) MoFlo customised cell sorter Biorad S3 sorter Fortessa analyser e e Cyan analyser LSR II analyser Aria II sorter

Find us at https:// www.med.cam.ac.uk/nihr‐cambridge‐brc‐cell‐phenotyping‐‐hub/

Cor Cor 46 Imaging

The Wellcome‐MRC Cambridge Stem Cell Institute Advanced Microscope Facility is able to offer imaging technologies, training and support.

The variety of instruments and image analysis options in the facility ensure that we are able to meet many imaging requirements in‐house. We are able to provide invaluable tools for investigating cellular processes in both fixed and living cells in 2D and 4D. This is achieved by means of conventional imaging, HCS, confocal microscopy, time‐lapse, widefield & FRAP, FRET, FCS and FLIM.

We also offer post‐processing and image analysis tools for image volumes (deconvolution, 3D reconstruction, quantification) and assist users by developing custom analysis protocols.

Our range of equipment includes:

2x Leica SP5 TCS confocal microscopes Zeiss 710 confocal microscope with FLIM Zeiss 880 Multiphoton microscope Zeiss 700 confocal microscope Andor Revolution XD Spinning Disk confocal microscope Leica Matrix HCS screening/live cell imaging microscope Zeiss Imager fluorescence microscope GE Incell 2200 High Content Imager Essen Incucyte Zoom live cell imager Nikon Biostation IM live cell imager

CONTACT US: [email protected]

Facilities Computing

CITIID has a local high‐performance computing (HPC) cluster and is configured similar to the University of Cambridge HPC, to which it has access via two 10Gb links.

There are 200TB of networked data storage available, which is supported by a team of four people, led by Stuart Williams.

CONTACT US: [email protected]

47 Core Immuno‐ metabolism

The CITTID immuno‐metabolism facility offers cutting‐edge high resolution liquid chromatography mass spectrometry (LC‐MS) to solve small molecule (less than 2000 Da)

analytical needs. Whether you are interested in highly bespoke hypothesis driven work, absolute quantitation, tracking stable labelled substrates through core metabolic pathways or untargeted global metabolomic profiling with fragmentation based metabolite ID, the immuno‐metabolism facility can tailor the analysis to your needs.

We also have a suite of data processing packages to deliver fast turnaround and get the most information out of your data from large scale pattern recognition studies to small scale quantitative work.

We currently use Thermo Scientific orbitrap (FTMS) based high‐resolution MS coupled to a best‐in‐class Vanquish UHPLC+ chromatography system for conventional analytes such as lipids and less polar aqueous compounds, and a Dionex Ion Chromatography system for highly polar difficult to measure species.

With over 15 years’ experience in the field of small molecule MS we can meet any analytical challenge. The following equipment is available: Thermo Scientific Q‐Exactive Plus Orbitrap mass spectrometer Thermo Scientific Horizon UHPLC+ liquid chromatography Thermo Scientific Integrion ion chromatography system Software: Excalibur V4.1, Tracefinder Clinical V4.1, Comound Discoverer V2.1

CONTACT US: [email protected]

Facilities Bio ‐ Safety Level 3

CL‐3 containment facilities in CITIID allow scientists to work with non‐attenuated clinical organisms. As such, they provide a critical research capacity that allows investigators to study the emergence of lethal pathogens with global importance.

CONTACT US: [email protected]

Core 48 W H E R E T O FIND U S

Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID)

Jeffrey Cheah Biomedical Centre

Cambridge Biomedical Campus

University of Cambridge

Puddicombe Way

Cambridge

CB2 0AW

www.citiid.cam.ac.uk

The CITIID Directory was First Published in April 2019

Information was correct at the time of publication