Download the abstract book - EMBO Conference Tuberculosis 2016

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Infection wikipedia , lookup

Bacterial cell structure wikipedia , lookup

Globalization and disease wikipedia , lookup

Bacterial morphological plasticity wikipedia , lookup

Triclocarban wikipedia , lookup

Molecular mimicry wikipedia , lookup

Magnetotactic bacteria wikipedia , lookup

Tuberculosis wikipedia , lookup

Transcript
THANKS TO OUR SPONSORS
LabEx IBEID
TABLE OF CONTENTS
Page
General information ................................................................................... 6
Social events.............................................................................................. 7
Map of the campus .................................................................................... 8
Scientific program ...................................................................................... 9
Oral presentations.................................................................................... 21
Poster sessions
Poster session 1 ................................................................................. 87
Poster session 2 ............................................................................... 201
Poster session 3 ............................................................................... 259
Poster session 4 ............................................................................... 339
Authors and co‐authors index ................................................................ 393
Participant list......................................................................................... 409
Reproduction or exploitation, in any form, of the data included in this document are forbidden.
Chairs of the conference
Roland Brosch (Chair) and Pedro Alzari, Marco Bellinzoni, Carmen Buchrieser, Laleh
Majlessi, Giulia Manina, Roxane Simeone, Ludovic Tailleux (co-chairs)
Institut Pasteur, Paris, France
Scientific Committee
Pedro Alzari, Institut Pasteur, Paris, France
Clifton Barry, NIH, Bethesda, USA
Priscille Brodin, INSERM and Institut Pasteur de Lille, France
Roland Brosch, Institut Pasteur, Paris, France
Stewart Cole, EPFL, Lausanne, Switzerland
Sabine Ehrt, Weill Cornell Medical College, New York, USA
Lalita Ramakrishnan, University of Cambridge, UK
Eric Rubin, Harvard TH Chan School of Public Health, Boston, USA
Theme of the conference
The principle theme of this EMBO conference “Tuberculosis 2016: Interdisciplinary research on
tuberculosis and pathogenic mycobacteria”, consists of the latest biological insights and
advances on pathogenic mycobacteria that are presently gained by new powerful technologies and
sophisticated approaches linked to the domains of genomics, genetics, cell biology, structural
biology, medicinal chemistry and/or immunology and how these advances might be used to cope
with the enormous problems that pathogenic mycobacteria cause for millions of people today.
Pathogenic mycobacteria correspond to a large group of medically important bacteria that cause the
major human diseases Tuberculosis, Leprosy, and Buruli ulcer, but are also increasingly important
in the context of emerging pathogens in patients with genetic disorders, such as cystic fibrosis
patients.
These different themes will be presented and discussed by world-renowned scientists from a broad
range of different disciplines. This international conference, to be held from September 19-23,
2016, in the Conference centre (CIS) within the campus of the Institut Pasteur in Paris will provide a
premium forum for the discussion and exchange of the recent advances in this timely and important
topic.
Conference Center of the Institut Pasteur - 25-28 rue du Dr Roux - 75724 Paris cedex 15 - France
www.tuberculosis2016.org – [email protected]
GENERAL INFORMATION
W ELCOME DESK  OPENING HOURS
On the Institut Pasteur campus, the "Plan Vigipirate Attentats" is on, so please make sure to have an
official ID or passport on you to enter the campus.
If your registration is fully covered, you will receive your complete congress kit including your badge,
the certificate of attendance, the conference program and if pre-booked ahead, the voucher for the
conference dinner on Wednesday evening. Please wear your badge at all time.
If registration was not fully covered, please come directly to the registration desk "on site payment".
We accept payment by cash or credit card (Visa or Mastercard).
REGISTRATION DESK opens at 2:00 pm on September 19, 2016 in the hall of the CIS Auditorium.
Monday
Tuesday
Wednesday
Thursday
Friday
September 19
September 20
September 21
September 22
September 23
2:00 pm
7:30 pm
8:00 am
7:00 pm
8:00 am
6:00 pm
8:00 am
7:00 pm
8:00 am
5:30 pm
ORAL SESSIONS
Scientific sessions are taking place in the main auditorium of the "Centre d’information
Scientifique", located in the building CIS.
POSTER SESSIONS
Four poster sessions are scheduled in the hall of the CIS and in the Espace Congrès:
Poster session 1: Hall of CIS + Espace Congrès
Tuesday September 20, 2016, 1:45 pm - 3:30 pm - [Posters 65-176]
Poster session 2: Hall of CIS
Wednesday September 21, 2016, 1:45 pm - 3:15 pm - [Posters 177-231]
Poster session 3: Hall of CIS
Thursday September 22, 2016, 1:30 pm - 3:00 pm - [Posters 232-308]
Poster session 4: Hall of CIS
Friday September 23, 2016, 1:45 pm - 3:15 pm - [Posters 309-359]
Poster numbers are in the programme book. Check the matching number on the board to display
your poster in the right place. Magnets are available at the welcome desk to mount your poster.
COFFEE BREAKS AND LUNCHES
Coffee breaks will be served in the hall of CIS.
Lunches buffet will be served in the hall of the CIS and in the Espace Congrès throughout the
conference. Access to lunches is limited to participants who registered.
6
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
SOCIAL EVENTS
You are invited to join us for the Tuberculosis 2016 on
• Monday 19 at 7:30 - 9:30 pm: Welcome cocktail (hall of CIS)
• Tuesday 20 at 7:00 - 9:00 pm: Wine & Cheese by the posters (hall of CIS)
Conference dinner is scheduled on Wednesday 21, at 8.00 pm in the ‘Salon Honnorat-Maison
internationale’ at the Cité internationale universitaire de Paris.
The Cité Internationale universitaire de Paris was set up as part of the inter-war pacifist movement.
Its founders, driven by a humanist ideal, sought to create “a school of human relations to promote
peace” with a view to fostering harmony between different nations by furthering friendship between
students, researchers and artists from all over the world.
Address: 17 boulevard Jourdan - 75014 Paris
Access: Metro Line 6 from Pasteur (Nation direction) – stop at Denfert-Rochereau
+ connection with RER line B (train) from Denfert-Rochereau (Massy Palaiseau direction) – stop at
Cité universitaire (one stop). Journey: 15/20 minutes.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
7
M AP OF THE CAMPUS
CIS Building
Welcome desk
Auditorium (plenary sessions)
Espace Congrès
Coffee breaks, lunches & cocktails
Poster sessions 1 - 4
8
Lunches
Poster session 1-4
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
th
Monday, 19 September 2016
2:00 Arrival, Registration and Poster set up
5:40 Welcome addresses by Prof. Christian Bréchot, General Director of the Institut
Pasteur and the Organizers
1
Opening Keynote Lecture Session
Chair:
6:00 pm
7:20 pm
Howard TAKIFF
1 Cellular and molecular insights on the survival of Mycobacterium tuberculosis in
6:00 phagosomes
P. Brodin
Center for Infection and Immunity, Institut Pasteur de Lille - INSERM U1019, Lille,
France
2 Pursuing Edward Jenner’s Revenge: Novel Strategies for Sterilizing
6:40 Mycobacterium tuberculosis Infections
W.R. Jacobs
Microbiology and Immunilogy, Albert Einstein College of Medicine, New York,
United States
7:20 Welcome reception
th
Tuesday, 20 September 2016
2
Mycobacterial Genomics and Evolution
Chair:
8:30 am
10:15 am
Philip SUPPLY
3 TB under the sea: re-thinking the origins of Mycobacterium tuberculosis
8:30 T. Stinear
Department of Microbiology and Immunology, University of Melbourne, Melbourne (VIC),
Australia
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
9
4 Comparative genomics and phylogenetics of Uruguayan Mycobacterium bovis
8:55 isolates provide insights into their genetic variability
M. Lasserre
Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay
5 Generalists and Specialists in Mycobacterium tuberculosis
9:10 S. Gagneux
Medical Parasitology & Infection Biology, Swiss Tropical & Public Health Institute, Basel,
Switzerland
6 Origin of M. tuberculosis Beijing family and its possible hypervirulent
9:35 determinants
Q. Gao
Fudan University, Shanghai, China
7 Programmable transcriptional repression in mycobacteria using an orthogonal
10:00 CRISPR interference platform
J. Rock
Harvard School of Public Health, Boston, United States
10:15 Coffee break
3
Mycobacterial Biology & Physiology I
Chair:
10:45 am
12:30 pm
Stefan NIEMANN
8 Cell envelope remodeling and evolution of tuberculosis bacilli virulence
10:45 C. Guilhot
CNRS, IPBS, Toulouse, France
9 Mycobacterium tuberculosis periplasmic protease MarP activates the
11:10 peptidoglycan hydrolase RipA during acid stress
H. Botella
Microbiology and Immunology Department, Weill Cornell Medical College, New York,
United States
10 The tailoring modifications of Mycobacterium tuberculosis polysaccharides
11:25 M. Jackson
Mycobacteria Research Laboratories, Colorado State University, Fort Collins, United
States
11 Drug use on a single cell level
11:50 H. Rego
Microbial Pathogenesis, Yale, New Haven, United States
10
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
12 Single-cell approaches to unravel mycobacterial individuality and persistence
12:05 G. Manina
Junior Group Microbial Individuality and Infection, Institut Pasteur, Paris, France
12:30 Lunch buffet (Hall of CIS + Espace Congrès)
1:45 Poster Session 1 - No. 65 - 176
4
Transport systems & Secretion in Mycobacteria
Chair:
3:30 pm
4:50 pm
Riccardo MANGANELLI
13 The role of the ESX-5 secretion system, from nutrient acquisition to immune
3:30 modulation
W. Bitter
VU university medical center, Amsterdam, The Netherlands
14 Intercellular communication links ESX-1 to ESX-4 in mycobacterial conjugation
3:55 K. Derbyshire
Department of Biomedical Sciences, University at Albany, United States
15 Structure of the ESX-5 Type VII Secretion System
4:10 K. Beckham
EMBL - Hamburg, Germany
16 The Tuberculosis Necrotizing Toxin
4:25 M. Niederweis
Department of Microbiology, University of Alabama at Birmingham, Homewood, United
States
4:50 Coffee break
5
Mycobacterial Biology & Physiology II
Chair:
5:15 pm
7:00 pm
Brian ROBERTSON
17 Stress resistance, cell division and in vivo persistence
5:15 S. Ehrt
Department of Microbiology & Immunology, Weill Cornell Medicine, New York, United
States
18 Inherited cell-surface wave-troughs mark future division sites in mycobacteria
5:40 H.A. Eskandarian
Global Health Institute, UPKIN, École Polytechnique Fédérale de Lausanne, Lausanne,
Switzerland
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
11
19 The inosine monophosphate dehydrogenase, GuaB2, is a vulnerable new
5:55 bactericidal drug target for tuberculosis
V. Mizrahi
Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Cape
Town, South Africa
20 Functional, regulatory, and evolutionary aspects of the phage-shock-protein
6:20 response of Mycobacterium tuberculosis
M. Gennaro
PHRI, Rutgers University, Newark, United States
21 Playing nice together - making cell walls in mycobacteria
6:35 E. Rubin
Immunology and Infectious Diseases Department, Harvard TH Chan Scholl of Public
Health, Boston, United States
7:00 Wine and Cheese
st
Wednesday, 21 September 2016
6
Biomarkers, Profiling and Beyond
Chair:
8:30 am
10:25 am
Nathalie WINTER
22 Human CD8+ T-cells recognizing peptides from Mycobacterium tuberculosis
8:30 presented by HLA-E have an unorthodox Th2-like, multifunctional, Mtb-inhibitory
phenotype and represent a novel human T-cell subset
T. Ottenhoff
Leiden University Medical Center, Leiden, The Netherlands
23 Profiling persistent tubercule bacilli from patient sputa during therapy predicts
8:55 early drug efficacy
S. Waddell
Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
24 Understanding and Intervening in HIV-1 associated Tuberculosis
9:10 R. Wilkinson
University of Cape Town, Cape Town, South Africa Imperial College London The Francis
Crick Institute, London, United Kingdom
25 Condensation of DNA as a generic stress response in Mycobacterium
9:35 tuberculosis: apoptosis in bacteria?
N. Van Der Wel
Electron Microscopy Center Amsterdam, Cell Biology and Histology, AMC, Amsterdam,
The Netherlands
26 TB Subunit Vaccine Immunity Expressed in The Lung
10:00 P.L. Andersen
Statens Serum Institut, Copenhagen, Denmark
10:25 Coffee break
12
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
7
Whole cell vacines and beyond
Chair:
10:50 am
12:30 pm
Daria BOTTAI
27 VPM1002 as a TB prime vaccine, but also as post-exposure vaccine on the
10:50 horizon
L. Grode
Business Development, Vakzine Projekt Management GmbH, Honnover, Germany
28 Mycobacterium bovis BCG Δzmp1 – a promising, novel live vaccine candidate
11:15 P. Sander
Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
29 ESX-1 type VII secretion, cytosolic pattern recognition and consequences for
11:40 vaccine design
R. Brosch
Institut Pasteur, Paris, France
30 MTBVAC: from Discovery to Evaluation in High-Burden Countries
12:05 C. Martin
University of Zaragoza, Zaragoza, Spain
12:30 Lunch buffet (Hall of CIS + Espace Congrès)
1:45 Poster Session 2 - No. 177 - 231
8
Biochemistry & Chemical biology
Chair:
3:15 pm
4:35 pm
Patrick BRENNAN
31 The dynamics of TB lesions revealed by PET/CT scanning using [F-18] trehalose
3:15 C. Barry
Tuberculosis Research Section - LCID, NIH, Bethesda, United States
32 Flipping trehalose monomycolate across the inner membrane of mycobacteria
3:40 S.S. Chng
National University of Singapore, Singapore
33 Mechanisms of high fidelity DNA replication in Mycobacterium tuberculosis
3:55 M. Lamers
Structural Studies, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
34 Structural basis of phosphatidylinositol mannosides biosynthesis in
4:10 mycobacteria
M. Guerin
CICbioGUNE, Spain
4:35 Coffee break
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
13
9
Biochemistry & Chemical biology and beyond
Chair:
5:00 pm
6:05 pm
Katarina MIKUSOVA
35 Amino acid sensing by protein kinase G in metabolic regulation and virulence
5:00 H. O'Hare
Dept of Infection, Immunity and Inflammation, University of Leicester, Leicester, United
Kingdom
36 Targeting topology modulators and topoisomerases of Mycobacterium
5:25 tuberculosis
V. Nagaraja
Jawaharlal Nehru Centre for Advanced Scientific research & Department Microbiology
and Cell Biology, Indian Institute of Science, Bangalore, India
37 In Search of Drugs to Shorten Treatment of TB
5:40 C. Nathan
Depts. of Microbiology & Immunology and Medicine, Weill Cornell Medicine, New York,
United States
8:00 Conference Dinner - Cité Universitaire de Paris
Thursday, 22
nd
September 2016
10 NTM & Emerging mycobacterial pathogens
Chair:
8:30 am
10:15 am
Caroline DEMANGEL
38 The distinct fate of smooth and rough mycobacterium abscessus variants inside
8:30 macrophages
J.L. Herrmann
UMR1173, Université de Versailles Saint Quentin en Yvelines, Versailles, France
39 Global spread of Mycobacterium abscessus clones amongst cystic fibrosis
8:55 patients
J. Bryant
Department of Medicine, University of Cambridge, Cambridge, United Kingdom
40 A multidisciplinary approach to decipher the epidemiology of Mycobacterium
9:10 ulcerans infection: where do we stand?
S. Eyangoh
Head of Mycobacteriology Service, Centre Pasteur du Cameroun, Yaounde, Cameroon
41 NOD2, mycobacteria and chronic enteritis
9:35 D. Montamat-Sicotte
Research Institute of the McGill University Health Centre, Montreal, Canada
14
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
42 New Approaches to Target Mycobacterium abscessus Infections
9:50 L. Kremer
Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé (CPBS), Centre
National de la Recherche Scientifique FRE3689, Université de Montpellier, France
10:15 Coffee break
Table on (socio economic) impact of
11 Round
mycobacterial research
10:45 am
12:30 pm
10:45 Gilla Kaplan (Bill & Melinda Gates Foundation)
11:00 Brigitte Gicquel (Institut Pasteur Paris and Institut Pasteur Shangai)
11:15 Hannu Laang (European Commission, Brussels, Belgium)
11:30 René Coppens (TBVI, Lelystad, The Netherlands)
11:45 Christian Lienhardt (WHO, Switzerland)
12:00 General Discussion
12:30 Lunch buffet (Hall of CIS + Espace Congrès)
1:30 Poster Session 3 - No. 232 - 308
12 Mycobacteria and Host Interaction / Responses
Chair:
3:00 pm
4:45 pm
Laleh MAJLESSI
43 C-type lectin receptor DCIR modulates immunity to tuberculosis by sustaining
3:00 type I interferon signaling in dendritic cells
O. Neyrolles
Institute of Pharmacology & Structural Biology, CNRS-University of Toulouse, Toulouse,
France
44 Immuno-metabolic regulation of M. tuberculosis infection by HIF-1α and nitric
3:25 oxide
S. Stanley
School of Public Health, Molecular and Cell Biology, University of California, Berkeley,
United States
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
15
45 T cells and TB - do we know what kind of T cell works best?
3:40 A.M. Cooper
Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
46 Type I interferon production by B cells controls the balance between
4:05 immunopathology and pathogen burden in lungs during Mycobacterium
tuberculosis infection
D. Hudrisier
CNRS, IPBS, Toulouse, France
47 Anti-microbial effector pathways used by T cells to restrict intracellular M.
4:20 tuberculosis replication
S. Behar
Microbiology and Physiological Systems, University of Massachusetts Medical School,
Worcester, United States
4:45 Coffee break
13 Host Pathogen interaction and beyond
Chair:
5:15 pm
7:00 pm
Peter PETERS
48 Title to be confirmed
5:15 J. Cox
Berkeley University, San Francisco, United States
49 CD36-mediated uptake of surfactant lipids by human macrophages promotes
5:40 intracellular growth of M. tuberculosis
C.E. Dodd
Microbiology, The Ohio State University, Columbus, United States
50 Infecting Amoebae with Mycobacteria to Study Conserved Mechanisms of Innate
5:55 Immunity
T. Soldati
Department of Biochemistry, University of Geneva, Geneva, Switzerland
51 Insights into the host-pathogen and microbiome interactions from dual RNA6:20 sequencing of tuberculous sputum
R. Lai
The Francis Crick Institute, London, United Kingdom
52 The minimal unit of infection: M. tuberculosis in the macrophage
6:35 D. Russell
Microbiology and Immunology, Cornell University, Ithaca, New york, United States
16
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
rd
Friday, 23 September 2016
14 Drug design-new developments
Chair:
8:30 am
10:25 am
Giovanna RICCARDI
53 Reseting acquired antibiotic resistance in Mycobacterium tuberculosis
8:30 A. Baulard
Center for Infection and Immunity, Institut Pasteur de Lille et INSERM, Lille, France
54 Mycobacterium tuberculosis CTP synthetase and pantothenate kinase: two
8:55 promising targets for the development of multitargeting drugs
M.R. Pasca
Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia,
Pavia, Italy
55 Antitubercular profile of GSK070 a Mycobacterium tuberculosis leucyl-tRNA
9:10 synthetase inhibitor
A. Mendoza-Losana
TbDPU, GSK, Tres Cantos, Spain
56 First clinical trial with the novel drug candidate PBTZ169: expected results and
9:35 surprises
V. Makarov
Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian
Academy of Sciences, Moscow, Russia
57 Griselimycins, a new class of anti-TB drugs
10:00 S. Lagrange
Infectious Diseases Unit, Sanofi, Marcy L'étoile, France
10:25 Coffee break
15 From animal models to man
Chair:
11:00 am
12:55 pm
Ludovic TAILLEUX
58 Animal Models of Tuberculosis for Efficacy Testing
11:00 A. Rawkins
National Infections Service, Public Health England, Salisbury, United Kingdom
59 Tuberculosis and One Health: comparative analyses of the human and bovine
11:25 tubercle bacilli
S. Gordon
School of Veterinary Medicine, University College Dublin, Dublin, Ireland
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
17
60 Comparative Efficacy of New Oxazolidinones in a NHP Model of Tuberculosis
18
11:50 evaluated by [ F]FDG PET/CT and Bacterial Burden
L.E. Via
Tuberculosis Research Section, NIAID, NIH, Bethesda, United States
61 Evolution of Drug Resistance in Mycobacterium tuberculosis
12:05 A. Pym
Research Laboratory, AHRI (African Health Research Institute), Durban, South Africa
62 Severe childhood tuberculosis as a genetic disorder
12:30 S. Boisson-Dupuis
INSERM Hôpital Necker/Rockefeller University, United States
12:55 Lunch buffet (Hall of CIS + Espace Congrès)
1:45 Poster Session 4 - No. 309 - 359
16
Chair:
Closing Keynote Lectures Session
3:15 pm
5:00 pm
Pedro ALZARI
63 Interactions of mycobacterial phenolic glycolipids with host macrophages
3:15 L. Ramakrishnan
University of Cambridge, Cambridge, United Kingdom
64 Tuberculosis drug development: the leaking pipeline
4:00 S. Cole
EPFL, Lausanne, Switzerland
4:45 Closing comments / End of congress
18
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
ORAL PRESENTATIONS
SUMMARY
Page
Opening Keynote Lecture Session ............................................. 21
Mycobacterial Genomics and Evolution ..................................... 23
Mycobacterial Biology & Physiology I ......................................... 28
Transport systems & Secretion in Mycobacteria ........................ 33
Mycobacterial Biology & Physiology II ........................................ 37
Biomarkers, Profiling and Beyond .............................................. 42
Whole cell vacines and beyond .................................................. 47
Biochemistry & Chemical biology ............................................... 51
Biochemistry & Chemical biology and beyond ........................... 55
NTM & Emerging mycobacterial pathogens ............................... 58
Mycobateria and Host Interaction/Responses ............................ 63
Host Pathogen interaction and beyond ...................................... 68
Drug design-new developments ................................................. 73
From animal models to man ....................................................... 78
Closing Keynote Lectures Session ............................................. 83
1
Opening Keynote Lecture Session
19/09/2016
Cellular and molecular insights on the survival of Mycobacterium tuberculosis in
phagosomes
P. Brodin
Center for Infection and Immunity, Institut Pasteur de Lille - INSERM U1019, Lille, France
The highly successful pathogen Mycobacterium tuberculosis persists and replicates in
macrophages. It is then crucial to decipher the host-pathogen cross-talk which allows
M. tuberculosis to establish a niche favorable to the infection within such professional phagocytic
cells. More precisely the arsenal deployed by the pathogen consists of various cellular mechanisms,
which need to be elucidated in detail, including inhibition of phagosome maturation, block of
phagosome acidification, vacuolar rupture, activation of the inflammasome and resistance to killing
by oxygenated metabolites.
To identify the molecular and cellular players involved in these multiple intracellular mechanisms,
we have implemented high content screening approaches, using a combination of RNAi screens
and dynamic visual phenotypic assays (relying on automated confocal fluorescence microscopy) to
monitor the trafficking and replication of M. tuberculosis inside host cells. We thus identified novel
key host signaling pathways, which will be presented here.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
21
2
Opening Keynote Lecture Session
19/09/2016
Pursuing Edward Jenner’s Revenge: Novel Strategies for Sterilizing Mycobacterium
tuberculosis Infections
W.R. Jacobs
Howard Hughes Medical Institute, Bronx Microbiology and Immunilogy, Albert Einstein College of
Medicine, New York, United States
In 1789, when Edward Jenner inoculated a child with material from a cowpox lesion to prevent
smallpox, he was using a related but milder infectious agent to protect against a deadly pathogen,
an approach that ultimately led to smallpox eradication. Notably, Jenner’s family members died from
consumption (tuberculosis, TB), also prevalent at that time and a disease that continues to have
devastating consequences. Researchers Calmette and Guérin followed Jenner’s inspiration of using
a related bovine disease as a vaccine source, developing the BCG vaccine for TB. However, this
vaccine has failed to eradicate TB, and despite 30 years of genetically engineering BCG and
Mycobacterium tuberculosis, the resulting vaccine candidates are not substantially improved over
BCG. Short-course chemotherapies for TB were a great advancement of the last century, but
sterilization of the bacillus within human hosts requires prolonged treatment. I hypothesize that
vaccine ineffectiveness and the need for prolonged chemotherapies result from M. tuberculosis cells
entering a state that resists killing, a property called persistence. New strategies are required to
study and kill persistent M. tuberculosis cells. This talk will describe laboratory approaches for
observing persistent cells, including new reporter mycobacteriophages that enable visualization of
persisters. Through genetic manipulation of M. tuberculosis and improved knowledge of how
isoniazid works, novel strategies have been developed to sterilize cultures of M. tuberculosis and
overcome persistence in vivo. I will propose how a greater knowledge of persister cell killing could
lead to better TB chemotherapies. Lastly, in line with Jenner’s model of testing radically different
approaches for vaccines, I will describe how years of failure to develop recombinant BCG vaccines
against herpes simplex virus (HSV)-1 and HSV-2 led to the testing of a genetically engineered HSV2 ΔgD construct as a vaccine candidate. Serendipitously, this HSV-2 mutant induces sterilizing
immunity against HSV-1 and HSV-2, even though, contrary to the accepted dogma for an effective
herpes vaccine, it fails to elicit neutralizing antibodies. Rather, this vaccine induces non-neutralizing
antibodies that mediate antibody-dependent, cell-mediated cytotoxicity (ADCC). The implications of
this new HSV vaccine strategy for developing more efficacious TB vaccines will be proposed.
22
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
3
Mycobacterial Genomics and Evolution
20/09/2016
TB under the sea: re-thinking the origins of Mycobacterium tuberculosis
S. Pidot2, H. Izumi1, N. West1, J. Fuerst1, I. Monk2, J. Porter2, K. Mangas2, T. Seemann3,
R. Brosch4, T. Stinear2
1
School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane 2Department
of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity 3Victorian Life
Sciences Computation Initiative, University of Melbourne, Melbourne, Australia 4Unit for Integrated
Mycobacterial Pathogenomics, Institut Pasteur, Paris, France
During a study of microbial diversity in marine sponges, a mycobacterium (strain name: FSD4b-SM)
was isolated in pure culture from tissue of a Fascaplysinopsis sp. sponge, taken from waters on the
Great Barrier Reef in Queensland, Australia at a depth of 26 metres. FSD4b-SM replicates slowly
(doubling every 10 days), growing preferentially in liquid sea-water media up to 30°C. Initial 16S
rRNA gene sequence comparisons suggested that it was closely related to the M. tuberculosis
complex (MTBC). We established the complete genome of FSD4b-SM. With a single circular
5,575,519 bp chromosome harbouring 4624 protein coding DNA sequences (CDS), it is larger than
the MTBC genomes. However, DNA and protein sequence comparisons of 2500 core CDS
confirmed this mycobacterium is the most closely related species yet discovered to the tubercle
bacilli. Despite the unusual origin of the bacterium, the FSD4b-SM genome revealed extensive
conservation of genes associated with M. tuberculosis pathogenesis, including ESX-1, ESX-3, ESX5 secretion system and associated effectors, PE/PPE proteins, and signature MTBC cell wall
glycolipids (e.g. PGLs, mycoketides). There were 418 CDS specific to FSD4b-SM with predicted
functions likely related to growth within the marine environment, including membrane antiporters
and permeases, production of osmoprotectants and novel secondary metabolite loci. There were
also 900 CDS present only in the MTBC. The discovery of FSD4b-SM (proposed name
Mycobacterium spongiae sp. nov.) adds to growing evidence that the recent evolutionary origins of
the MTBC were likely marine and it provides an additional important resource as we refine our
understanding of the genetic factors that shaped the evolution of M. tuberculosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
23
4
Mycobacterial Genomics and Evolution
20/09/2016
Comparative genomics and phylogenetics of Uruguayan Mycobacterium bovis isolates
provide insights into their genetic variability
M. Lasserre4, G. Greif4, M. Castro-Ramos1, A. Juambeltz1, H. Naya3, C. Robello4-2, L. Berná4
1
Departamento de Bacteriología, División de Laboratorios Veterinarios (DI.LA.VE.) 2Dpto. de
Bioquímica, Facultad de Medicina 3Unidad de Bioinformática 4Unidad de Biología Molecular,
Institut Pasteur de Montevideo, Montevideo, Uruguay
Bovine tuberculosis is one of the most important diseases in cattle as well as being a zoonotic risk.
Cattle slaughter as means of contingence results in losses in the production of dairy and meat
products. The existence of strain-specific phenotypes of Mycobacterium bovis, characterized by
having different ranges of incidence, prevalence and/or severity, highlights the necessity of strainlevel discrimination of M. bovis by means of molecular typing approaches.
We selected 37 local strains isolated for spoligotyping, and ten of these strains for whole genome
sequencing. Genomic DNA was paired-end sequenced and assembled, and comparative analysis
were performed.
Amongst the studied strains five spoligotype patterns were detected, belonging to the British clonal
complex European 1, which is rarely found in mainland Europe and the most frequent in South
America. Spoligotypes identified (SB0274, SB0145, SB0130, SB0140, SB1072) had a frequency
and distribution pattern showing local traits concordant with cattle movement and different from
what is found in Brazil and Argentina.
SNP calling against the reference AF2122/97 revealed homogeneous SNP distributions along the
genome. Among the SNP-densest regions, lppl and pks12 showed the highest mutations, the latter
being widely associated with pathogenesis and host immunomodulation in Mycobacterium
tuberculosis. MntH, arsB2 and moeB1 showed to be truncated in all strains, suggesting a potential
alteration against the reference that could preview a bigger genomic difference.
Comparative analysis split these strains into two groups: those with spoligotypes SB0145/SB0130,
and SB1072/SB0274. The former showed an absence of the region of difference RD3 that contains
ORFs from phiRv1 prophage, not absent in the latter. Truncated genes evidenced a pattern of
incidence between these groups, commonly belonging to strains of the same group. SNP
phylogenetics supported the segregation between these groups. Nonetheless, SNP PCA nor
genome multiple alignment showed a clustering adequate to these groups or locality of origin.
Our comparative study shows that there is a bigger pattern of genomic variability than the provided
with a spoligotype classification, the latter being correlated to SNPs but not to genomic structure.
This analysis sheds light on M. bovis genomic variability as well as the nature of Uruguayan strains.
24
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
5
Mycobacterial Genomics and Evolution
20/09/2016
Generalists and Specialists in Mycobacterium tuberculosis
S. Gagneux
Medical Parasitology & Infection Biology, Swiss Tropical & Public Health Institute, Basel,
Switzerland
Generalist and specialist species differ in the breadth of their ecological niche. Little is known on the
niche width of obligate human pathogens. We analyzed a global collection of Mycobacterium
tuberculosis Lineage 4 clinical isolates, the most geographically widespread cause of human
tuberculosis. We show that Lineage 4 comprises globally distributed and geographically restricted
sublineages, suggesting a distinction between generalists and specialists. Population genomic
analyses revealed that generalists exhibited a higher diversity in human T cell epitopes, and support
a European origin for the most common generalist sublineage. Hence, the global success of
Lineage 4 reflects distinct strategies adopted by different sublineages and human migration.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
25
6
Mycobacterial Genomics and Evolution
20/09/2016
Origin of M. tuberculosis Beijing family and its possible hypervirulent determinants
Q. Gao
Fudan University, Shanghai, China
Mycobacterium tuberculosis Beijing family is the most successful genotype and responsible for
more than a quarter of the global tuberculosis epidemic. As the predominant genotype in East Asia,
the Beijing family has been emerging in various areas of the world and is often associated with
disease outbreaks and antibiotic resistance. We characterized the global diversity of this family
based on whole-genome sequences of 358 Beijing strains and show that the Beijing strains
endemic in East Asia are genetically diverse, whereas the globally emerging strains mostly belong
to a more homogenous subtype known as “modern” Beijing. Phylogeographic and coalescent
analyses indicate that the Beijing family most likely emerged around 30,000 y ago in southern East
Asia, and accompanied the early colonization by modern humans in this area. By combining the
genomic data and genotyping result of 1,793 strains from across China, we found the “modern”
Beijing sublineage experienced massive expansions in northern China during the Neolithic era and
subsequently spread to other regions following the migration of Han Chinese, supporting a parallel
evolution of the Beijing family and modern humans in East Asia. The dominance of the “modern”
Beijing sublineage in East Asia and its recent global emergence are most likely driven by its
hypervirulence, which might reflect adaption to increased human population densities linked to the
agricultural transition in northern China. We further analyzed online available sequencing data of
1082 M. tuberculosis Beijing isolates and determined the genetic changes that were commonly
present in modern Beijing strains but absent in ancient Beijing strains. These genetic changes
include 44 single nucleotide polymorphisms (SNPs) and 2 short genomic deletions, which
significantly enriched in the function category of regulatory proteins. We are now applying RNA-Seq
and Lipidomics analysis to further elucidate the mechanisms that underlie the successful expansion
of modern Beijing strains.
26
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
7
Mycobacterial Genomics and Evolution
20/09/2016
Programmable transcriptional repression in mycobacteria using an orthogonal CRISPR
interference platform
J. Rock1, F. Hopkins1, A. Chavez2, M. Diallo1, M. Chase1, E. Gerrick1, J. Pritchard1, G. Church2,
E. Rubin1, C. Sassetti6, D. Schnappinger5, S. Fortune1-3-4
1
Harvard School of Public Health 2Wyss Institute for Biologically Inspired Engineering, Harvard
University, Boston 3The Broad Institute of MIT & Harvard 4The Ragon Institute of MGH, Harvard
and MIT, Cambridge 5Department of Microbiology and Immunology, Weill Cornell Medical College,
New York 6University of Massachusetts Medical School, Worcester, United States
Mycobacterium tuberculosis (Mtb) is a globally important pathogen for which new drugs and drug
regimens are desperately needed. Treatment of Mtb infection requires multidrug therapy both to
minimize the evolution of drug resistance and to ensure complete clearance of the pathogen.
However, in the process of drug development, it has been difficult to identify drug target
combinations that are likely to display therapeutic synergy, largely as a result of laborious genetic
manipulations in Mtb and the inability to interrogate multiple genes simultaneously. CRISPR
interference (CRISPRi) repurposes the CRISPR/Cas9 immune system for the sequence-specific
control of gene expression and, in model bacteria, has allowed robust and regulated knockdown of
gene expression. However, in Mtb, the existing Streptococcus pyogenes-based CRISPRi system is
of limited utility because of relatively poor knockdown efficiency and proteotoxicity. We
hypothesized that orthologous Cas9 alleles from other bacterial species might allow more robust
control of gene expression in Mtb with less toxicity. We screened eleven diverse Cas9 alleles in vivo
and identified four that are broadly functional for targeted gene knockdown in mycobacteria. The
most efficacious of these alleles, the CRISPR1 allele from Streptococcus thermophilus (dCas9Sthe#1),
typically achieves 20-100 fold knockdown of endogenous gene expression. In contrast to other
CRISPRi systems, we show that gene knockdown in mycobacteria is robust when targeted far from
the transcriptional start site, thereby allowing high resolution phenotyping in the context of bacterial
operons. Finally, we demonstrate the utility of this system by investigating synthetic genetic and
chemical interactions within the mycobacterial folate synthesis pathway, an important but
controversial drug target for drug resistant Mtb. We anticipate that this CRISPRi system will have
broad utility for functional genomics, drug target screening, and genetic interaction mapping.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
27
8
Mycobacterial Biology & Physiology I
20/09/2016
Cell envelope remodeling and evolution of tuberculosis bacilli virulence
C. Guilhot
CNRS, IPBS, Toulouse, France
The etiologic agent of tuberculosis (TB) are bacteria from the Mycobacterium tuberculosis complex
which gathers sub-species each able to cause disease in humans; the most efficient of them in
humans is M. tuberculosis, which remains a major public health problem worldwide. Persistence
and efficient transmission are the hallmark of this disease and determine the TB pandemics.
However, the bacterial functions required for these key steps of the infectious cycle are mostly
unknown. M. tuberculosis is thought to have emerged from a pool of mycobacteria resembling
Mycobacterium canettii, a group of bacterial strains suspected to be more adapted to environment
and which caused few cases of human TB, mostly in East Africa. An intriguing question is what
adaptations were associated with the enhanced capacity of M. tuberculosis to invade humans and
to become the highly successful pathogen actually circulating worldwide. Later a phylogenetic
lineage of the M. tuberculosis complex specialized on non–human mammals and lost the ability to
sustain in humans. The key determinants of these evolutions of tubercle bacilli virulence are still
mysterious. However, recent findings have suggested that discrete rearrangements of the cell
envelope composition have been key determinant to these evolutions. These results will be
presented and discussed.
28
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
9
Mycobacterial Biology & Physiology I
20/09/2016
Mycobacterium tuberculosis periplasmic protease MarP activates the peptidoglycan
hydrolase RipA during acid stress
H. Botella4, J. Vaubourgeix4, W. Xu4, S. Song4, H. Makinoshima2-1, M.H. Lee3, M.S. Glickman2,
S. Ehrt4
1
Division of Translational Research, Exploratory Oncology Research & Clinical Trial Center,
National Cancer Center, Kashiwa, Chiba, Japan 2Immunology program, Memorial Sloan Kettering
Cancer Center 3Medicine, Weill Cornell Medical college 4Microbiology and Immunology
Department., Weill Cornell Medical College, New York, United States
Mycobacterium tuberculosis (Mtb) can persist in the human host in a latent state for decades, in part
because it has the ability to withstand numerous stresses imposed by host immunity. Former
studies have established the essentiality of the periplasmic protease MarP for Mtb to survive in
acidic media and establish and maintain infection in mice. Here, we document that—when
subjected to acidic stress—MarP cleaves the peptidoglycan hydrolase RipA, a process required for
RipA’s activation. Failure of RipA processing in MarP-deficient cells leads to cell elongation and
chains formation, a hallmark of progeny cell separation arrest. To our knowledge, this is the first
example of a protease that activates a peptidoglycan-degrading enzyme by proteolytic cleavage.
Our results suggest that sustaining peptidoglycan hydrolysis may be essential to Mtb’s survival in
acidic conditions.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
29
10
Mycobacterial Biology & Physiology I
20/09/2016
The tailoring modifications of Mycobacterium tuberculosis polysaccharides
M. Jackson2, S. Angala2, M. Mc Neil2, G. Larrouy-Maumus1, M. Gilleron1, L. Shi2, H. Pham2,
H. Škovierová2, J. Nigou1, W. Wheat2
1
IPBS-CNRS, Toulouse, France 2Mycobacteria Research Laboratories, Colorado State University,
Fort Collins, United States
The covalent modification of (lipo)polysaccharides with discrete substituents plays important roles in
the physiology of Gram-positive and Gram-negative bacteria and their interactions with the host.
This presentation will review what is known of the tailoring modifications of the major cell envelope
polysaccharides of Mycobacterium tuberculosis, arabinogalactan and lipoarabinomannan, their
biosynthetic origin and biological functions.
30
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
11
Mycobacterial Biology & Physiology I
20/09/2016
Drug use on a single cell level
H. Rego2, E. Rubin1
1
Harvard School of Public Health, Boston 2Microbial Pathogenesis, Yale, New Haven, United
States
Even in a genetically identical population, bacterial cells display a remarkable amount of
physiological diversity on a single cell level, which can lead to important phenotypic differences.
This seems especially true for the human pathogen Mycobacterium tuberculosis, which causes
tuberculosis (TB). TB is notoriously difficult to treat because small subpopulations die more slowly
despite being genetically identical to the vast majority of bacteria that are quickly killed by
antibiotics. While we have known about this phenomenon since antibiotics were first discovered,
little is understood about the underlying mechanisms, especially outside of the well-studied
organism E. coli. Using a combination of fluorescence single-cell methods and bacterial genetics I
have been investigating this question in mycobacteria. Mycobacteria create asymmetry from the
very beginning – at cell division – and I show that this asymmetry propagates to differential antibiotic
killing in the population. I then identify a gene – lamA - which upon deletion creates a more
homogenous population of cells that die more rapidly with antibiotic treatment. These results
suggest that mycobacteria create heterogeneity at a very high frequency with specificity and that
subversion of this heterogeneity is a viable option toward improved TB therapy.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
31
12
Mycobacterial Biology & Physiology I
20/09/2016
Single-cell approaches to unravel mycobacterial individuality and persistence
G. Manina
Junior Group Microbial Individuality and Infection, Institut Pasteur, Paris, France
Clonal cells grown under identical conditions exhibit strikingly different behavior, also known as
phenotypic heterogeneity. While part of this heterogeneity is caused by inherent randomness of
intracellular processes, most of cell-to-cell variation grounds on deterministic bases. Both social
context and surrounding environment can greatly impact the phenotypic spectrum of individual cells,
possibly resulting in beneficial variation. We found that also Mycobacterium tuberculosis displays
phenotypic heterogeneity at steady state with respect to growth potential and metabolic activity, and
this variation rises when cells are subjected to environmental perturbations. The ability of
M. tuberculosis to explore a wide phenotypic landscape may be associated with its tendency to
persist not only to protracted antibiotic therapy but also to the multifaceted host milieu. We are
currently exploring whether and how phenotypic heterogeneity contributes to mycobacterial
persistence in its broadest sense.
The combination of fluorescently-labelled bacteria, real-time microfluidic microscopy and
multiparametric analysis enables quantitative and predictive understanding of the behavior of
individual cells over time, under tightly controlled environmental conditions and within hostbiomimetic platforms. By studying mycobacteria at the single-cell and subpopulation level, we aim to
discriminate stochastic noise from robust phenotypic switches and their correlation with bacterial
fitness. We strive to elucidate the molecular bases of phenotypic heterogeneity and its implications
for pathogenesis and adaptation, giving special attention to the quiescent bacterial reservoirs and
their signatures of persistence. We postulate that subverting phenotypic heterogeneity, by finetuning the behavior of discrete subpopulations, may aid us to weaken bacilli and tackle the hurdle of
persistent tuberculosis. In conclusion, the identification of subpopulation-specific instead of averagepopulation biomarkers will be instrumental to develop enhanced disease control strategies and to
possibly shorten the treatment duration.
32
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
13
Transport systems & Secretion in Mycobacteria
20/09/2016
The role of the ESX-5 secretion system, from nutrient acquisition to immune modulation
L. Ates5, E. Houben4, R. Ummels5, C. Jiménez5, R. Warren3, A. Abdallah2, A. Pain2,
R. Hernández-Pando1, W. Bitter5-4
1
Nat Inst of Med Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico 2KAUST, Thuwal,
Saudi Arabia 3Stellenbosch University, Stellenbosch, South Africa 4VU university 5VU university
medical center, Amsterdam, The Netherlands
Mycobacteria are well protected from effectors of the immune system and from antibiotics by their
unusual diderm cell envelope. Nonetheless, pathogenic mycobacteria require a large number of
secreted proteins for nutrient acquisition and for survival inside the host. Therefore, specialized
secretion systems must be in place to transport proteins to the cell surface or into the environment,
without affecting membrane permeability. Most of the known effector proteins are transported via
the different type VII secretion (T7S) systems. The most recently evolved T7S system is ESX-5,
which is unique for the slow-growing mycobacteria. ESX-5 is responsible for the secretion of dozens
of substrates belonging to the PE and PPE families. A number of pathogenic mycobacteria,
including M. tuberculosis and M. marinum, produce an extraordinary large number of these PE and
PPE proteins, suggesting that they play a role in virulence.
We study the secretion of PE and PPE proteins by ESX-5 and the role of these proteins in
mycobacterial survival and virulence. We have shown that ESX-5 is essential for M. marinum, but
that this essentiality can be rescued by increasing the permeability of the outer membrane, either by
altering its lipid composition or by the introduction of the heterologous porin MspA. Examination of
phenotypes on defined carbon sources revealed that an esx-5 mutant is strongly impaired in the
uptake and utilization of hydrophobic carbon sources. Therefore, we propose that ESX-5 system is
responsible for the transport of cell envelope proteins that are required for nutrient acquisition.
These proteins probably compensate for the lack of MspA-like porins in slow-growing mycobacteria.
In our search for novel ESX-5 components we identified two independent transposon mutants in M.
marinum that were deficient in PE_PGRS secretion but situated outside of the esx-5 locus. Both
mutants had a transposon insertion in a single specific ppe gene. Although secretion of many
different PE_PGRS proteins was blocked, this mutation had no effect on bacterial growth. Deletion
of the orthologous gene in M. tuberculosis also blocked PE_PGRS secretion. Proteomic analysis
showed that in fact the two largest subsets of ESX-5 substrates, i.e. PPE-MPTR and PE-PGRS, are
not secreted by this mutant. Interestingly, hypervirulent clinical M. tuberculosis isolates of the Beijing
lineage have a natural mutation in this gene and a concomitant loss of PE/PPE secretion.
Restoration of this secretion defect reverted the hypervirulence phenotype of such a Beijing strain.
Therefore, these specific PE/PPE substrates seem to play an important role in virulence
attenuation.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
33
14
Transport systems & Secretion in Mycobacteria
20/09/2016
Intercellular communication links ESX-1 to ESX-4 in mycobacterial conjugation
T. Gray1, R. Clark2, N. Boucher2, P. Lapierre2, C. Smith2, K. Derbyshire1
1
Department of Biomedical Sciences, University at Albany 2Division of Genetics, New York State
Department of Health, Wadsworth Center, Albany, United States
ESX (Type 7) secretion systems in mycobacteria have many diverse roles. ESX-1 alone appears to
play three roles in the process of Distributive Conjugal Transfer (DCT), involving the transfer of
chromosomal DNA from donor to recipient strains of Mycobacterium smegmatis. Loss-of-function
studies have shown that ESX-1 suppresses DNA transfer in the donor, but the recipient requires
ESX-1 activity to receive DNA. GWAS-like analyses of transconjugants further showed that genes
within ESX-1 determine the mating identity of the conjugal strains. Reasoning that DCT requires
coordination between participating strains, we hypothesized that the diverse functions exhibited by
ESX-1 secretion are consistent with intercellular communication networks. We developed a SNPguided RNA-seq approach to examine the strain-specific transcriptional responses to co-culture
under mating conditions to identify the effects of communication. One of the most highly induced
transcripts was esxUT, encoding the hetero-dimeric secretion substrate EsxUT. ESX-4 is the
evolutionary progenitor for the other mycobacterial ESX secretion systems, but has no described
function to date. The co-culture transcriptional response we observed was specific to the recipient
strain, and to esx4, since the other esx systems (1 and 3) remained transcriptionally unchanged.
This recipient ESX-4 response required a functional recipient ESX-1 system, indicating a
dependency not previously seen between paralogous ESX systems. Importantly, ESX-1 mutant
donors elicited a greater esxUT transcription response in the co-cultured recipients, consistent with
intercellular communication. Mutation of several genes in the seven-gene esx4 locus in the recipient
strain prevented DNA transfer, but did not affect DCT when mutated in the donor. These data
identify a biological role and provide a quantifiable assay for the enigmatic ESX-4 secretion
apparatus in M. smegmatis. Collectively, this study supports a model wherein ESX-1 coordinates
intercellular communication between co-cultured conjugal donor and recipient strains, leading to an
activation of ESX-4, and culminating in DCT. Our findings begin to explain some of the disparate
functions of ESX-1 for DCT in M. smegmatis, and suggest that the many ESX systems of
actinobacteria may also play an unexpected novel role: mediating intercellular communication.
34
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
15
Transport systems & Secretion in Mycobacteria
20/09/2016
Structure of the ESX-5 Type VII Secretion System
K. Beckham1, L. Ciccarelli2, C. Bunduc3, A. Parret1, W. Bitter4, E. Houben3, T. Marlovits2,
M. Wilmanns1
1
EMBL - Hamburg 2University Medical Center Eppendorf, Hamburg, Germany 3Vrije
Universiteit 4VU University Medical Center, Amsterdam, The Netherlands
Bacteria utilize a diverse repertoire of secretion machineries to facilitate the transport of a range of
substrates in and out of the cell. The structure and function of these systems from Gram-negative
bacteria have been the subject of investigation for many years, furthering our understanding of hostpathogen interactions mediated by these systems. However, the secretion machineries from Grampositive bacteria are less well understood, despite their importance in the virulence in several
pathogens. Arguably the most the important human pathogen, Mycobacterium tuberculosis, relies
on the activity of the T7SS to infect the human host. Pathogenic mycobacteria encode up to five
T7SSs within ESX loci (ESX-1 to 5) which are structurally similar but functionally distinct as these
systems have been shown to play defined roles within the cell. This functional diversity is thought to
be mediated partially by the specific range of substrates that are secreted by each ESX-system, for
example the ESX-5 system secretes a diverse range of PE/PPE substrates that modulate the
infection process whereas the ESX-3 system has a role in iron transport.
Currently, little is known about the structure of the T7SS and the mechanism by which it
translocates substrates across the diderm mycomembrane. Previous studies have shown that the
T7SS complex comprises four conserved components, which assemble to form a 1.5 MDa complex
(Houben et al., 2012). However, the stoichiometry and arrangement of the core components within
this complex is unknown. To investigate this further, we have reconstituted a functional ESX-5
system in Mycobacterium smegmatis, which allowed for the analysis of the role of specific
substrates in ESX-5 functioning and their interactions (see also the abstract of Bunduc et al.). Using
the reconstituted system we optimized the purification protocol of the ESX-5 membrane complex.
Characterization of the complex using DLS, small-angle X-ray scattering and electron microscopy
has provided novel insights into this integral-membrane machinery. Here we report the first threedimensional reconstruction of the structure of the T7SS using electron microscopy. These novel
insights into the structure of this nanomachine further our understanding of the molecular
mechanisms of substrate translocation across the mycomembrane.
Houben et al. (2012) Composition of the type VII secretion system membrane complex. Molecular
Microbiology, 86: 472–484.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
35
16
Transport systems & Secretion in Mycobacteria
20/09/2016
The Tuberculosis Necrotizing Toxin
M. Niederweis
Mycobacterium tuberculosis (Mtb) induces necrosis of infected cells to evade immune responses.
The Mtb protein CpnT consists of an N-terminal channel domain that is used for uptake of nutrients
across the outer membrane and a secreted C-terminal domain. This C-terminal domain causes
necrotic cell death in eukaryotic cells and was named Tuberculosis Necrotizing Toxin (TNT).
Infection experiments revealed that TNT gains access to the macrophage cytosol and constitutes
the main cytotoxicity factor of Mtb in macrophages. The purified TNT protein hydrolyzes the
essential coenzyme NAD+. A non-catalytic TNT mutant showed no cytotoxicity in macrophages or
in zebrafish zygotes, thus demonstrating that the NAD+ glycohydrolase activity is required for TNTinduced cell death. The crystal structure of TNT revealed a new NAD+ glycohydrolase fold. TNT
homologs of previously unknown functions are found in more than 300 bacterial and fungal species.
Thus, TNT represents the founding member of a toxin family widespread in pathogenic
microorganisms.
36
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
17
Mycobacterial Biology & Physiology II
20/09/2016
Stress resistance, cell division and in vivo persistence
S. Ehrt, R. Wang, H. Botella, K. Kreutzfeldt
Department of Microbiology & Immunology, Weill Cornell Medicine, New York, United States
Among the factors that contribute to the success of M. tuberculosis as a pathogen is its ability to
withstand potentially bactericidal host defenses and to resist elimination by an activated immune
system. Acidification of the macrophage phagosome represents one such antimycobacterial
defense mechanism. We identified acid hyper-susceptible M. tuberculosis mutants and report their
phenotypic and mechanistic characterization that highlights a link between stress resistance, cell
division and pathogenesis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
37
18
Mycobacterial Biology & Physiology II
20/09/2016
Inherited cell-surface wave-troughs mark future division sites in mycobacteria
H.A. Eskandarian
Global Health Institute, UPKIN, École Polytechnique Fédérale de Lausanne, Lausanne,
Switzerland
Cell division is tightly controlled in space and time in order to maintain cell size and ploidy within
narrow bounds. In bacteria, the canonical Minicell (Min) and nucleoid occlusion (Noc) systems
together ensure that division is restricted to midcell after completion of chromosome segregation. It
is unknown how division site selection is controlled in bacteria that lack homologs of the Min and
Noc proteins, including mycobacteria responsible for tuberculosis and other chronic infections.
We use correlated time-lapse optical and atomic force microscopy to demonstrate that
morphological landmarks (waveform troughs) on the undulating surface of mycobacterial cells
correspond to future sites of FtsZ contractile ring formation and cell division. The manifestation of
these features are only visible by long-term time-lapse AFM, for which we have developed to our
knowledge the first system capable of continuous imaging for up to one week. Newborn cells inherit
wave-troughs from the (grand) mother cell and ultimately divide at the center-most wave-trough,
making these morphological features the earliest known landmark of future division sites. Wavetroughs are established at subpolar regions, as cell material in mycobacteria is added at the poles.
Off-center wave-troughs are shifted towards the cell center by the successive addition of new wavetroughs. In cells lacking the chromosome partitioning (Par) system, missegregation of chromosomes
is accompanied by asymmetric cell division at off-center wave-troughs, resulting in the formation of
anucleate cells. Our observations suggest a bipartite model of division site selection, in which
inherited cell-surface wave-troughs are “licensed” sites for cell division and symmetrically
segregated chromosomes suppress division at off-center wave-troughs.
Our study focuses our search for a molecular mechanism determining division-site selection in
mycobacteria to a point much earlier in time and spatially far from midcell.
38
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
19
Mycobacterial Biology & Physiology II
20/09/2016
The inosine monophosphate dehydrogenase, GuaB2, is a vulnerable new bactericidal drug
target for tuberculosis
V. Singh4, S. Donini3, A. Pacitto6, C. Sala5, R. Hartkoorn5, N. Dhar5, G. Keri2, D. Ascher6,
G. Mondesert1, A. Vocat5, A. Lupien5, R. Sommer5, H. Vermet1, S. Lagrange1, J. Buechler7,
D. Warner4, J. Mckinney5, J. Pato2, S. Cole5, T. Blundell6, M. Rizzi3, V. Mizrahi4
1
Sanofi-Aventis Research & Development, Marcy L'etoile, France 2Vichem Chemie, Budapest,
Hungary 3University of Piemonte Orientale, Novara, Italy 4Institute of Infectious Disease &
Molecular Medicine, University of Cape Town, Cape Town, South Africa 5Ecole Polytechnique
Fédérale de Lausanne, Lausanne, Switzerland 6University of Cambridge, Cambridge, United
Kingdom 7Alere, San Diego, United States
VCC234718, a molecule with growth inhibitory activity against Mycobacterium tuberculosis (Mtb),
was identified by phenotypic screening of a 15,344-compound library. Sequencing of a VCC234718resistant mutant identified a Tyr487Cys substitution in the inosine monophosphate dehydrogenase,
GuaB2, which was subsequently confirmed to be the primary target of VCC234718 in Mtb.
VCC234718 inhibits Mtb GuaB2 with a Ki value of 100 M and is uncompetitive with respect to IMP
and NAD+. This compound binds at the NAD+ site, after IMP has bound, and makes direct
interactions with IMP; therefore, the inhibitor is uncompetitive. VCC234718 forms strong pi
interactions with the Tyr487 residue side chain from the adjacent protomer in the tetramer,
explaining the resistance-conferring mutation. In addition to sensitizing Mtb to VCC2344718,
depletion of GuaB2 was bactericidal in Mtb in vitro and in macrophages. When supplied at a high
concentration (≥125 µM), guanine alleviated the toxicity of VCC234718 treatment or GuaB2
depletion via purine salvage. However, transcriptional silencing of guaB2 prevented Mtb from
establishing an infection in mice, confirming that Mtb has limited access to guanine in this animal
model. Together, these data provide compelling validation of GuaB2 as a new TB drug target.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
39
20
Mycobacterial Biology & Physiology II
20/09/2016
Functional, regulatory, and evolutionary aspects of the phage-shock-protein response of
Mycobacterium tuberculosis
P. Datta2, J. Ravi2, A. Barton2, R. Khan2, R. Manganelli1, M. Gennaro2
1
University of Padova, Padova, Italy 2PHRI, Rutgers University, Newark, United States
The phage shock protein (Psp) response, which is enacted by a multi-gene system extensively
studied in Gram-negative organisms, protects bacteria from envelope stress through a cascade of
protein interactions that stabilize the cell membrane. In Enterobacteria, the Psp response
participates in membrane polarization maintenance, divalent metal transport, and bacterial
virulence. A key component of the Psp response is PspA, a protein that stabilizes the inner
membrane when the cell envelope structure/composition is altered. PspA orthologs are widely
distributed in eubacteria, archea, cyanobacteria, and higher plants. In Gram-positive bacteria, the
Psp system has been studied in Bacillus subtilis (where it is called Lia) and, more recently,
discovered in Mycobacterium tuberculosis. In these two organisms, the PspA ortholog is associated
with gene modules that differ from each other and from the E. coli paradigm. We will present
functional and regulatory characteristics of the Psp system of M. tuberculosis, an envelope-stressresponsive four-gene operon that includes
clgR (transcriptional regulator), pspA, a third
gene encoding an integral membrane protein,
and
a
fourth
gene
of
unknown
function. Moreover, we will compare and
contrast regulatory and functional properties
and gene-expression dynamics observed in
M. tuberculosis with those of the E. coli and
B. subtilis systems. We will also review
conservation and genomic context of the Psp
system in Firmicutes and Actinobacteria, and
contrast the four-gene operon structure
uniquely found in tuberculous mycobacteria
with the gene configuration identified in nontuberculous
mycobacteria
and
other
Actinobacteria. Since Psp functions have
been
linked
to
bacterial
virulence,
susceptibility
to
membrane-perturbing
antibiotics and associated phenotypes such
as biofilm formation, understanding the Psp
system of M. tuberculosis might help identify
novel targets for antimicrobial therapeutics.
40
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
21
Mycobacterial Biology & Physiology II
20/09/2016
Playing nice together - making cell walls in mycobacteria
E. Rubin
Immunology and Infectious Diseases Department, Harvard TH Chan Scholl of Public Health,
Boston, United States
Biosynthesis of the mycobacterial cell wall requires the coordination of a complex network of
enzymes that both synthesize and degrade different components. Many of these enzymes appear
to have redundant functions. However, while some overlap, many have been adapted to have
unique functions. Some only interact with specific partners while others have evolved to fulfill
completely new roles. This complexity offers mycobacteria new opportunities for regulation.
Conversely, however, it also represents a set of vulnerabilities that can be exploited therapeutically,
both new targets and new chances to alter killing by existing drugs.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
41
22
Biomarkers, Profiling and Beyond
21/09/2016
Human CD8+ T-cells recognizing peptides from Mycobacterium tuberculosis presented by
HLA-E have an unorthodox Th2-like, multifunctional, Mtb-inhibitory phenotype and
represent a novel human T-cell subset
T. Ottenhoff2, K.E. Meijgaarden2, M.C. Haks2, N. Caccamo1, F. Dieli1, S.A. Joosten2
1
Central Laboratory for Advanced Diagnostic and Biomedical Research (CLADIBIOR),
Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di Palermo, Palermo
90129, Italy 2Leiden University Medical Center, Leiden, The Netherlands
Mycobacterial antigens are not only presented to T-cells by classical HLA-class Ia and HLA-class II
molecules, but also through alternative antigen presentation molecules such as CD1a/b/c, MR1 and
HLA-E. We recently described mycobacterial peptides that are presented in HLA-E and recognized
by CD8+ T-cells. Using T-cell cloning, phenotyping, microbiological, functional and RNA-expression
analyses, we found that these T-cells can exert cytolytic or suppressive functions, inhibit
mycobacterial growth, yet express GATA3, produce Th2 cytokines (IL-4,-5,-10,-13) and activate Bcells via IL-4. In TB patients, Mtb specific cells were detectable by peptide-HLA-E tetramers, and IL4 and IL-13 were produced following peptide stimulation. The frequencies were highest in untreated
TB patients and declined following successful treatment. These results identify a novel human T-cell
subset with an unorthodox, multifunctional Th2 like phenotype and cytolytic or regulatory capacities,
which is involved in the human immune response to mycobacteria and demonstrable in active TB
patients’ blood. The results challenge the current dogma that only Th1 cells are able to inhibit Mtb
growth and clearly show that Th2 like cells can strongly inhibit outgrowth of Mtb from human
macrophages. These insights significantly expand our understanding of the immune response in
infectious disease.
42
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
23
Biomarkers, Profiling and Beyond
21/09/2016
Profiling persistent tubercule bacilli from patient sputa during therapy predicts early drug
efficacy
I. Honeyborne5, T. Mchugh5, I. Kuittinen1, A. Cichonska1, D. Evangelopoulos5, K. Ronacher2,
P. Van Helden2, S. Gillespie7, D. Fernandez-Reyes6, G. Walzl2, J. Rousu1, P. Butcher4, S. Waddell3
1
Helsinki Institute for Information Technology, Aalto University, Espoo, Finland 2Department of
Science and Technology, Stellenbosch University, Stellenbosch, South Africa 3Brighton and
Sussex Medical School, University of Sussex, Brighton 4Institute for Infection and Immunity, St
George's University of London 5Centre for Clinical Microbiology 6Department of Computer Science,
University College London, London 7Medical and Biological Sciences Building, University of St
Andrews, St Andrews, United Kingdom
Background: New treatment options are needed to maintain and improve therapy for tuberculosis,
which caused the death of 1.5 million people in 2013 despite potential for an 86% treatment
success rate. A greater understanding of Mycobacterium tuberculosis (M.tb) bacilli that persist
through drug therapy will aid drug development programs. Predictive biomarkers for treatment
efficacy are also a research priority.
Methods and Results: Genome-wide transcriptional profiling was used to map the mRNA
signatures of M.tb from the sputa of 15 patients before and 3, 7 and 14 days after the start of
standard regimen drug treatment. The mRNA profiles of bacilli through the first two weeks of
therapy reflected drug activity at 3 days with transcriptional signatures at days 7 and 14 consistent
with reduced M.tb metabolic activity similar to the profile of pre-chemotherapy bacilli. These results
suggest that a pre-existing drug-tolerant M.tb population dominates sputum before and after early
drug treatment, and that the mRNA signature at day 3 marks the killing of a drug-sensitive subpopulation of bacilli. Modelling patient indices of disease severity with bacterial gene expression
patterns demonstrated that both microbiological and clinical parameters were reflected in the
divergent M.tb responses; evidence that factors such as bacterial load and disease pathology
influence the host-pathogen interplay and the phenotypic state of bacilli. Transcriptional signatures
were also defined that predicted measures of early treatment success (rate of decline in bacterial
load over three days, TB test positivity at 2 months, and bacterial load at 2 months).
Conclusions: This study defines the transcriptional signature of M.tb bacilli that have been
expectorated in sputum after two weeks of drug therapy, characterizing the phenotypic state of
bacilli that persist through treatment. We demonstrate that variability in clinical manifestations of
disease are detectable in bacterial sputa signatures, and that the changing M.tb mRNA profiles 0-2
weeks into chemotherapy predict the efficacy of treatment 6 weeks later. These observations
advocate assaying dynamic bacterial phenotypes through drug therapy as biomarkers for treatment
success.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
43
24
Biomarkers, Profiling and Beyond
21/09/2016
Understanding and Intervening in HIV-1 associated Tuberculosis
R. Wilkinson
University of Cape Town, Cape Town, South Africa Imperial College London The Francis Crick
Institute, London, United Kingdom
The commonest opportunistic infection worldwide that occurs in HIV-1 infected persons is
tuberculosis (TB). HIV-1 co-infection predisposes to both infection by, and reactivation of TB, and
modifies its natural history and clinical presentation. An increase in extrapulmonary disease is wellrecognized, and early or subclinical TB disease characterised by very few or no symptoms is also
common. Immunodiagnostic methods to ascertain TB sensitisation in HIV-1 infected persons are
compromised in sensitivity.
The obvious immune defect caused by HIV-1 is a progressive reduction in CD4 T cell numbers that
correlates with both increasing risk of TB and the likelihood of extrapulmonary dissemination.
However, and unlike most opportunistic conditions in HIV-1 infected persons, the risk of TB is
increased from the time of acquisition of HIV-1 before CD4 deficiency is profound. This raises the
question of whether there is additional qualitative defect in CD4 function.
Antiretroviral therapy (ART) reduces susceptibility to TB in HIV-1 infected persons via viral
suppression thus allowing partial immune restoration. Work has shown ART is associated with
expansion of both terminally differentiated and effector memory M. tb. antigen-specific CD4.
Conversely the occurrence of ART-induced inflammatory reactions in the presence of a high antigen
load in active tuberculosis (TB-IRIS) brings into focus the importance of pathological immunity in TB
and how poorly it is understood in humans. Recent interest has been in interaction between
reconstituting innate and acquired responses in the induction of TB-IRIS. There is evidence of
myeloid activation, in particular a signature of inflammasome activation with downstream
inflammatory cytokine elevation.
This presentation will review recent work in these areas and the translational consequences of
those findings.
44
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
25
Biomarkers, Profiling and Beyond
21/09/2016
Condensation of DNA as a generic stress response in Mycobacterium tuberculosis:
apoptosis in bacteria?
N. Van Der Wel1, E. Scutigliani1, D. Picavet1, E. Scholl1, A. Den Hertog2, H. Van Veen1
1
Electron Microscopy Center Amsterdam, Cell Biology and Histology, AMC 2Biomedical research,
KIT, Amsterdam, The Netherlands
Multidrug-resistant tuberculosis is a grave threat to public health worldwide. Even in Europe, about
10% of the Mycobacterium tuberculosis cases are confronted with resistance. Recently we have
discovered that M. tuberculosis condenses its DNA in response to antibiotic stress. Condensation is
visualized using high resolution fluorescence and electron microscopy (Figure 1). DNAcondensation of is an established mechanism to survive antibiotic stress, and in developing
multidrug resistance. Here, we demonstrate that condensation appears already 1 hour after
antibiotic stress and does not affect the viability of M. tuberculosis. Interestingly patient derived,
multidrug-resistant M. tuberculosis strains are more capable of condensing their DNA than drugsusceptible strains. These results imply that by condensing their DNA, mycobacteria can avoid the
effects of antibiotics, which facilitates resistance development. Our study provides the first principal
evidence that DNA-condensation is a reversible, generic stress response of viable M. tuberculosis,
and understanding this mechanism could be crucial for the development of new therapeutic
approaches against latent and multidrug resistant TB.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
45
26
Biomarkers, Profiling and Beyond
21/09/2016
TB Subunit Vaccine Immunity Expressed in The Lung
P.L. Andersen
Statens Serum Institut, Copenhagen, Denmark
The capacity of CD4 T-cells to protect against Mycobacterium tuberculosis (Mtb) is governed by
their ability to localize to the lung site of infection. The subunit vaccine H56/CAF01 confer durable
protection and elicits polyfunctional CD4 T-cells that are preferentially localized to the lung
parenchyma. These lung-resident T-cells have an intermediate state of Th1 differentiation and are
also found in the lung vasculature and peripheral circulation of vaccinated animals, but not controls.
This population efficiently trafficks into the Mtb-infected lung parenchyma after adoptive transfer.
Thus, durable immunity elicited by H56/CAF01 vaccination is associated with the maintenance of
circulating less differentiated CD4 T-cells that selectively home to the lung.
I will discuss these findings in relation to recent data from the analysis of the immune profile
obtained with the H56 subunit vaccine in ongoing clinical trials in QFT+ and QFT-individuals.
46
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
27
Whole cell vacines and beyond
21/09/2016
VPM1002 as a TB prime vaccine, but also as post-exposure vaccine on the horizon
L. Grode, M.Cotton, A. Hesseling, J. Knaul, S.H.E Kaufmann and B.Eisele
Business Development, Vakzine Projekt Management GmbH, Honnover, Germany
VPM1002 is a live recombinant vaccine against tuberculosis (TB). As BCG is not sufficiently
effective to stop the spread of TB, two modifications have been implemented in VPM1002
(rBCG∆ureC::Hly+) to improve its immunogenicity. Two phase I studies in healthy adult volunteers
and one phase IIa study in healthy newborn infants were performed. All clinical Phases were
randomized, controlled studies which evaluated safety and immunogenicity of VPM1002 in
comparison with BCG, including multiparameter flow cytometry to characterize the quality of the T
cell response following immunization. Safety and tolerability results from all clinical trials showed no
serious adverse reactions after VPM1002 vaccination. VPM1002 is currently in a Phase II doubleblinded clinical trial in newborn infants and a Phase III clinical trial in adults evaluating VPM1002 as
a TB post-exposure vaccine will commence by the end of this year in India.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
47
28
Whole cell vacines and beyond
21/09/2016
Mycobacterium bovis BCG Δzmp1 – a promising, novel live vaccine candidate
P. Sander
Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
Immunization with the live vaccine strain Mycobacterium bovis Bacille Calmette-Guérin (BCG)
confers significant but still incomplete protection against tuberculosis (TB). We hypothesized that
interference with pathogen-mediated subversion mechanisms of the host’s immune response may
provide a rationale for TB vaccine development and the improvement of BCG. Zmp1 (Rv0198c) is a
mycobacterial zinc metallopeptidase which shows a high degree of structural and biochemical
conservation to human proteases Neprilysin and ECE-1. Genetic inactivation of zmp1 interferes with
mycobacterial induced phagosome maturation arrest, induces innate immune mechanisms upon
mycobacterial infection of macrophages and attenuates Mycobacterium tuberculosis. Relief from
Zmp1-mediated phagosome maturation arrest facilitates presentation and enhances
immunogenicity of mycobacterial antigens in in vitro and in vivo BCG infection models. In a guinea
model, vaccination with BCG Δzmp1 strain confers improved protection over BCG against a low
dose aerosol challenge with M. tuberculosis. Furthermore, BCG Δzmp1 deletion mutants exhibit
decreased virulence in a SCID mouse model, as compared to parental strains. The improved safety
profile and the enhanced protective efficacy indicate that BCG Δzmp1 is a promising, novel live
vaccine candidate.
48
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
29
Whole cell vacines and beyond
21/09/2016
ESX-1 type VII secretion, cytosolic pattern recognition and consequences for vaccine
design
R. Brosch
Institut Pasteur, Paris, France
Recent insights into the implication of ESX-1 type VII secretion in cytosolic pattern recognition of
Mycobacterium tuberculosis, the etiologic agent of human tuberculosis and its bacterial products,
inside the cytosol of host phagocytes, open novel prospects for rational design of whole cell
vaccines. Mycobacterium bovis BCG (Bacille Calmette-Guérin) and Mycobacterium microti, the only
two vaccines used in large-scale vaccination programs against tuberculosis, are both ESX-1
deletion mutants that are unable to induce ESX-1-mediated vacuole-to-cytosol communication and
downstream innate immune signaling. After previous ESX-1 complementation of BCG with the ESX1 system of M. tuberculosis has shown an increase of protective efficacy but also an increase in
virulence, here we have used the ESX-1 from Mycobacterium marinum to complement BCG. We
found that heterologous expression of the ESX-1 from M. marinum, a class II organism, in BCG, led
to a recombinant strain (BCG::ESX-1Mmar) that was able to induce phagosomal rupture and
consequent innate immune signaling by activating the cGas/STING/TBK1/type I interferon axis and
enhancing inflammasome activity. Consistently, the BCG::ESX-1Mmar induces higher proportions of
CD8+ T-cell effectors against mycobacterial antigens shared with BCG and generates polyfunctional
CD4+ Th1 cells specific to ESX-1 antigens, ultimately displaying superior protective efficacy against
a M. tuberculosis challenge, relative to parental BCG in a mouse model. This strain can be seen as
an example of a promising rBCG candidate that combines low virulence with enhanced protection
abilities.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
49
30
Whole cell vacines and beyond
21/09/2016
MTBVAC: from Discovery to Evaluation in High-Burden Countries
B. Gicquel1, J. Gonzalo-Asensio2, N. Aguilo2, D. Marinova2, E. Broset2, S. Alvarez2, I. Perez2,
C. Martin2
1
Institut Pasteur, Paris, France 2University of Zaragoza, Zaragoza, Spain
The only available vaccine in use against TB in humans today is BCG, a live attenuated vaccine
derived from the bovine pathogen Mycobacterium bovis. BCG presents variable protection against
pulmonary forms of TB. Genomic comparative studies have shown loss of a number of major
mycobacterial antigens and about 23% of M. tuberculosis-specific human T-cell epitopes in BCG
relative to the human pathogen Mycobacterium tuberculosis. MTBVAC is a new live tuberculosis
vaccine based on a genetically attenuated phoP-fadD26-deletion mutant of M. tuberculosis. The
presence of stable deletion mutations in two independent virulence genes abrogates the risk of
reversion to virulence. MTBVAC has shown a comparable or superior safety and immunogenicity
profile to BCG in different preclinical animal models including new-born mice model (Arbues et al
Vaccine 2013 and Broset el al mBio 2015, Aguilo et al Tuberculosis 2016).
A first-in-human MTBVAC clinical trial was recently completed successfully in healthy adults in
Lausanne, Switzerland sponsored by Biofabri (NCT02013245) (Spertini et al LRM 2015). In this
trial, when MTBVAC was given at the same dose as BCG (5x10⁵ CFU), there were more
responders in the MTBVAC group than in the BCG group, with a greater frequency of polyfunctional
CD4+ central memory T cells. MTBVAC is the first live-attenuated M. tuberculosis vaccine to enter
clinical trials and to date has shown a comparable safety profile to BCG. A notable finding in the first
trial was the absence of ESAT-6 and CFP-10-specific T cell responses at the end of the study,
suggesting that interferon-g release assays (IGRAs) could be utilized as study endpoints in future
efficacy trials to test efficacy against M. tuberculosis infection. The immunogenicity data show that
MTBVAC is at least as immunogenic as BCG. Altogether these data supported the advanced
clinical development in high-burden countries where TB is endemic. A dose-escalation safety and
Immunogenicity study to compare MTBVAC to BCG in newborns with a safety arm in adults is
currently ongoing in South Africa sponsored by Biofabri (NCT02729571). MTBVAC is developed
with the goal to provide improved efficacy over BCG for use in new-borns, adolescents and adults
as a preventive strategy against tuberculosis in high-burden countries (Arregui et al PeerJ 2016).
Funding: Biofabri, TuBerculosis Vaccine Initiative (TBVI). The Norwegian Agency for Development
Cooperation (NORAD). BIO2014 5258P and TBVAC2020 643381 Spanish and European grants.
References:
• Aguilo N, Uranga S, Marinova D et al MTBVAC vaccine is safe, immunogenic and confers
protective efficacy against Mycobacterium tuberculosis in newborn mice. Tuberculosis 2016;
96:71-4.
• Arbues A, Aguilo JI, Gonzalo-Asensio J, et al. Construction, characterization and preclinical
evaluation of MTBVAC, the first live-attenuated M. tuberculosis-based vaccine to enter clinical
trials. Vaccine 2013; 31(42):4867-73
• Arregui S, Sanz J, Marinova D, et al. On the impact of masking and blocking hypotheses for
measuring the efficacy of new tuberculosis vaccines. PeerJ. 2016 Feb 11;4:e1513.
• Broset E, Martín C, Gonzalo-Asensio J.Evolutionary landscape of the Mycobacterium
tuberculosis complex from the viewpoint of PhoPR: implications for virulence regulation and
application to vaccine development. MBio. 2015 Oct 20;6(5):e01289-15.
• Spertini F, Audran R, Chakour R, et al. Safety of human immunisation with a live-attenuated
Mycobacterium tuberculosis vaccine: a randomised, double-blind, controlled phase I trial. The
Lancet Respiratory Medicine 2015; 3:953-62.
50
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
31
Biochemistry & Chemical biology
21/09/2016
The dynamics of TB lesions revealed by PET/CT scanning using [F-18] trehalose
C. Barry
Tuberculosis Research Section - LCID, NIH, Bethesda, United States
PET/CT scanning is receiving increasing attention as a useful means of evaluating TB disease
status and response to therapy however current scans are limited in that the most commonly used
probe - 2-[18F]-fluorodeoxyglucose (FDG) - reports only on host inflammatory processes and not on
bacterial viability. To circumvent this problem we have synthesized trehalose derivatives containing
[18F] and applied these to imaging TB-infected rabbits and non-human primates. Serially scanning
with FDG followed by FDT (fluorodeoxytrehalose) revealed that sites of rapid trehalose
incorporation and sites of high levels of inflammation are largely non-overlapping within individual
animals. These results suggest that local areas with high levels of inflammation effectively suppress
rapid bacterial growth while new lesions rapidly develop that are not subject to inflammatory
pressure.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
51
32
Biochemistry & Chemical biology
21/09/2016
Flipping trehalose monomycolate across the inner membrane of mycobacteria
Z. Xu2, G. Poce1, S.S. Chng2
1
Sapienza University of Rome, Rome, Italy 2National University of Singapore, Singapore
The mycobacterial outer membrane (OM) is characterized by the presence of mycolic acids, which
are C60-C90 long, branched chain fatty acids, either existing in the forms of trehalose mono- and
diesters, or covalently attached to peptidoglycan via arabinogalactan polysaccharides. Being the
major component in the OM, mycolic acids make this bilayer extremely hydrophobic and render the
membrane impervious to many antibiotics. Mycolic acids are synthesized in the cell as trehalose
monomycolates (TMMs), and have to be translocated across the inner membrane (IM) and the
aqueous periplasm before being functionalized onto the cell wall. While the biosynthetic pathway of
TMM and the final steps of assembly at the OM have been well characterized, how TMM is
transported across the IM and periplasm are still not clear. Recently, an essential IM protein MmpL3
has been implicated in TMM transport across the cell envelope and is believed to be inhibited by
multiple pharmacophores. Here, we present direct biochemical evidence for the function of MmpL3
in flipping TMM across the IM. Furthermore, we demonstrate that a couple of potential MmpL3
inhibitors tested directly target the TMM flippase. Our work provides fundamental insights into
mycolic acid transport and validates MmpL3 as a viable target for the development of new
antibiotics against mycobacterial infections.
52
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
33
Biochemistry & Chemical biology
21/09/2016
Mechanisms of high fidelity DNA replication in Mycobacterium tuberculosis
J. Rock2, U. Lang1, S. Fortune2, M. Lamers1
1
Structural Studies, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom 2Harvard
School of Public Health, Boston, United States
Antibiotic resistance in Mycobacterium tuberculosis (Mtb) is increasing at an alarming rate.
Resistance in Mtb is predominantly caused by point mutations, yet the mechanisms that govern the
mutation rate in this bacterium are poorly understood. Importantly, mycobacteria lack DNA
mismatch repair, an evolutionary conserved mechanism that removes replication errors from the
DNA. However, they do not show increased mutation rates when compared to E. coli or S.
typhimurium, suggesting that in mycobacteria, the DNA replication machinery itself may be capable
of unusually high fidelity DNA synthesis. To understand how Mtb controls its mutation rate, we have
characterized the replicative DNA polymerase from Mtb, DnaE1. We show that high fidelity DNA
replication relies on a non-canonical exonuclease, the PHP domain, that is distinct from the
replicative exonuclease in E. coli as well as the eukaryotic exonucleases. We further show that
inhibition of the PHP exonuclease renders mycobacteria hypersensitive to nucleotide analogs that
are used in antiviral therapy, indicating that the replicative DNA polymerase from Mtb is an attractive
target for novel antimicrobial drugs.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
53
34
Biochemistry & Chemical biology
21/09/2016
Structural basis of phosphatidylinositol mannosides biosynthesis in mycobacteria
M. Guerin
CICbioGUNE, Spain
Many cellular reactions involve both hydrophobic and hydrophilic molecules that reside within the
chemically distinct environments defined by the phospholipid-based membranes and the aqueous
lumens of cytoplasm and organelles. Enzymes performing this type of reaction are required to
access a lipophilic substrate located in the membranes and to catalyze its reaction with a polar,
water-soluble compound.[1] Here we focus on two enzymes involved in the early steps of the
phosphatidylinositol mannosides (PIMs) biosynthetic pathway, unique glycolipids found in abundant
quantities in the inner and outer membranes of the cell envelope of all Mycobacterium species.
They are based on a phosphatidylinositol lipid anchor carrying one to six mannose residues and up
to four acyl chains. PIMs are considered not only essential structural components of the cell
envelope but also the precursors of the lipoglycans lipomannan and lipoarabinomannan, important
molecules implicated in host-pathogen interactions in the course of tuberculosis. Of particular
relevance, we demonstrate the occurrence of a conformational switch during the catalytic cycle of
the retaining glycosyltransferase PimA, the enzyme that start the pathway, involving both β-strand–
to–α-helix and α-helix–to–β-strand transitions. [2] These structural changes seem to modulate
catalysis and are promoted by interactions of the protein with anionic phospholipids in the
membrane surface. Although scant structural information is currently available on protein catalysis
at the lipid-water interface, our studies demonstrate that protein-membrane interactions might entail
unanticipated structural changes in otherwise well conserved protein architectures, and suggests
that similar changes may also play a functional role in other membrane enzymes. Finally, we report
the crystal structures of PatA, an essential membrane associated acyltransferase that transfers a
palmitoyl moiety from palmitoyl–CoA to the 6-position of the mannose ring added by PimA, in the
presence of its naturally occurring acyl donor palmitate and a nonhydrolyzable palmitoyl–CoA
analog. The structures reveal an α/β architecture, with the acyl chain deeply buried into a
hydrophobic pocket that runs perpendicular to a long groove where the active site is located.
Enzyme catalysis is mediated by an unprecedented charge relay system, which markedly diverges
from the canonical HX4D motif. Our studies establish the mechanistic basis of substrate/membrane
recognition and catalysis for an important family of acyltransferases, providing exciting possibilities
for inhibitor design.
References:
1. Forneris, F.; Mattevi, A. Science 2008, 321, 213-216.
2. Giganti et al., Nat. Chem. Biol. 2015, 11, 16-18. Highlighted in the News and Views
Section: Brodhun F, Tittmann K. Nat. Chem. Biol. 2015, 11, 102-103.
3. Albesa-Jove et al., Angew. Chem. Int. Ed. Engl. 2015, 54, 9898-9902.
4. Albesa-Jove et al., Nat. Commun. 2016, 7, 10906.
54
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
35
Biochemistry & Chemical biology and beyond
21/09/2016
Amino acid sensing by protein kinase G in metabolic regulation and virulence
H. O'Hare
Dept of Infection, Immunity and Inflammation, University of Leicester, United Kingdom
Protein kinase G (PknG) has previously been implicated in metabolic regulation and virulence of
Mycobacterium tuberculosis. This multi-domain kinase has been the subject of diverse studies.
Amongst the various putative functions, evidence is accumulating that the link to metabolic
regulation is likely to be through its substrate GarA. Together these proteins form a molecular switch
to control the tricarboxlic acid cycle and glutamate synthesis/degradation. Phosphorylation of GarA
by PknG inactivates it, thus genetic disruption of garA or pknG should have opposing effects on
metabolism, while GarA variants lacking phosphorylation sites should mimic the effects of pknG
deletion. Both the kinase and substrate (with intact phosphorylation sites) were required in infection
models, highlighting the importance of responsiveness of this regulatory system, but the garA
mutant was markedly more attenuated than pknG deficient M. tuberculosis. Combined with the
growth defects of these strains, this hints that M. tuberculosis may experience glutamate starvation
in vivo. Glutamate itself was a stimulus for rapid phosphorylation of GarA in bacterial cells. We
propose that M. tuberculosis and other actinobacteria may sense external amino acids via a novel
complex of PknG with transmembrane GlnX and lipoprotein GlnH.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
55
36
Biochemistry & Chemical biology and beyond
21/09/2016
Targeting topology modulators and topoisomerases of Mycobacterium tuberculosis
V. Nagaraja
Jawaharlal Nehru Centre for Advanced Scientific research & Department Microbiology and Cell
Biology, Indian Institute of Science, Bangalore, India
DNA topoisomerases catalyze the topological reactions that are essential for cell survival.
Contrasting activities of different kinds of topoisomerases ensure to regulate the supercoiling,
relaxation and other topological reactions. Most mycobacteria possess only one topoisomerase I
and a DNA gyrase as representatives of type I and II group. Our earlier studies have revealed that
mycobacterial topoisomerase I and DNA gyrase are distinct from other prokaryotic and eukaryotic
topoisomerases in many characteristics. Topoisomerase I is site specific and a unique carboxy
terminal domain is involved in strand passage reaction. Mycobacterial gyrase also exhibits distinct
features and is a strong decatenase unlike the E.coli enzyme.
In order to maintain topological homeostasis of the genome, topoisomerases catalyse DNA
cleavage, strand passage and rejoining of the ends. Thus, although they are essential housekeeping enzymes, they are the most vulnerable targets; arrest of the reaction after the first transesterification step leads to breaks in DNA and cell death. We have targeted both DNA gyrase and
topoisomerase1 from mycobacteria. The latter, although essential, has no inhibitors described so
far. We have now characterized the first set of small molecule inhibitors that inhibit the enzyme with
bactericidal effect.
In addition to topoisomerases, nucleoid associated proteins(NAPs), are the key topology modulators
responsible for DNA compaction and genome organization. We have targeted HU, an essential
protein from M. tuberculosis to develop small molecule inhibitors by structure based design. Overexpression of HU leads to alteration in the nucleoid architecture. The crystal structure of the Nterminal half of HU reveals a cleft that accommodates duplex DNA. Based on the structure, we have
designed inhibitors which bind to the protein and affect its interaction with DNA, de-compact the
nucleoid and inhibit cell growth. Chemical probing using the inhibitors reveal the importance of HU
regulon in M.tuberculosis.
56
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
37
Biochemistry & Chemical biology and beyond
21/09/2016
In Search of Drugs to Shorten Treatment of TB
C. Nathan, B. Gold, G. Lin, J. Vaubourgeix, T. Lupoli, K. Saito, S. Somersan-Karakaya, T. Warrier
Depts. of Microbiology & Immunology and Medicine, Weill Cornell Medicine, New York City, New
York, United States
Pyrazinamide’s ability to shorten treatment of TB while being active in vitro largely against nonreplicating (NR) Mycobacterium tuberculosis (Mtb) points to the clinical value of finding new drugs
active against NR Mtb. Yet, the prolonged course even of PZA-containing regimens suggests poor
activity against some other subpopulation of Mtb. We approach these challenges by distinguishing
three forms of phenotypic tolerance1, including a form in which stressed Mtb no longer forms
colonies on agar but retains disease-causing potential: differentially detectable (DD) Mtb. We take
three routes to search for compounds active against Mtb showing phenotypic tolerance to most TB
drugs: (1) whole cell screening under NR conditions; (2) target-based screening against enzymes
that Mtb needs to survive under stresses that block replication; and (3) testing active agents from (1)
and (2) against DD Mtb in vitro. Screens (1) and (2) are directed against what was called class II
phenotypic tolerance1, now called class IIa because under those NR conditions, without drug, the
number of CFU neither increases nor decreases during the assay, in contrast to conditions that
reduce CFU but leave DD Mtb, whose phenotypic tolerance is termed class IIb. Corresponding
actives are (1) novel cephalosporins, (2) species-selective inhibitors of the Mtb proteasome and (3)
a natural product-based nitrofuranylcalanolide. We hypothesize that rapid cure of TB will require
achieving four goals in each infected physical compartment. Overcoming heritable resistance will
require drug(s) for which 1/(product of their frequencies of resistance) > number of viable Mtb.
Overcoming class I phenotypic tolerance1 (displayed by a small fraction of cells in a replicating
population) will require n drugs where n > number of mechanisms of class I phenotypic tolerance
displayed by different individual bacteria toward those drugs. Overcoming class IIa phenotypic
tolerance will require a drug that kills NR Mtb detectable as CFU. Finally, overcoming class IIb
phenotypic tolerance will require a drug that kills DD Mtb. The number of drugs required to meet all
four goals could in theory be as few as 1 but an upper bound on the number is undefinable— a
daunting prospect.
1. Nathan, C. Bacterial pathogenesis: Fresh approaches to anti-infective therapies. Science
Translational Medicine 4: 1-13, 2012
We acknowledge for support: Tri-Institutional TB Research Unit (NIH grant U19 AI1111043); Bill &
Melinda Gates Foundation TB Drug Accelerator; Tri-Institutional Therapeutics Discovery Institute;
Milstein Program in Chemical Biology and Translational Medicine.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
57
38
NTM & Emerging mycobacterial pathogens
22/09/2016
The distinct fate of smooth and rough Mycobacterium abscessus variants inside
macrophages
J.L. Herrmann
UMR1173, Université de Versailles Saint Quentin en Yvelines, Versailles, France
Mycobacterium abscessus is a pathogenic, rapidly growing mycobacterium responsible for
pulmonary and cutaneous infections in immunocompetent patients and in patients with Mendelian
disorders, such as cystic fibrosis (CF). M. abscessus is known to transition from a smooth (S)
morphotype with cell surface-associated glycopeptidolipids (GPL) to a rough (R) morphotype lacking
GPL. M. abscessus S and R variants are able to grow inside macrophages but are present in
morphologically distinct phagosomes. The S forms are found as single bacteria within phagosomes
characterized by a tightly apposed phagosomal membrane and the presence of an electron
translucent zone (ETZ) surrounding the bacilli. In contrast, infection with the R form leads to
phagosomes containing more than two bacilli, surrounded by a loose phagosomal membrane and
lacking the ETZ. In contrast to the R variant, the S variant is capable to restrict the intraphagosomal
acidification and induces less apoptosis and autophagy. Unexpectedly, the membrane of
phagosomes enclosing the S forms showed signs of alteration such as breaks or partial
degradation. Although such events were not frequently encountered, they suggest that the S form is
capable to provoke phagosome-cytosol communications. The mycobacterial constituents and
molecular mechanisms involved in the rupture, or at least alteration, of the M. abscessus Scontaining phagosome membrane remain as yet unknown. It has been shown clearly that M.
abscessus is not equipped with the ESX-1 apparatus, which mediates establishment of cytosol
contact for slow-growing pathogenic mycobacteria such as Mycobacterium tuberculosis,
Mycobacterium marinum and Mycobacterium kansasii. Ongoing work with a panel of defined
mutants, will hopefully allow us to identify the putative membrane-damaging constituents of the S
strain, which are obviously mediated by an ESX-1-independent mechanism. To summarize, it
seems to be rather difficult to strictly compare the behaviors of S and R morphotypes within in vitrogrown macrophages cultures and patients, because in patients they might occur at dedicated
stages during progression of the infection and disease. S and R variants can most likely be
regarded as two representatives of the same isolate, which can co-exist and/or evolve differently in
response to host immunity. Nonetheless, we provide compelling evidence that, at the cellular scale,
M. abscessus S imitates phenotypic traits of pathogenic SGM and that the loss of cell-wall
associated lipids, namely GPL, can result in the acquisition of an RGM intracellular behavior with a
peculiar extracellular state characterized by a very high replication capacity.
58
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
39
NTM & Emerging mycobacterial pathogens
22/09/2016
Global spread of Mycobacterium abscessus clones amongst cystic fibrosis patients
J. Bryant3-4, D. Grogono3, D. Rodriguez-Rincon3, I. Everall4, K. Brown2-3, P. Moreno1, D. Verma5,
E. Hill5, J. Drijkoningen3, C. Haworth2, S. Harris4, D. Ordway5, J. Parkhill4, R.A. Floto2-3
1
EMBL-EBI 2Cambridge Centre for Lung Infection, Papworth Hospital 3Department of Medicine,
University of Cambridge, Cambridge 4Wellcome Trust Sanger Institute, Hinxton, United
Kingdom 5Mycobacteria Research Laboratory, Colorado State University, Fort Collins, United
States
Lung infections with Mycobacterium abscessus have increased in frequency worldwide, emerging
as an important global threat to individuals with cystic fibrosis (CF) where they cause accelerated
inflammatory lung damage and death.
M. abscessus was previously thought to be independently acquired by susceptible individuals from
the environment. However, using whole genome sequencing and detailed epidemiological analysis
of a cohort of patients attending the CF centre at Papworth Hospital, we found strong evidence for
transmission between patients. This observation is supported by our subsequent whole genome
analysis of a global collection of 1,080 clinical isolates from 517 patients, showing that the majority
of infections are from densely clustered M. abscessus genotypes with low levels of diversity,
indicating a high level of human associated spread. Moreover, the phylogeny reveals the presence
of three recently emerged dominant circulating clones that have global spread. We found that these
clones are associated with worse clinical outcomes and show increased virulence in both cell-based
and mouse infection models.
Within patients we found evidence of significant genetic diversity and evolutionary adaptation
through the processes of convergent evolution and hypermutation, demonstrating the propensity of
M. abscessus to evolve from an environmental organism into a transmissible human pathogen.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
59
40
NTM & Emerging mycobacterial pathogens
22/09/2016
A multidisciplinary approach to decipher the epidemiology of Mycobacterium ulcerans
infection: where do we stand?
S. Eyangoh
Head of Mycobacteriology Service, Centre Pasteur du Cameroun, Yaounde, Cameroon
Buruli ulcer (BU) is a severe human debilitating skin and soft tissue disease caused by the
bacterium Mycobacterium ulcerans. BU has been reported in 33 countries mainly located in Central
and West Africa. Amidst the rising number of BU cases, in 1998, the World Health Organization
declared BU as an emerging neglected tropical disease. Since then, several scientific studies to
better understand BU have been conducted. Despite increase in knowledge about the disease, the
mode of disease transmission is still unclear. As a result, it has been difficult to propose and
effective prevention strategy for BU.
Here, we present the results of a multidisciplinary approach to describe the dynamics of M. ulcerans
in the aquatic environment, and to understand the links between M. ulcerans transmission, human
activities and their evolutions, in anthropologically modified environments. This approach allowed; to
obtain an array of results.
1. Age and sex were identified as risk factors for BU and population was classified in three main
groups:
Children 5-15 years (especially males)
Women 15-49 years
Adults over 60 years;
2. individual risk factor analysis (habits/ activities) showed the protection effect of bed nets, good
care of wounds and wearing shoes during activities and contact with stagnant water identified
as risk factor
3. Spatial analysis of time aggregated incidence rate of BU in Akonolinga- a BU endemic site,
from 2002–2012 identified the Nyong river as a major risk factor for BU;
4. BU incidence in Akonolinga varies significantly by season and this seasonal variation is linked
to the fluctuation of M. ulcerans occurrence in the environment. Fluctuation in the occurrence
of M. ulcerans in the environment is probably driven by the dynamics of freshwater ecosystems
of the Nyong River;
5. M. ulcerans dynamic was shown geographically, in aquatic communities and in freshwater
ecosystem;
6. Seasonal fluctuations of M. ulcerans in freshwater ecosystems;
7. Potential biotic and abiotic factors as drivers of M. ulcerans
8. The ecological traits (living on aquatic vegetation or bottom of water column and
macropredator) may predispose water bug taxa to acquire M. ulcerans from the aquatic
environment.
These results improve our understanding of the epidemiology of BU and could have direct public
health implication in improving early detection, guiding treatment decentralization and
implementation of prevention strategies.
60
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
41
NTM & Emerging mycobacterial pathogens
22/09/2016
NOD2, mycobacteria and chronic enteritis
D. Montamat-Sicotte2, J.D. Lalande2, F. Mcintosh2, M. Paquet3, L. Kreitmann2, D. Houle2,
M. Divangahi1, M. Behr2-1
1
Department of Microbiology and Immunology, McGill International TB Centre 2Research Institute
of the McGill University Health Centre 3Université de Montréal, Faculté de médecine Vétérinaire,
Montreal, Canada
Nucleotide-binding oligomerization domain-containing protein 2 (NOD2) is an intracellular pattern
recognition receptor that preferentially recognizes N-glycolyl MDP produced by mycobacteria. In
humans, loss-of-function mutations in NOD2 predispose to Crohn’s disease. We set out to study the
outcome of gastrointestinal infections as a function of NOD2, using Nod2-/- mice that phenocopy
the human mutations. To do so, we tested bacteriologic, histopathologic and immunologic outcomes
in Nod2+/+ and Nod2-/- mice following infection with Listeria monocytogenes (LIST), E. coli strain
LF82 and Mycobacterium avium paratuberculosis (MAP).
Following intrajejunal infection with 109 bacteria, LIST and E. coli resulted in acute gastroenteritis,
with Nod2-independent mortality in the first week following infection. With a lower dose of 108,
infected mice that survived acute infection cleared the pathogens by 8 weeks after infection and
there was no pathology in their small intestine at this time-point. In contrast, mice infected with 109
MAP survived the full 8 weeks of infection without gross morbidity (normal weight compared to PBS
sham infections) and no mortality. Bacteriologic analysis revealed that MAP-infected Nod2-/- mice
had greater numbers of bacteria in the small intestine, the liver and the spleen, compared to
Nod2+/+ infected controls. Histopathologic analysis revealed granulomatous inflammation in the
serosa of the ileum. Responses were quantified via FACS analysis, which revealed increased
numbers of neutrophils and monocytes in the small intestine of Nod2/- mice; by co-staining, we
determined that the Nod2-/- group had more TNFα producing neutrophils. To understand the
potential role of these cells in the host response, we conducted ex vivo studies of MAP infection.
Nod2-/- macrophages showed reduced recognition and clearance of MAP infection. Nod2-/dendritic cells showed reduced recognition of MAP infection, and when loaded with Ovalbuminexpressing MAP, reduced OVA-specific T-cell proliferation. Nod2-/- neutrophils showed reduced
recognition and clearance of MAP infection.
Together, our results show that of the three intestinal pathogens tested, only MAP resulted in a
chronic infection and produced Nod2-dependent outcomes. The recruitment of cells to the small
intestine that had impaired responses to MAP infection may contribute to a climate of chronic
inflammation in the gut that drives ongoing pathology.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
61
42
NTM & Emerging mycobacterial pathogens
22/09/2016
New Approaches to Target Mycobacterium abscessus Infections
L. Kremer
Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé (CPBS), Centre National de
la Recherche Scientifique FRE3689, Université de Montpellier, France
Mycobacterium abscessus (Mabs) is a rapid growing pathogenic species capable to induce severe
and chronic pulmonary infections and is particularly harmful for cystic fibrosis patients. Transitioning
from a smooth (S) high-glycopeptidolipid (GPL) producer to a rough (R) low-GPL producer is
associated with increased virulence, involving the formation of massive serpentine cords and
abscesses. The intrinsic resistance of Mabs to most commonly available antibiotics seriously limits
chemotherapeutic treatments. Therefore, new targets and drugs are urgently needed. Until recently,
the identification of new chemotypes and the description of their mechanism of action have been
largely hampered by our restricted knowledge of the physiopathological events characterizing Mabs
infections and by the lack of genetic tools and appropriate animal models for in vivo drug
assessments.
To better understand the natural resistance of Mabs to antiTB drugs, a deletion of MAB_4780,
encoding a putative dehydratase, was created in the R variant. Unexpectedly, the mutant exhibited
alteration of the mycolic acid composition and a pronounced defect in cording. This correlated with
an extremely attenuated phenotype in wild-type and in immunocompromised zebrafish. This
indicates that MAB_4780 is required for Mabs cording and for the successful establishment of acute
and lethal infections. Thereby, targeting MAB_4780 may represent an attractive anti-virulence
strategy to control Mabs infections. The combination of our dehydratase assay with a highresolution crystal structure of MAB_4780 opens the way to identify such specific inhibitors.
In a different approach, we demonstrated the usefulness of a phenotypic screen for bactericidal
compounds against Mabs using a previously validated library of compounds active against M.
tuberculosis. A new compound exhibiting potent activity against Mabs in vitro and in infected
macrophages and zebrafish was identified. This lead compound, PIPD1, targets the mycolic acid
transporter MAB_4508 of the MmpL family. Interestingly, multiple single mutations in MAB_4508
conferring high resistance to PIPD1 defined a potential PIPD1-binding pocket when mapped on a
MAB_4508 structural model. This emphasizes a yet unexploited structure class with promising
translational development possibilities against Mabs.
62
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
43
Mycobateria and Host Interaction/Responses
22/09/2016
C-type lectin receptor DCIR modulates immunity to tuberculosis by sustaining type I
interferon signaling in dendritic cells
A. Troegeler, D. Hudrisier, O. Neyrolles
Institute of Pharmacology & Structural Biology, CNRS-University of Toulouse, Toulouse, France
Immune response against pathogens is a tightly regulated process that must ensure microbial
control while preserving integrity of the infected organs. Tuberculosis (TB) is a paramount example
of a chronic infection in which antimicrobial immunity is protective in the vast majority of infected
individuals, but can become detrimental if not finely tuned. Here, I will discuss our recent results that
show that C-type lectin dendritic cell (DC) immunoreceptor (DCIR) is required to modulate lung
inflammation and bacterial burden in TB. In mice, we found that DCIR deficiency impairs STAT1mediated type I interferon (IFN) signaling in DCs, leading to increased production of interleukin-12
(IL-12) and increased differentiation of T helper-1 (Th1) cells during infection. As a consequence,
while DCIR-deficient animals control M. tuberculosis better than wild-type animals, they also
develop more inflammation. Altogether, our study reveals a novel pathway by which a C-type lectin
modulates the equilibrium between infection-driven inflammation and pathogen’s control by
sustaining type I IFN signaling in DCs.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
63
44
Mycobateria and Host Interaction/Responses
22/09/2016
Immuno-metabolic regulation of M. tuberculosis infection by HIF-1α and nitric oxide
J. Braverman, K. Sogi, S. Stanley
School of Public Health, Molecular and Cell Biology, University of California, Berkeley, Berkeley,
United States
IFN-γ is critical for immunity to Mycobacterium tuberculosis. We show that the transcription factor
Hypoxia Inducible Factor 1-α (HIF-1α) is a master regulator of IFN-γ dependent signaling that
promotes inducible antibacterial immunity. Mice lacking HIF-1α in the myeloid lineage are extremely
susceptible to infection with M. tuberculosis. HIF-1α is required for optimal production of a large
number of critical immunity genes and promotes the production of inflammatory cytokines,
chemokines and eicosanoids. In addition, HIF-1α mediates IFN-γ dependent metabolic transitions in
macrophages that promote control of infection by enhancing immune responses and influencing
nutrient availability to M. tuberculosis. We demonstrate that HIF-1α and inducible nitric oxide
synthase (iNOS) are linked in a positive feedback loop in which each promotes optimal expression
of the other. Thus, nitric oxide plays a major immunomodulatory role by promoting a broad array of
antibacterial defense mechanisms. Further, we provide evidence that the interconnected regulation
of HIF-1α and iNOS is important to achieve proper inflammatory balance during infection.
64
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
45
Mycobateria and Host Interaction/Responses
22/09/2016
T cells and TB - do we know what kind of T cell works best?
A.M. Cooper
Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
Mycobacterium tuberculosis (Mtb) has been evolving with its human host for many generations and
has developed the capacity to manipulate the human immune response. One working model of the
interaction between Mtb and the human host is that the bacterium induces both a strong
inflammatory and a strong acquired immune response and then actively regulates these responses
at the primary lesion site. This induction and subsequent regulation then results in an infectious
lesion in the lung while maintaining a relatively ambulatory host. CD4 T cells play a critical yet
contradictory role in this process by both controlling disseminated disease while promoting the
development of the lesion in the lung that mediates transmission. It is not surprising that our ability
to vaccinate against tuberculosis (TB) has not been totally successful as our attempts have largely
mimicked what the bacterium does in vivo. In order to overcome the current impasse in vaccine
development we need to define the factors which impact the phenotype and function of protective
CD4 T cells. Vaccination using a variety of T cell epitopes can impact the long term protetction
mediated by T cells, the nature of these T cells will be discussed.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
65
46
Mycobateria and Host Interaction/Responses
22/09/2016
Type I interferon production by B cells controls the balance between immunopathology and
pathogen burden in lungs during Mycobacterium tuberculosis infection
A. Bénard5-9, I. Sakwa10, P. Schierloh1, M. Del Carmen Sasiain1, I. Mercier5-6-7-9-4, L. Jouneau2,
P. Boudinot2, L. Tailleux3, T. Al-Saati8, R. Lang12, J. Rehwinkel15, S.H. Kaufmann11,
V. Anton-Leberre9-7-4-6, A. O'garra13-14, B. Gicquel3, S. Fillatreau10, O. Neyrolles5-9, D. Hudrisier5-9
1
IMEX-CONICET, Caba, Argentina 2Virologie et Immunologie Moléculaires, INRA, Jouy-EnJosas 3Institut Pasteur, Paris 4CNRS UMR5504 5CNRS, IPBS 6UMR792, INRA 7INSA 8INSERM,
US006 9Université de Toulouse, Toulouse, France 10DRFZ 11Max Planck Institute of Infection
Biology, Berlin 12Institute of Clinical Microbiology, Erlangen, Germany 13MRC National Institute for
Medical Research 14NHLI, Imperial College, London 15MRC Human Immunology Unit, Oxford,
United Kingdom
The type I interferon (IFN) pathway decisively influences the outcome of Mycobacterium
tuberculosis (Mtb) infection. Here, we show that B lymphocytes, which accumulate in the lungs of
Mtb-infected animals and patients, play a significant role in the type I IFN response associated with
tuberculosis (TB). Through global transcriptome analyses, we discovered that B cells were activated
by type I IFN during disease, and produced this cytokine upon direct stimulation with Mtb. The
production of type I IFN by B cells involved the dinucleotide sensor STING and the C-type lectin
Mincle, and was markedly antagonized by intrinsic MyD88 signaling. Thus, B cells from mice with a
B cell-restricted deficiency in Myd88 displayed an enhanced type I IFN production. This resulted in
reduced inflammation-driven tissue damage, while providing a favorable environment for Mtb
multiplication. B cells from the pleural fluid of TB patients similarly displayed a striking expression of
type I IFN, implicating this pathway in TB pathogenesis in human. Altogether, our findings identify
type I IFN-producing B cells as key modulators in the balance between host tissue protection and
bacterial burden during TB, and further emphasize the critical roles of cytokine production by B cells
in health and disease.
66
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
47
Mycobateria and Host Interaction/Responses
22/09/2016
Anti-microbial effector pathways used by T cells to restrict intracellular M. tuberculosis
replication
S. Behar
Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester,
United States
Identifying anti-microbial effector pathways used by T cells to restrict intracellular M. tuberculosis
replication could help improve vaccine efforts and offer new immunotherapy targets. Apart from
IFNγ and TNF, there are several other T cell functions that have been described to contribute to an
antimicrobial response. Early during the immune response, IL-17 enhances control of M.
tuberculosis, in part by promoting granuloma formation. CD8+ T cell cytolytic activity also contributes
to protection in both mice and humans. We have recently discovered that IL-21 and GM-CSF are
produced by T cells and contribute to host resistance. IL-21 signaling plays a crucial role in T cell
responses during Mycobacterium tuberculosis infection.
IL-21 augments CD8+ T cell priming, promotes T cell accumulation in the lungs, and enhances T cell
cytokine production. IL-21 is produced predominantly by activated CD4+ T cells and has pleiotropic
effects on immunity via the IL-21 receptor (IL-21R), a member of the common gamma chain (γc)
cytokine receptor family. IL-21R-/- mice have increased lung bacterial burden and earlier mortality
compared to WT mice. To causally link the immune defects with host susceptibility, we use an
adoptive transfer model to show that IL-21R-/- T cells transfer less protection than WT T cells.
These results prove that IL-21 signaling has an intrinsic role in promoting the protective capacity of
T cells. Thus, the net effect of IL-21 signaling is to enhance host resistance to M. tuberculosis.
GM-CSF-/- mice are highly susceptible to infection with Mycobacterium tuberculosis and clinical data
has shown that anti-GM-CSF neutralizing antibodies can lead to increased susceptibility to
tuberculosis in otherwise healthy people. GM-CSF activates human and murine macrophages to
inhibit intracellular M. tuberculosis growth. We have previously shown that GM-CSF produced by
iNKT cells inhibits growth of M. tuberculosis. However, the general role of T cell-derived GM-CSF
during infection has not been defined. We find that non-conventional and conventional T cells
produce GM-CSF during M. tuberculosis infection. Early during infection, non-conventional iNKT
cells and γδ+ T cells are the main source of GM-CSF, a role subsequently assumed by conventional
CD4+ T cells as the infection progresses. Although we find that T cell production of GM-CSF can be
important for host defense, production of GM-CSF by radioresistant cells is quantitatively more
important in the mouse model. Thus, beyond the major contribution of GM-CSF production by
radioresistant cells, we identify T cell production of GM-CSF to be potential modifiable effector
function that contributes to host resistance against M. tuberculosis.
These data show that IL-21 and GM-CSF are T cell products that modify the outcome of
tuberculosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
67
48
Host Pathogen interaction and beyond
22/09/2016
Title to be confirmed
J. Cox
Berkeley University, San Francisco, United States
68
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
49
Host Pathogen interaction and beyond
22/09/2016
CD36-mediated uptake of surfactant lipids by human macrophages promotes intracellular
growth of M. tuberculosis
C.E. Dodd4-1, C.J. Pyle2-1, M. Rajaram1, L.S. Schlesinger3-1-4
1
Center for Microbial Interface Biology 2College of Pharmacy 3Department of Microbial Infection &
Immunity 4Microbiology, The Ohio State University, Columbus, United States
Mycobacterium tuberculosis (M.tb) is an extremely salient pathogen and causes a human death
every twenty seconds. M.tb is also capable of enduring for decades inside of the human body and
therefore a substantial reservoir of latently infected individuals exists as a lurking threat to global
health. A historical scourge of the human species, M.tb is now highly adapted to the lung
environment and is capable of intracellular survival within alveolar macrophages (AMs). As the
immunological guardians of the alveolar airspace, AMs not only phagocytose inhaled microbes and
particulate matter but are also crucial in the catabolism of lung surfactant, a lipid-protein complex
which lines the tissue. Macrophage phenotype and behavior is regulated by surfactant and M.tb can
utilize host lipids as a carbon source during infection. We therefore investigated which receptor(s)
contribute to surfactant uptake and whether the presence of those surfactant lipids within human
macrophages prior to infection with M.tb enhances bacterial survival. We show that preformed
scavenger receptor CD36 is redistributed from an intracellular pool to the cell membrane following
exposure to lipids and proteins found in lung surfactant. Over days in culture with surfactant
components, CD36 transcript and protein levels are increased relative to resting macrophages.
siRNA knockdown of CD36 inhibits the ability of human macrophages to acquire the most abundant
lipid species in surfactant, dipalmitoylphosphatidylcholine (DPPC). DPPC uptake is specifically
mediated by CD36, as acquisition of another surfactant lipid, phophatidylglycerol, is unaffected by
CD36 knockdown. Furthermore, Scavenger Receptor-A knockdown macrophages retain the ability
to acquire DPPC. Finally, when human macrophages are cultured in surfactant lipids prior to
infection with M.tb, bacterial growth is increased significantly. This surfactant-mediated growth
advantage does not occur if CD36 is knocked down prior to culturing macrophages in surfactant.
We conclude that CD36 contributes to surfactant lipid uptake by human macrophages and that M.tb
is able to exploit this function in an endogenously lipid-filled AM. Ongoing experiments are
investigating whether surfactant lipids are readily available as a carbon source for M.tb early during
infection.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
69
50
Host Pathogen interaction and beyond
22/09/2016
Infecting Amoebae with Mycobacteria to Study Conserved Mechanisms of Innate Immunity
T. Soldati
Department of Biochemistry, University of Geneva, Geneva, Switzerland
The first line of defence against bacteria infections are phagocytic cells of the innate immune
system. These cells kill bacteria via oxygen-dependent (e.g. ROS) and oxygen-independent (e.g.
chemicals, enzymes and microbicidal peptides) mechanisms. While multicellular organisms use
phagocytosis to kill microbes and initiate a sustained immune response, phagocytic amoebae
internalise bacteria as nutrients, via mechanisms of recognition, signalling and killing that are
surprisingly conserved. In particular, at the transition to multicellularity, eukaryotic organisms
acquired NOX enzymes to generate ROS. Dictyostelium is a social amoeba that feeds by
phagocytosis and has a rudimentary but highly conserved cell-intrinsic immune system. It is
genetically and biochemically tractable and has emerged as a powerful and experimentally versatile
host model organism. In particular, we study the mechanisms of infection by Mycobacterium
marinum, a close cousin of M. tuberculosis, which uses similar virulence strategies to re-program
and proliferate inside macrophages. We use this Dictyostelium/M. marinum model to study the interrelationships between host and pathogen in terms of nutritional immunity, acquisition of metabolites,
interference with the membrane trafficking and cytsokeletal processes. For example, recently, we
have used this system to study the role of iron, zinc and their transporters during mycobacterial
infection. In addition, we have focused on the monitoring of the impact of the TOR pathway and the
interplay between membrane damage and autophagy. Furthermore, we discovered that M. marinum
can use fatty acids derived from both host phospholipids and triacyglycerols and that this lipidbased metabolism is not directly linked with dormancy. We also used this complex system to identify
anti-infective compounds that impact on the fate of the infection without significant antibiotic activity.
70
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
51
Host Pathogen interaction and beyond
22/09/2016
Insights into the host-pathogen and microbiome interactions from dual RNA-sequencing of
tuberculous sputum
R. Lai5, T. Cortes4, S. Marais1, M. Burke5, N. Rockwood2-1, A. Garza-Garcia5, D. Young3-5,
R. Wilkinson2-5-1
1
Clinical Infectious Diseases Research Initiative, University of Cape Town, Cape Town, South
Africa 2Department of Medicine 3MRC Centre for Molecular Bacteriology and Infection, Imperial
College London 4Department of Pathogen Molecular Biology, London School of Hygiene and
Tropical Medicine 5The Francis Crick Institute, London, United Kingdom
Background: Transmission of Mycobacterium tuberculosis (Mtb) is via aerosol droplets of
infectious sputum. Understanding the phenotypes of the tubercle bacilli and their microenvironment
during this transition phase may illuminate new strategies to reduce transmission.
Methods: Sputum samples were collected from 17 patients with active TB disease, among which 8
were co-infected with HIV-1. Additional sputa were collected from 8 patients who were respiratory
symptomatics with diseases other than TB. Total RNA was extracted from sputa as well as
reference laboratory Mtb (H37Rv) cultures grown to exponential and stationary phase. We
performed a one-step, ultra-deep dual RNA-Seq on tuberculous and control sputa to survey the
global transcriptome of the host, Mtb and the resident microbiota.
Result: The transcriptional profile of Mtb most closely related that of actively replicating bacteria in
laboratory culture, with significantly increased expression of transcripts linked to cholesterol
metabolism and zinc deprivation, and reduced expression of the PhoP regulon implicated in control
of virulence determinants. Approximately 1% of the total bacterial reads in sputum mapped to Mtb.
Transcriptional profiling of commensal bacteria in the sputum provided further support for a low zinc
environment and suggested an integrated, interdependent metabolic network. The ability to utilize
cholesterol may reduce pressure on Mtb to compete for common nutrients with the established
microbiota in the respiratory tract.
In the host transcriptomes, sputa from TB patients displayed a strong type I and II interferon and
TNF-mediated inflammatory responses compared to those from non-TB controls, similar to whole
blood signatures already reported. We also observed transcriptional activity corresponding to the
Warburg effect in TB sputa, implicated by increased expression of genes associated with glycolysis
and decreased expression of genes associated with oxidative phosphorylation.
Conclusion: RNA-sequencing allows simultaneous characterization of host, Mtb and commensal
bacteria and gives a comprehensive snapshot on the oral and respiratory microenvironment. The
knowledge acquired offers insight into pathogenesis and on future strategies to better prevent Mtb
transmission.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
71
52
Host Pathogen interaction and beyond
22/09/2016
The minimal unit of infection: M. tuberculosis in the macrophage
D. Russell
Microbiology and Immunology, Cornell University, Ithaca, New York, United States
The interaction between Mycobacterium tuberculosis (Mtb) and its host cell is highly complex and
extremely intimate. Were it not for the disease, one might regard this interaction at the cellular level
as an almost symbiotic one. The metabolic activity and physiology of both cells are shaped by this
co-existence. We believe that where this appreciation has greatest significance is in the field of drug
discovery. Evolution rewards efficiency, and recent data from many groups indicate that Mtb has
evolved to utilize the environmental cues within its host to control large genetic programs or
regulons. But these regulons may represent chinks in the bacterium’s armor because they include
off-target effects, such as the constraint of Mtb’s metabolic plasticity. A prime example is how the
presence of cholesterol within the host cell appears to limit the ability of Mtb to fully utilize or
assimilate other carbon sources. We believe firmly that to understand the physiology of Mtb, and to
identify new drug targets, it is imperative that the bacterium be interrogated within the context of its
host cell. The constraints induced by the environmental cues present within the host cell need to be
preserved and exploited. The Mtb-infected macrophage truly is the “minimal unit of infection”.
72
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
53
Drug design-new developments
23/09/2016
Reseting acquired antibiotic resistance in Mycobacterium tuberculosis
A. Baulard
Center for Infection and Immunity, Institut Pasteur de Lille et INSERM, Lille, France
Antibiotic resistance is one of the biggest threats to human health. Amongst the scariest worldwide
progressions of this scourge is that of multi- and extensively-drug resistant Mycobacterium
tuberculosis. For some key antituberculosis pro-antibiotics, resistance mechanisms are mainly
driven by mutations in the bacterial enzymatic pathway required for their bioactivation. We have
developed drug-like molecules waking-up a cryptic alternative bio-activation pathway of ethionamide
in M. tuberculosis to circumvent the classic bioactivation pathway where mutations are observed in
resistant clinical strains. The first of its kind molecule, named SMARt-1 (Small Molecule Aborting
Resistance), not only fully reverses ethionamide-acquired resistance and successfully treats
ethionamide-resistant infection in mice, but also increases the basal sensitivity of bacteria to
ethionamide.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
73
54
Drug design-new developments
23/09/2016
Mycobacterium tuberculosis CTP synthetase and pantothenate kinase: two promising
targets for the development of multitargeting drugs
L.R. Chiarelli4, M. Esposito4, B.S. Orena4, G. Mori4, N. Buttari4, G. Degiacomi3, F. Gosetti2,
M. Manfredi2, S. Ekins7, K. Mikusova5, M. Bellinzoni1, R. Manganelli3, E. Marengo2,
L. Ballell-Pages6, G. Riccardi4, M.R. Pasca4
1
Structural microbiology Unit, Pasteur Institut, Paris, France 2Department of Science and
Technological Innovation, University of Piemonte Orientale, Alessandria 3Department of Molecular
Medicine, University of Padova, Padova 4Department of Biology and Biotechnology "Lazzaro
Spallanzani", University of Pavia, Italy, Pavia, Italy 5Department of Biochemistry, Comenius
University of Bratislava, Bratislava, Slovakia 6Tres Cantos Medicines Development Campus,
GlaxoSmithKline, Madrid, Spain 7Collaborative Drug Discovery, Burlingame, United States
Tuberculosis is responsible for about 1.5 million deaths/year, and the emergence and spread of
Mycobacterium tuberculosis multidrug-resistant strains has prompted the discovery for new drugs.
In a previous work (Mori et al., 2015), we validated the CTP synthetase PyrG as the target of
thiophene-carboxamide (82) and carbamothioyl-propanamide (88) prodrugs activated by EthA, and
of the (026) sulfonic active metabolite of 82.
In order to confirm PyrG as valuable TB target, testing the possibility of cross-reactivity of the
inhibitors with the human enzyme, we expressed and purified the human CTP synthetase 1. The
enzymatic inhibition assays of CTP synthase of PyrG inhibitors are in progress.
Recently, by genetic and biochemical approaches, we found a secondary target of these three PyrG
inhibitors. In particular, biochemical studies confirmed that panthotenate kinase PanK is inhibited by
the EthA-activated compounds, as well as by the active metabolite 026. These evidences
suggested that PyrG and PanK enzymes could be successfully used for the screening of compound
libraries, in order to identify potential multitargeting hits.
For this reason, the publically available GSK TB-set library of antitubercular compounds was firstly
assayed against PyrG activity, identifying 3 pyridine-thiazoles as effective, ATP-competitive
inhibitors (Ki=3-20 μM). PyrG was also genetically validated as target of these 3 GSK compounds,
while metabolic labeling studies demonstrated that the compounds affect different biosynthesis
pathways involving PyrG.
In order to verify if these compounds could be possible multitargeting hits, they were assayed
against recombinant PanK enzyme activity, finding that one of them, the N-(4-(pyridin-2-yl)thiazol-2yl)-[1,3]dioxolo[4',5':4,5]benzo[1,2-d]thiazol-6-amine, affected PanK. Biochemical analysis
demonstrated that the compound inhibits PyrG and PanK with a similar competitive inhibition
mechanism, and the molecular docking confirmed that it is able to interact with the ATP binding site
of both proteins.
In conclusion, the two targets PyrG and PanK have been demonstrated to be suitable tools for
identification of new potential multitargeting antitubercular compounds.
74
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
55
Drug design-new developments
23/09/2016
Antitubercular profile of GSK070 a Mycobacterium tuberculosis leucyl-tRNA synthetase
inhibitor
A. Mendoza-Losana2, E. Perez-Herran2, A. Palencia1, S. Cusack1, X. Li3, Y. Zhou3, M.R.K. Alley3,
D. Barros-Aguirre2
1
EMBL, Grenoble, France 2TbDPU, GSK, Tres Cantos, Spain 3Anacor Phamaceuticals Inc, Palo
Alto, United States
Background: Protein synthesis inhibitors play an important role in the treatment of tuberculosis.
The essential protein synthesis enzyme leucyl-tRNA synthetase (LeuRS) has two catalytic sites, an
aminoacylation site, which charges tRNALeu with leucine and an editing or proof-reading site that
hydrolyses incorrectly charged tRNALeu. The application of the oxaborole tRNA trapping (OBORT)
mechanism (Rock & Mao et al., 2007 Science 316: 1759-1761) to Mycobacterium tuberculosis
LeuRS led to GSK070, a new anti-tubercular agent with excellent potency and selectivity.
Material/methods: MIC determination, frequency of spontaneous resistance and other
microbiological experiments were performed as described by Parish & Roberts, 2015 (Mycobacteria
Protocols 3, Methods in Molecular Biology). Clinical isolates were obtained through the Spanish
National Institute of Health and antibacterial activity determined using the BACTEC MGITTM 960
System (Becton Dickinson). The GSK070 co-crystal structure was obtained with the editing domain
of M. tuberculosis LeuRS using amino acid residues from Gly309 to Ile515 in the presence of AMP
and the inhibitor.
Results: GSK070 was co-crystalised with the editing domain of M. tuberculosis LeuRS and was
found in the typical OBORT binding mode with the 4-chlorobenzoxaborole core occupying the
amino acid substrate site, while the boron atom formed two covalent bonds with the cis-diol from the
ribose of AMP. The 3-aminomethyl group from the inhibitor also plays a critical role as it interacts
with two negatively charged residues (Asp447, Asp450) and participates in an H-bond with the
carbonyl of Met441. These interactions yield a potent inhibitor of the enzyme that retains activity
against sensitive, MDR and XDR M. tuberculosis clinical isolates, exhibiting an MIC90 of 0.16 μM
with an MIC range of ≤0.02-0.31 µM. The frequency of spontaneous resistance is in the range of
other antituberculars and no cross-resistance in vitro has been observed with other antituberculars
that are in the clinic or in development. Further confirming GSK070’s unique mode of action, we
observed mutations in the leuS gene from these resistant mutants.
Conclusions: GSK070 has excellent activity against a set of 98 strains including drug-sensitive,
multidrug and extremely drug-resistant strains of M. tuberculosis. Its frequency of spontaneous
resistance is in the range of other clinically relevant antituberculars and most of the resistant
mutations map to the editing site of M. tuberculosis LeuRS. Finally, GSK070 does not show crossresistance with other antituberculars.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
75
56
Drug design-new developments
23/09/2016
First clinical trial with the novel drug candidate PBTZ169: expected results and surprises
V. Makarov1, S. Cole2
1
Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of
Sciences, Moscow, Russia 2Global Health Institute, Ecole Polytechnique Fédérale de Lausanne,
Lausanne, Switzerland
PBTZ169 is novel drug candidate with high efficacy in animals models and its combination
treatment of PBTZ169 with BDQ and pyrazinamide was shown to be more efficacious than the
standard treatment for tuberculosis in a mouse model. The target of PBTZ169 is famous DprE1, an
essential enzyme in cell wall biosynthesis. Furthermore this drug candidate demonstrated during
preclinical research “drug like” properties what made it an attractive drug candidate to treat TB in
humans.
During first clinical trials several cohorts of the healthy volunteers were treated by the single doses
of PBTZ169 as well as two weeks repeated treatment was chosen for two maximal doses. As
expected PBTZ169 was well tolerated and no significant toxicity effects was observed during the
trials.
The study of the metabolism shown that human metabolism of PBTZ169 is very different from
microbial or animals compound transformation. So main pathway of microbial, mice and less rats
metabolism connected with reduction processes, but human metabolism mainly connected with
oxidation processes. Due this difference we observed several new active metabolites of PBTZ169 in
human and now we can conclude that animal antituberculosis activity of PBTZ169 is a result not
only activity of the drug itself but it is a result of the sum activity of the drug and its metabolites.
Direct antimicrobial plasma activity was studied and such activity was observed for 24 hours after
human treatment for some doses. This data gets high chance for good efficacy of PBTZ169 in
human for treatment TB infection.
76
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
57
Drug design-new developments
23/09/2016
Griselimycins, a new class of anti-TB drugs
S. Lagrange
Infectious Diseases Unit, SANOFI, Marcy L'etoile, France
Griselimycin is a natural cyclic peptide isolated from Streptomyces species. Lead optimization of
Griselimycin resulted in identification of a novel synthetic analog derivative Cyclohexylgriselimycin
(CGM). DNA Polymerase sliding clamp (DnaN) has been identified as the target. CGM profiling
confirmed the interest of this new class for TB treatment. An update on this program will be done
and the selection of the most promising advanced lead for further late preclinical studies will be
presented.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
77
58
From animal models to man
23/09/2016
Animal Models of Tuberculosis for Efficacy Testing
A. Rawkins
National Infections Service, Public Health England, Salisbury, United Kingdom
Pre-clinical testing of novel therapies and vaccines in animal models of tuberculosis provides
essential information on safety, immunogenicity and efficacy which supports the development of
new products towards use in humans.
The species most commonly used to test the efficacy of vaccines are mice, guinea pigs and nonhuman primates. Efficacy is measured by comparing the relative severity of disease in terms of
bacterial burden, survival and/or analyses which describe the severity of the key pathological
features. Bacterial burden can be measured robustly in small animal species and enables relatively
rapid screening of large numbers of candidate vaccines in order to select the most promising for
testing in more relevant and complex animal models such as non-human primates or models which
more closely replicate human TB. These include models of HIV/TB co-infection, latent or reactivated
TB, simulation of neonatal or adolescent vaccination strategies and the use of more relevant strains
or doses of M. tuberculosis for challenge, including natural transmission between animals. Studies
are on-going at PHE with funding by the Bill and Melinda Gates Foundation to establish a guinea
pig model of naturally acquired infection using experimentally infected macaques as the source of
aerosols. Data generated in a pilot study have demonstrated the feasibility of transmission between
macaques and guinea pigs.
An increase in the complexity of the animal model can result in greater variation between animals
with regards to the measurements of disease severity and there is a balance to be struck between
the relevance of the model to humans and the ability to control variables and interpret the data.
Sophisticated methods of measuring bacterial burden and disease pathology, in-life by advanced
imaging have been developed and these significantly reduce this variation.
With scarce resources and for ethical reasons, it is imperative that data generated in animals are
informative. Evaluation of the same vaccine in early stage clinical trials and in animals, in parallel, is
feasible and greatly enhances the value of the animal studies and offers the potential to identify
correlates of protection which would significantly improve and accelerate the development of an
improved TB vaccine.
78
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
59
From animal models to man
23/09/2016
Tuberculosis and One Health: comparative analyses of the human and bovine tubercle
bacilli
S. Gordon
School of Veterinary Medicine, University College Dublin, Dublin, Ireland
The One Health concept seeks to improve human, animal and environmental health by building on
synergies across these areas. Comparative studies across human and animal TB offer a unique
area in which to explore such One Health synergies; indeed in his classic studies in the late 19th
century on the differentiation of human and bovine tubercle bacilli, Theobald Smith already
recognised the potential for such comparative work to “lead eventually to more light on the whole
subject of tuberculosis from the preventive as well as the therapeutic side” (Smith 1898). In this
presentation I will discuss work on comparative analyses of M. tuberculosis and M. bovis as the
exemplar human- and animal-adapted members of the M. tuberculosis complex, and look to what
these studies can teach us about the virulence, evolution and host adaptation of the complex as a
whole. The presentation will encompass comparative virulence studies of M. tuberculosis and M.
bovis in bovine experimental infections, the use of transcriptomics to define differential responses of
bovine alveolar macrophages to infection with M. tuberculosis and M. bovis, and exploration of
potential host tropic virulence mechanisms across the bacilli.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
79
60
From animal models to man
23/09/2016
Comparative Efficacy of New Oxazolidinones in a NHP Model of Tuberculosis evaluated by
[18F]FDG PET/CT and Bacterial Burden
L.E. Via1, G. Marriner1, Y. Park1, K.K. Robbins1, J. Sarathy2, M. Zimmerman2, B. Prideaux2,
C. Chen2, H. Boshoff1, V. Dartois2, J. Flynn3, C. Barry1
1
Tuberculosis Research Section, NIAID, NIH, Bethesda 2PHRI, Rutgers University,
Newark 3Department of Microbiology and Molecular Genetics, University of Pittsburgh School of
Medicine, Pittsburgh, United States
The number of new cases of multidrug resistant tuberculosis (MDR-TB) is increasing worldwide.
New chemotherapeutics are needed to augment treatment for MDR-TB, with the ultimate goal of a
new drug regimen useful for both drug susceptible and resistant disease. Linezolid (LIN) is used in
salvage treatment of MDR-TB, but it has an unfavorable toxicity profile with long-term use.
Therefore, other potentially less toxic oxazolidinones are being evaluated for TB. The common
marmoset, a small, non-human primate (NHP) model, is proving useful in assessing Mycobacterium
tuberculosis (MTB) drug efficacy. Five oxazolidinones were evaluated in the marmoset and mouse
models using in vivo imaging and terminal assays. Marmosets were infected with MTB for
approximately 7 weeks, given a pre-treatment 18F-fluorodeoxyglucose (FDG) PET/CT scan, and
randomized into treatment groups. The NHP were administered oral daily doses of LIN, AZD5847
(AZD), Radezolid (RAD), Sutezolid (SUT), or Tedizolid (TED) adjusted to match the T>MIC of the
expected human dose. Serial PET/CT scans were performed during the 1 or 2 months of treatment
and computationally evaluated to assess TB-associated abnormalities. C57BL/6 mice were similarly
infected and treated for 1 or 2 months with oxazolidinone doses equivalent to the T>MIC achieved
in the marmoset. After treatment, the animals were euthanized and the bacterial burden was
assessed. Marmosets treated with the oxazolidinones showed reduced or stabilized lung disease by
PET/CT and increased survival post-infection, compared to controls. Animals treated with LIN, AZD,
SUT and TED experienced the most consistent response showing reductions in both abnormal lung
volume and FDG uptake. Lesion and organ bacterial burdens were significantly reduced in
marmosets administered LIN, AZD and SUT. Quantitative reductions in abnormal lung volumes and
FDG uptake were highly associated with lower bacterial burdens. Studies of drug distribution into
epithelial lining fluid, penetration into caseum, and in vitro PK and PD assays partially predicted the
activity of these agents. In the mice, SUT promoted the most significant reduction in bacterial
burden followed by LIN with the other agents showing only modest activity. These results support
further investigation of SUT, AZD, and possibly TED as components in new drug regimens.
80
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
61
From animal models to man
23/09/2016
Evolution of Drug Resistance in Mycobacterium tuberculosis
A. Pym
Research Laboratory, AHRI (African Health Research Institute), Durban, South Africa
The continued advance of antibiotic resistance threatens the treatment and control of many
infectious diseases. This is exemplified by the largest global outbreak of extensively drug resistant
(XDR) tuberculosis (TB) identified at Tugela Ferry, KwaZulu-Natal, South Africa, in 2005. It is
unclear whether the emergence of XDR-TB was due to recent inadequacies in TB control or other
factors. Using whole genome sequencing on clinical and historical isolates we demonstrate the
evolution of drug resistance over a 50 year period and identify a common mutational pathway
responsible for multiple episodes of de novo evolution. By combining association and correlated
evolution tests with strategies for amplifying signal from rare variants, we were also able to identify
new mutations associated with resistance. One of these was a loss-of-function mutations in ald
(alanine dehydrogenase) which conferred resistance to D-cycloserine and emerged independent of
antibiotic selection in the evolution of animal adapted strains of the M. tuberculosis complex.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
81
62
From animal models to man
23/09/2016
Severe childhood tuberculosis as a genetic disorder
S. Boisson-Dupuis
INSERM Hôpital Necker/Rockefeller University, United States
Tuberculosis (TB) is caused by Mycobacterium tuberculosis (Mtb) and has long been considered
exclusively as an infectious disease. Up to one third of the world population is infected by Mtb,
however, only 10% of infected people develop clinical TB, including about 5% within two years after
infection (known as primary tuberculosis). This form predominantly affects children, is clinically
subacute and often spreads via the blood to tissues outside the lungs. The remaining 5% of infected
individuals develop TB later in life, years or decades after infection, typically due to microbial
reactivation from latency, which results in the chronic pulmonary form of adult TB. The genetic
factors of the host have been shown to play an important role in humans. The investigation of a rare
syndrome designated as Mendelian susceptibility to mycobacterial disease (MSMD), which is
characterized by susceptibility to poorly virulent mycobacteria (the vaccine strain, BCG, and
environmental mycobacteria), has led to the identification of a set of 10 human genes essential for
antimycobacterial immunity, all involved in IFN-γ-mediated immunity (upstream or downstream from
IFN-γ). In this context, unsurprisingly, several MSMD patients were also shown to be susceptible to
TB. In addition, children with complete IL-12Rβ1 or TYK2 deficiency were reported to display severe
TB, providing the first proof of principle that severe childhood TB may result from single-gene inborn
errors of immunity. They all display impaired production of IFN-γ in response to IL-12/IL-23,
probably accounting for their susceptibility to TB. These results strongly support the hypothesis that
childhood TB may result from a collection of single-gene inborn errors of immunity, at least in a
fraction of patients. However, most children with TB lack a genetic etiology. The laboratory has
recruited 200 children from Turkey, Iran and Morocco with severe tuberculosis and completed
Whole Exome Sequencing on each. Some of the results obtained will be presented during the
seminar, highlighting that TB of childhood results from a collection of diverse single-gene inborn
errors of immunity in a significant proportion of children.
82
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
63
Closing Keynote Lectures Session
23/09/2016
Interactions of mycobacterial phenolic glycolipids with host macrophages
L. Ramakrishnan
University of Cambridge, Cambridge, United Kingdom
We have developed the zebrafish model of tuberculosis where we can exploit the genetic tractability
and optical transparency of the larval stage to monitor and modulate host-pathogen interactions in
real-time. More recently, we have developed the zebrafish as a model for leprosy. I will present work
showing the interactions of mycobacterial phenolic glycolipids with host macrophages that
contribute to pathogenesis in both tuberculosis and leprosy.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
83
64
Closing Keynote Lectures Session
23/09/2016
Tuberculosis drug development: the leaking pipeline
S. Cole
EPFL, Lausanne, Switzerland
Tuberculosis (TB) was one of the first infectious diseases to be rationally treated and the golden
age of TB drug discovery led not only to new antibiotics but also to the development of combination
therapy. Until recently, no new TB drugs had been developed since the 1960s but, in response to
the HIV-pandemic and widespread resistance to first- and second-line drugs, a pipeline of candidate
TB drugs has now been established. I will review the scientific progress that fed this pipeline and
emphasize the important role played therein by public-private-partnerships. I will also highlight the
slow progress in clinical trials and extensive attrition rate, both compounded by lack of funding and
industrial commitment. As a result of these factors and poor coordination the pipeline has many
leaks.
84
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
POSTER SESSION 1
Tuesday, September 20
SUMMARY
Mycobacterial Genomics and Evolution ........................................ 87
Mycobacterial Biology & Physiology ............................................ 117
Transport systems & Secretion in Mycobacteria ......................... 184
65
Mycobacterial Genomics and Evolution
20/09/2016
Detection of genomic mutations in KatG, inhA and rpoB genes of Mycobacterium
tuberculosis isolates using polymerase chain reaction and multiplex allele-specific
polymerase chain reaction
A.D. Khosravi1, H. Goodarzi1, S.M. Alavi1, M. Afzali Behbahani2
1
Infectios and Tropical Diseases Research Center, Ahvaz Jundishapur University of Medical
Sciences, Ahvaz 2Tehran University of Medical Sciences, Tehran, Iran
Background: Isoniazid (INH) and Rifampin (RIF) are the most effective first line antibiotics against
Mycobacterium tuberculosis (MTB). Mutations in several genes are determining the resistance of
MTB to INH, with the most common gene target of KatG, and resistance to RIF is due to mutation in
rpoB gene.[1] The aim of present study was to determine the mutations in the regions related to RIF
and INH resistance in MTB strains isolated from tuberculosis patients in Ahvaz, Iran.
Methods: We characterized 80 clinical isolates of confirmed MTB to analyze the most commonly
observed INH and RIF mutations.[2,3] PCR analysis with subsequent sequencing were used to detect
mutations related to RIF and INH INH resistance. The multiplex allele-specific-PCR, was performed
as a comparative assay and for evaluation of this method. [4]
Results: The sequencing of the 250-bp region of katG codon 315, revealed point mutations at 5
different codons in 13.7℅ of the MTB isolates. The sequencing of the 270-bp central region of the
rpoB gene, revealed point mutations at 7 different codons in 12 (15%) of the MTB isolates. The
results obtained with multiplex allele-specific-PCR assay, were in concordance to PCR-sequencing
with high sensitivity and specificity for katG315, inhA15, and rpoB (531, 516, 526).
Conclusions: The result of this study suggested that molecular techniques can be used as a rapid
tool for the identification of drug resistance in clinical isolates of MTB. Both DNA sequencing and
multiplex allele-specific -PCR technique gave a high value sensitivity for the detection of RIF and
INH mutations and detecting multi-drug resistant tuberculosis cases.
Key words: M. tuberculosis, resistance, Isoniazid, Rifampin, mutation
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
87
66
Mycobacterial Genomics and Evolution
20/09/2016
Whole Genome Analysis of Clinical Mycobacterium bovis in Ghana Contradicts its
Classification Among Ancient Lineages of the Mycobacterium tuberculosis Complex
I.D. Otchere2-1, S. Harris4, S.L. Busso4, A. Asante-Poku2, S. Osei-Wusu2, K. Koram2, J. Parkhill4,
S. Gagneux3, D. Yeboah-Manu2
1
Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra 2Noguchi
Memorial Institute for Medical Research, University of Ghana, Accra 3Swiss TPH, Basel 4Wellcome
Trust Sanger Institute, Genome Campus, Cambridge University, UK, Cambridge, Ghana
Background: Tuberculosis caused by Mycobacterium bovis is a re-emerging problem in both
livestock and humans. With the scare of drug resistant strains, it imperative that an understanding of
the causative agent is realized for proper control of M. bovis associated TB.
Methods: Mycobacterium bovis (15) among 1740 Mycobacterium tuberculosis complex (MTBC)
isolates from Northern and Greater Accra regions of Ghana were screened for susceptibility to INH
and RIF. Five of the M. bovis strains were whole-genome sequenced and added to a pool of L4, L5,
L6 and global collection of MTBC genomes for comparative analysis.
Results: Mycobacterium bovis was isolated from 3/560 females and 12/1380 males. The average
age of the patients infected with M. bovis was 46.8 years (7 to 72 years). Four out of 212 (1.9%) TB
patients from the Northern compared to 11/1714 isolates from the Greater Accra regions were
infected with M. bovis. Two of the 15 (13.3%) bovine strains were isolated from HIV positive patients
whereas 6 (37.5%) were from patients who were in constant contact with livestock. The bovine
strains were susceptible to the 2 drugs except 2 INH (with katG S315T) and 1 RIF (with Q432P and
I1491S) mono-resistant strains. Maximum likelihood phylogenetic tree with 100 bootstraps and
rooted on M. canettii showed clustering of the 5 bovine TB genomes with the animal strains of the
global collection and were closer to MAF L6 than L5. Pan-genome analyses of the 5 M. bovis
genomes showed shared 3824 core-genes and 320 shell-genes. Comparing the presence and/or
absence of genes among the genomes, we found the bovine strains have relatively smaller genome
similar to L6 as compared to L4 and L5. We found the density of SNPs within the ESAT6 secretion
system was higher (1.4) than L6 (0.9), L5 (1.0) and L4 (0.1). We observed a number of mutations
within some house-keeping genes essential for in vitro growth in the bovine strains.
Conclusion: The bovine strains cluster with L6 and share relatively smaller genome compared to
L5 and L4. This challenges the classification of M. bovis as ancient and L4 as modern since the
MTBC evolves by genome deletions.
88
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
67
Mycobacterial Genomics and Evolution
20/09/2016
Transcriptional and proteomic profiling of diverse clinical isolates of Mycobacterium
tuberculosis
A. Banaei-Esfahani2-4, B. Collins2, O. Schubert2-5, A. Trauner1, S. Borrell1, S. Gygli1, M. Coscolla1,
S. Gagneux1, R. Aebersold2-3
1
Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health
Institute, Basel 2Department of Biology, Institute of Molecular Systems Biology, ETH
Zurich 3Faculty of Science, University of Zurich 4PhD Program in Systems Biology, University of
Zurich and ETH Zurich, Zurich, Switzerland 5Departments of Human Genetics, University of
California, Los Angeles, United States
Mycobacterium tuberculosis (Mtb) as one of the most successful human pathogens still has
remained a major source of mortality worldwide. Increasing number of drug resistant strains as well
as its ability in co-infection with HIV/AIDS place a massive burden on health care systems.
Mycobacterium tuberculosis complex (MTBC) comprises seven human-adapted lineages. Three
lineages (2-4) have been referred to as evolutionarily “modern” due to a deletion in genomic region
namely TbD1. Advent of next-generation sequencing and advanced proteomics in addition to new
computational approaches has paved the way for studying and curing tuberculosis more
systematically. Although Mtb is considered as highly clonal specie, genomic data show that two
human adopted MTBC strains differ by about 1200 SNPs on average while two thirds of SNPs in
coding regions are non-synonymous. This observation can propose that majority of these SNPs are
functional. In this study we profiled proteome of six fully sequenced clinical isolates from lineages
one (ancient lineage) and two (modern lineage). Quantification of about 80% of expressed genes
(~2500 proteins) using SWATH-MS allowed us to characterize the strains effectively. RNA
sequencing also enabled analysis of expression across the transcriptomes. PCA analysis admits
that the strains stand near to each other based on their relationship. We have corroborated that
DosR regulon has higher expression in lineage two at proteome level. It can be an appropriate
explanation for phenotypic behavior of modern lineages embedding more virulent strains. Around
one fourth of proteins including several enzymes and transcription factors, show significantly
differential expression between lineage one and two. Surprisingly number of changes at proteome
level as function of genomic differences (SNPs) behaves linearly. It’s also noted that every 7-8
SNPs is responsible for each protein change approximately. Overall these findings elucidate the
value of an integrated multilayered omics approach to characterize the underlying mechanisms
involved in Tuberculosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
89
68
Mycobacterial Genomics and Evolution
20/09/2016
Identification of mutations associated with resistance to the novel anti-tuberculosis drug
delamanid by a Whole Genome Sequencing approach
S. Battaglia5, E. Schena5, L. Nedialkova2, A.M. Cabibbe5, E. Borroni5, A. Trovato5-4, S. Niemann1,
C. Utpatel1, M. Merker1, S. Hoffmann-Thiel2-3, H. Hoffmann2-3, D.M. Cirillo5
1
Research Center Borstel, Molecular Mycobacteriology, German Center for Infection Research
(DZIF), Partner Site Borstel, Borstel 2Institute of Microbiology and Laboratory Medicine, IML red
GmbH, WHO Supranational Reference Laboratory of TB 3Institute of Microbiology and Laboratory
Medicine, IML red GmbH, WHO Supranational Reference Laboratory of TB, Germany Synlab MVZ,
Gauting, Germany 4Center for Translational Genomics and Bioinformatics (CTGB), San Raffaele
Scientific Institute 5Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation,
and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
Among the new anti-TB drugs, delamanid (DLM) has been conditionally approved for the therapy of
MDR-TB in 2014. This compound impairs the biosyntesis of mycolic acids and requires activation by
F420-dependent nitroreductase encoded by ddn gene. F420 cofactor is synthetized by enzymes
coded by fbiA, fbiB, fbiC and fgd1 genes. Polymorphisms in these genes were shown to be involved
in in vitro DLM resistance (DLM-R). We characterized the mutations leading to DLM resistance in a
panel of M. tuberculosis complex clinical isolates. A total of 250 MTBC strains representing eleven
different lineages and all drug resistance profiles were included in this study. Minimal inhibitory
concentrations (MICs) of DLM were determined by Bactec 960 MGIT system and resazurin
microtiter assay (REMA). Genomic DNA was extracted by CTAB method for Whole Genome
Sequencing (WGS) analysis. Paired-end (101 bp) libraries were prepared using Nextera XT DNA
Sample Preparation kit and sequenced on HiSeq 2500 platform (Illumina Inc). Samples showing a
coverage ≥ 20X were considered for SNPs calling by PhyResSE web-tool. DLM susceptible isolates
showed MICs between 0.03 and 0.0005 mg/L in REMA and between 0.06 and 0.0005 ml/L in MGIT.
WGS analysis on MTB isolates revealed fifteen non-synonymous mutations in the five genes
screened. Two polymorphisms were previously reported as lineage-specific for Harlem and M.
africanum 2. WGS analysis of DLM-S strains revealed the presence of nine new non-synonymous
mutations, while analysis of the five DLM-R strains (REMA MIC ≥ 32 mg/L, MGIT MIC ≥ 16 mg/L)
revealed the presence of three ddn W88Stop mutations and one fbiA K250Stop change in four
clustered Beijing strains (W148 clone), and absence of mutations in the last DLM-R isolate.
Moreover, NGS technique allowed us to rapidly analyse others potentially DLM-R related genes,
possibly involved in the emergence of DLM-R phenotype. It is worth noting that the population of
strains analysed were isolated from patients never exposed to DLM showing that mutations
inducing DLM-R phenotype are pre-existing in MTBC clinical strains and potentially associated to
specific clones.
90
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
69
Mycobacterial Genomics and Evolution
20/09/2016
Genetic characterization of a non-canonical MMR-like pathway in mycobacteria
A. Castañeda-García2, A. Prieto3, A. Rojas3, J. Rodriguez-Beltran3, T. Tønjum1, S. Waddell4,
A. Doherty4, J. Blazquez2-3
1
Oslo University Hospital, Oslo, Norway 2Centro Nacional de Biotecnologia, Madrid 3Instituto de
Biomedicina de Sevilla (IBIS)-CSIC, Seville, Spain 4University of Sussex, Brighton, United Kingdom
Mismatch Repair (MMR) is a near ubiquitous pathway that removes mismatched nucleobases from
DNA. MMR is essential for the maintenance of genome stability. Despite its major importance, an
apparent MMR mechanism is absent in mycobacteria. However, these bacteria exhibit rates and
spectrum of spontaneous mutations similar to MMR-bearing species, suggesting the existence of an
alternative MMR pathway. To identify novel mutation avoidance components in mycobacteria, a
Mycobacterium smegmatis insertion mutant library was generated and screened for spontaneous
mutation to rifampicin resistance, used as a hypermutator hallmark. As a result, a novel gene,
named mmrE, was identified as a key anti-mutator gene in Mycobacterium. MmrE, a putative DNArepair enzyme, is required for mutation avoidance and anti-recombination, hallmarks of canonical
MMR, and bears no structural homology to known MMR factors. Indeed the ΔmmrE phenotypes are
almost identical to those produced by the MMR deficiency in other bacteria (very high mutation rates
with transition-biased mutational spectrum and increased homeologous recombination rates).
Furthermore, mmrE and known MMR genes are mutually exclusive in their distribution among
organisms, as expected for alternative pathways. Finally, naturally occurring mmrE polymorphisms
in clinical M. tuberculosis isolates with diminished MmrE activity produce hypermutable phenotypes,
as occurs with MMR-bearing bacteria. In summary, our results strongly suggest that a novel MMRlike mechanism operates in organisms as mycobacteria that possess MmrE, but lack MutS-MutL.
Therefore, MmrE appears as an essential piece that, together with the high-fidelity DNA-polymerase
DnaE1 and its PHP domain-proofreader, maintain low mutation rates (~10-10 mutations per base per
generation) in mycobacteria, ensuring genome stability and DNA fidelity. Understanding the
mechanisms and pathways that influence mutation rates may unveil new strategies to predict and to
combat the development of drug resistance.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
91
70
Mycobacterial Genomics and Evolution
20/09/2016
Impact of the M. tuberculosis Genetic Background on the Acquisition of Drug Resistanceconferring Mutations
S. Borrell1-2, J. Feldmann1-2, R. Castro1-2, S. Gagneux1-2
1
Swiss Tropical and Public Health Institute 2University of Basel, Basel, Switzerland
The extent of genetic diversity in M.tuberculosis (Mtb) is more pronounced than was traditionally
believed, and can be classified into seven different geographically distributed lineages. The average
inter lineage genetic distance is approximately 2000 single nucleotide polymorphisms (SNPs). The
impact of this diversity on the baseline metabolism is still unknown. One of these lineages; the
Beijing lineage has repeatedly been associated with drug resistant tuberculosis (DR-TB). The
reason for this association is still not clear. To evaluate the influence of genetic background on the
acquisition of specific Isoniazid (INH) resistance-conferring mutations, and to explore whether the
Beijing lineage is better adapted to the potential physiological effects than other phylogenetical
distant lineages, associations between lineage and INH-DR were sought.
We performed Lüria-Delbruck fluctuation assay (LDFA), and found lineage-specific differences in
the baseline INH-resistance acquisition rate, with Beijing showing a higher rate compared to the
other lineages. Lineage specific INH-associated mutations were also explored. In addition, since
Beijing intra lineage diversity shows a phylogeographical structure and epidemiological studies have
evidenced differences on the Beijing-DR association depending on the strain geographical origin,
we further explored the impact of intra lineage diversity on the basal INH-DR mutation rate
performing LDFA of a collection of clinical Beijing strains globally distributed.
Overall, our findings support a role of Mtb lineage diversity in the emergence of global drug
resistance and consequently on the specific evolution of drug-resistance within each lineage.
92
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
71
Mycobacterial Genomics and Evolution
20/09/2016
HIV/TB coinfection reduces selective constraints on Mycobacterium tuberculosis
D. Brites1-2, E. Wampande3, J. Hattendorff1-2, K. Eisenach5, H. Boom4, M. Joloba3, S. Gagneux1-2
1
Tuberculosis Research Unit, Swiss Tropical and Public Health Institute 2University of Basel, Basel,
Switzerland 3Department of Medical Microbiology, Makerere University, Kampala,
Uganda 4Tuberculosis Research Unit, Case Western Reserve University and University Hospitals
of Cleveland, Cleveland 5Department of Pathology, University of Arkansas for Medical Sciences,
Little Rock, United States
The incidence of tuberculosis (TB) has increased in many parts of the world, partially fueled by HIV
coinfections. Given the importance of T-cell responses both in providing protective immunity against
tuberculosis, but as well as drivers of lung pathology and ultimately mediators of transmission, the
immunocompromised host environment is likely to affect the life cycle of M. tuberculosis. We
therefore hypothesized that the immunocompromised host environment affects the fitness of M.
tuberculosis and tested whether and how HIV/M. tuberculosis coinfections impact the genetic
structure of a M. tuberculosis population. To this end, we apply molecular population genomics to
whole-genome polymorphism data from a population of 180 M. tuberculosis strains circulating in
HIV infected and uninfected individuals sampled across 12 years in Kampala, a city with a high
burden of TB/HIV coinfections. Our results suggest that HIV coinfections have not subdivided this
M. tuberculosis population significantly. The HIV status of the host seems however to affect the fate
of mutations of M. tuberculosis strains significantly; M. tuberculosis from HIV co-infected hosts
harbor an excess of mutations present at low frequencies. This excess is significant both for
synonymous and nonsynonymous mutations pointing to an exacerbated role of genetic drift in M.
tuberculosis from HIV/TB coinfections. This could reflect that M. tuberculosis from HIV infected
patients is less transmitted. Supporting this, we show that M. tuberculosis from patients with lower
CD4+ T-cells (≤250 cells/ul) also tend to have more mutations which are uniquely found in the
population (i.e. where not transmitted), than M. tuberculosis strains which infect patients with higher
CD4+ T-cell numbers. Our study supports findings from epidemiological studies suggesting that
transmission of M. tuberculosis from HIV coinfected hosts is reduced, and suggests that M.
tuberculosis fitness is reduced by HIV/TB coinfections. However, it also provides evidence that the
continuous progression of M. tuberculosis/HIV syndemics can suply M. tuberculosis with genetic
diversity and hence provide opportunities for bacterial adaptation. The latter might facilitate the
evolution of resistance to anti-TB drugs.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
93
72
Mycobacterial Genomics and Evolution
20/09/2016
Comparison of phenotypic and genotypic methods for the detection of resistance to
second line injectable drugs in multidrug-resistant clinical isolates of Mycobacterium
tuberculosis in France
F. Brossier, A. Pham, C. Bernard, V. Jarlier, A. Aubry, N. Veziris, W. Sougakoff
French NRC for Mycobacteria, Pitié-Salpêtrière Hospital, Paris, France
Background: The second line injectable drugs (SLID, i.e. the aminoglycosides amikacin (AMK) and
kanamycin (KAN) and the cyclic peptide capreomycin (CAP)) are key drugs for the treatment of
multidrug-resistant tuberculosis (MDR-TB). SLID act by binding to 16S rRNA and inhibiting protein
synthesis. Mutations at positions 1401, 1402 and 1484 in the 16S rRNA gene (rrs) have been
associated with cross-resistance to these drugs. In addition to rrs modifications, mutations in the
promoter region of the eis gene, which encodes an aminoglycoside acetyltransferase, and in the
tlyA gene, which encodes a putative rRNA methyltransferase, are associated with resistance (R) to
KAN and CAP in Mycobacterium tuberculosis, respectively. The goal of the present study was to
compare the sequencing data of SLID resistance-associated genes to the corresponding results of
phenotypic drug susceptibility testing (DST) by the proportion method.
Methods: 206 MDR M. tuberculosis clinical isolates collected in 2010-2014 at the French Reference
Center for Mycobacteria were included, 153 susceptible to AMK/KAN/CAP, and 53 resistant to at
least 1 of the 3 SLID (23 R-AMK/CAP/KAN, 22 monoR-KAN, 3 R-AMK/KAN, 2 R-CAP/KAN, 3
monoR-CAP) including 29 XDR. DST was performed on Lowenstein-Jensen medium.
Results: Among the 53 strains resistant to at least 1 of the SLID, mutations in rrs accounted for
resistance to AMK, CAP and KAN for 81%, 75% and 44% isolates, respectively, while mutations in
eis promoter were detected in 44% of the isolates resistant to KAN. By contrast, no mutations in tlyA
were observed in the isolates resistant to CAP. Among the 153 isolates susceptible to the 3 SLID,
145 showed no mutation, 1 harbored unknown T1404C plus G1473A mutations in rrs and 7 had an
eis promoter mutation.
Conclusions: The discrepancies observed between the genotypic (on the primary culture) and
phenotypic drug susceptibility testing could be explained by i) a low level of resistance not detected
by phenotypic testing (n=8 strains), ii) a low percentage of resistant mutants not detected by
sequencing of drug resistance-associated genes on the primary culture (n=8 strains), and iii) to
other resistance mechanisms not yet characterized (n=7 strains).
94
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
73
Mycobacterial Genomics and Evolution
20/09/2016
Genetic diversity of Mycobacterium tuberculosis strains spread in the Northwestern region
of Russia
E. Chernyaeva2, M. Rotkevich2, A. Yurchenko2, V. Zhuravlev1, N. Solovieva1, M. Shulgina1,
A. Lapidus2, S. Obrien2
1
St. Petersburg Research Institute of Phtisiopulmonology 2Dobzhansky Center for Genome
Bioinformatics, St. Petersburg State University, St. Petersburg, Russia
The principal goal of the project is to develop a genome-wide variations catalogue of
Mycobacterium tuberculosis strains spread in the Russian Federation for clinical and
epidemiological purposes.
For whole genome sequencing we chose DNA samples obtained from strains revealed in St.
Petersburg (n=20), Leningrad region (n=18), Republic of Karelia (n=11) and Kaliningrad region
(n=26). Whole genome sequencing (WGS) were performed using Illumina MiSeq platform. M.
tuberculosis H37Rv reference genome (NC_000962.3) was used for SNP and Indel calling using
bowtie2 and samtools packages. RaxML package was used for phylogenetic analysis,
Mycobacterium canettii genome was used as outgroup. R-package was used for statistical analysis.
The majority of sequenced isolates formed 5 known phylogenetic groups corresponding to Beijing,
LAM, Ural, Haarlem and T spoligotypes. More than a half of sequenced isolates in each region
belonged to Beijing family. The second highest prevalence group in St. Petersburg, Leningrad
region and Karelia was LAM famity, however in Kaliningrad it was a T family which was rearly
identified in Leningrad region and was not detected in St. Petersburg and Karelia. Ural genetic
family was identified in 15% of M. tuberculosis isolates from Kaliningrad and only few isolates from
St. Petersburg, Leningrad region and Karelia belonged to this genetic family.
Thus, genome-wide analysis of M. tuberculosis isolates obtained in the Northwestern region of the
Russian Federation, revealed a difference in population structure of M. tuberculosis isolates
obtained in Kaliningrad region compared to other regions. However, all studied geographical
regions are characterized by prevalence of Beijing genetic family.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
95
74
Mycobacterial Genomics and Evolution
20/09/2016
Genome-wide analysis of Mycobacterium tuberculosis strains isolated from patients with
tuberculous spondylitis
E. Chernyaeva2, M. Rotkevich2, A. Yurchenko2, V. Zhuravlev1, N. Solovieva1, M. Shulgina1,
S. Obrien2
1
St. Petersburg Research Institute of Phtisiopulmonology 2Dobzhansky Center for Genome
Bioinformatics, St. Petersburg State University, St. Petersburg, Russia
The aim of the study is to find nucleotide polymorphisms (SNPs and InDels) associated with
different types of pulmonary tuberculosis (TB) and tuberculous spondylitis (TS) based on whole
genome sequencing data of Mycobacterium tuberculosis strains, collected in different regions of the
Russian Federation.
Genomic DNA of 146 M. tuberculosis strains was sequenced using Illumina MiSeq platform. M.
tuberculosis H37Rv reference genome (NC_000962.3) was used for SNP and Indel calling using
bowtie2, samtools and GATK packages. RaxML package was used for phylogenetic analysis,
Mycobacterium canettii genome was used as outgroup. R-package was used for statistical analysis.
Studied M. tuberculosis isolates were collected from patients with different clinical TB outcome: 55
were isolated from patients with TS; 68 – from patients with pulmonary TB (dissiminated, infiltrative
and fibro-cavernous TB); 23 – from patients with mixed TB (pulmonary and extrapulmonary).
To perform genome association study a phylogenetic analysis was conducted. Pulmonary and
extrapulmonary M. tuberculosis isolates were found in all phylogenetic clusters. Beijing genotype
was prevalent in all groups of M. tuberculosis isolates, however the prevalence of Beijing genotype
among TS isolates was higher than among pulmonary TB isolates (76% and 67%, respectively).
Meanwhile pulmonary TB isolates belonged to LAM family that TS isolates (16% and 5%,
respectively).
SNPs and InDels associated with phylogenetic clusters were removed and the rest genetic variants
were used for associations study. Statistical analysis allowed to identify SNPs and InDels
associated with clinical patterns (p ≤ 10-5). Over 40 SNPs discriminating infiltrative and fibrocavernous TB and 15 SNPs discriminating TS and fibro-cavernous TB were found in CDS and
intergenic regions. However, there were no reliable SNPs found discriminating pulmonary TB in
general and extrapulmonary TB. InDels discriminating pulmonary and extrapulmonary TB were
identified in PE_PGRS genes and need to be validated by Sanger sequencing.
96
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
75
Mycobacterial Genomics and Evolution
20/09/2016
Predicting the impact of global genomic variation of Mycobacterium tuberculosis in the
bacterial regulatory network
Á. Chiner-Oms4, F. González-Candelas4-3-2, I. Comas1-2
1
Tuberculosis Genomics Unit, Biomedicine Insitute of Valencia 2CIBER in Epidemiology and Public
Health 3Infection and Public Health, Foundation for the Promotion of Health and Biomedical
Research of Valencia Region 4Cavanilles Institute of Biodiversity and Evolutionary Biology,
University of Valencia, Valencia, Spain
We have constructed a set of computational models to predict the expression of Mycobacterium
tuberculosis genes from a previously published regulatory network and a very large expression
dataset.
The accuracy of the models was assessed by testing them versus a set of random models. The final
models were also tested with two different expression datasets. We use these models to predict the
expression of a mutated strain of M. tuberculosis lacking an important transcription factor called
phoP. Combining the information derived from these robust models obtained we propose a new,
more robust regulatory network.
We have also tested a set of genome-wide single nucleotide polymorphisms and deletions from a
large collection of clinical strains. We noted that some transcription factors from the regulatory
network were mutated or deleted in several of the seven M. tuberculosis lineages. These
transcription factors are embedded in regulatory subnetworks involving several metabolic,
transcription and transport related processes.
We speculate that the regulatory network may vary, albeit only slightly, among lineages and these
differences could be related to some of the phenotypic and epidemiological differences observed
among M. tuberculosis strains of different lineages.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
97
76
Mycobacterial Genomics and Evolution
20/09/2016
SMRT Genome Assembly Corrects Reference Errors, Resolving the Genetic Basis of
Virulence in Mycobacterium tuberculosis
A. Elghraoui, S. Modlin, F. Valafar
biomedical Informatics Research Center, San Diego State University, San Diego, United States
The genetic basis of virulence was investigated with the publication of the Mycobacterium
tuberculosis H37Ra genome, the avirulent counterpart to the M. tuberculosis reference strain
H37Rv. Variants with respect to H37Rv affecting 56 genes were identified as H37Ra-specific and
potentially causal to its attenuated virulence. These genomic differences have been subsequently
explored in at least 19 studies. Such analysis, however, heavily relies on the accuracy of the
sequences. While the H37Rv reference genome has had several corrections to date, that of H37Ra
is unmodified since its original publication. Here, we report the sequencing, assembly, and finishing
of the H37Ra genome with single-molecule sequencing, a platform capable of exceeding the
accuracy of Sanger-based technologies. Our assembly reveals that the number of H37Ra-specific
variants is less than half of what was determined based on the current H37Ra reference sequence.
PE/PPE family genes, which are intractable to typical short-read sequencing platforms because of
their repetitive and GC-rich nature, were over-represented in the set of genes with all reported
H37Ra-specific variants contradicted. Furthermore, we identified a sequencing error in H37Ra that
masked a true variant which, when considered in the context of previous work, corresponds to a
sequencing error in the H37Rv reference genome. By identifying sequencing errors in the H37Ra
reference genome, the conclusions of several studies are undermined and in some cases
invalidated. Through the reduction of genes with variants of unknown effect on virulence, our
assembly allows for a more focused and pertinent inquiry into the genetic factors that may
contribute to the difference in virulence between H37Ra and H37Rv. Our assembly also illustrates
the power and utility of single-molecule sequencing in producing accurate and fully resolved de
novo assembled genomes of mycobacteria.
Funding: This work has been supported by grants from National Institute of Allergy and Infectious
Diseases (NIAID) grant RO1AI05185 and National Science Foundation (NSF) grant 0966391.
98
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
77
Mycobacterial Genomics and Evolution
20/09/2016
HADTB: Hub for Aggregated Data in Tuberculosis, An Aggregated Database for Sharing,
Annotating, and Analyzing Tuberculosis (TB) Genomic Data
D. Oh, Y. Kim, A. Elghraoui, F. Valafar
biomedical Informatics Research Center, San Diego State University, San Diego, United States
Recent advances in sequencing have generated vast amounts of genomics data. Existing
databases are either not reliably maintained or do not integrate well with existing analysis tools. As
such, it is difficult to use this data for reliable knowledge generation that could be used in bedside
practice or in public health efforts in controlling outbreaks. To promote collaboration and global
analysis, a single database that can aggregate publicly available (genomic and meta) data, provide
multi-facetted dissemination interface, and be easily incorporated in bioinformatics pipelines is
essential. HADTB is an integrated database that allows users to query, upload, and annotate TB
data. Its multi-modal interface allows easy incorporation into custom analysis pipelines. HADTB is
community driven and self-sustaining.
The backend of HADTB is built on a MySQL database and mapped using SQLAlchemy. The Flask
framework is used to create REST API calls where users can access the database through a simple
URL. The front end is built using AngularJS framework. This provides more dynamic views than
PHP or other alternatives. The web-interface of HADTB is divided into three unique groups; genes,
variants and isolates. All the fields in each of these groups are relationally mapped to one another
which allows users to retrieve all associated genomic and metadata in a single table view.
Moreover, built in report function provides users with broad range of powerful statistical and
graphical methods for easy visualization.
In addition to community contribution, HADTB automatically aggregates publicly available raw
sequencing reads from published papers and uniformly processes them for variant calling. Along
with independent gene, variant, isolate, and literature data, the database also features relational
mapping where each entity can be viewed with its interactive counterparts for pathway and
regulatory analysis. Curation status promotes database integrity by differentiating user uploaded
experimental data from verified published data. HADTB provides multiple access points: a web
interface, a console client, a Python library, and a REST API.
Funding: This work has been supported by grants from National Institute of Allergy and Infectious
Diseases (NIAID) grant R01AI05185 and National Science Foundation (NSF) grant 0966391.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
99
78
Mycobacterial Genomics and Evolution
20/09/2016
Identification of Conditionally-Essential Genes of the M. kansasii Genome to Understand
the Evolution of Mycobacterial Virulence
M. Ghanem1-3-4, F. Mcintosh1-4, J. Wang5, M. Behr1-2-3-4
1
McGill International TB Centre 2Department of Medicine 3Department of Microbiology and
Immunology, McGill University 4Research Institute of the McGill University Health Centre, Montreal,
Canada 5University of Michigan, Ann Arbor, United States
Mycobacterium kansasii, commonly isolated from tap water, is an NTM that can cause a pulmonary
TB-like disease. However, there are no records showing person-to-person transmission and
infection is usually cleared in healthy individuals. Phylogenetic analyses performed in our lab have
shown M. kansasii to be the most closely related environmental species to the M. tuberculosis
complex (MTC). Genomic comparisons revealed a genome 2-Mbp larger than that of M.
tuberculosis, with orthologues in M. kansasii for at least 65% of all M. tuberculosis antigens. From
this, we infer that certain M. tuberculosis virulence genes are not specific to the MTC, but instead
are generic factors that contribute to the pathogenesis of disease by multiple mycobacteria.
To identify genes that contribute to M. kansasii survival, we use transposon sequencing; a random
genome-wide mutagenesis technique that will identify which mutants thrive (anti-virulence genes) or
succumb (virulence genes) following pulmonary infection. This method makes use of MycoMarT7; a
mycobacteriophage engineered to integrate transposons into mycobacterial genomes in a TAdinucleotide-specific manner. The M. kansasii genome includes ~ 95,000 TA sites dispersed
throughout its genome, making MycoMarT7 an ideal mutagenic agent for our purposes. We have
created 4 mutant libraries with the goal of covering a large portion of the M. kansasii genome. We
have also validated two high-dose infection models in C57/BL6 mice, enabling us to ensure that a
greater number of bacteria are studied than the number of genes in the M. kansasii genome.
Following a 6-week infection, Illumina HiSeq Next Generation Sequencing will then be used to
quantify two output pools, contrasting in vitro growth on Middlebrook agar and the lung
homogenate. Both output pools will be contrasted with the input (initial inoculum).
This study aims to identify genes that have played important roles during the evolution of different
mycobacterial pathogens. These genes are classified into 4 categories: (1) shared amongst all
slow-growing mycobacteria, (2) unique to M. kansasii, (3) shared between M. kansasii and other
NTMs excluding the M. tuberculosis complex and (4) shared between M. kansasii and M.
tuberculosis excluding other NTMs.
100
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
79
Mycobacterial Genomics and Evolution
20/09/2016
Complementation of Mycobacterium tuberculosis-specific genes in M. kansasii to
investigate the evolution of TB as a human pathogen
M. Ghanem2-4-1, J. Wang6, F. Mcintosh3-1, M. Reed2-4-1-3, P. Domenech4-1, D. Young5, B. Moody5,
M. Behr2-4-1-3
1
McGill International TB Centre 2Department of Microbiology and Immunology, McGill
University 3Department of Medicine, McGill University Health Centre 4Research Institute of the
McGill University Health Centre, Canada 5Division of Rheumatology, Immunology and Allergy,
Brigham and Women’s Hospital, Harvard Medical School 6Department of Microbiology and
Immunology, University of Michigan, United States
Our group has recently executed a genomic comparison between Mycobacterium tuberculosis and
M. kansasii, an environmental species that causes a TB-like non-transmissible lung disease.
These findings support the hypothesis that the latter can be used as a pertinent model to assess
the genetic requirements that led to the emergence of M. tuberculosis as a professional human
pathogen.
In the present project, we are using M. kansasii as a surrogate host, via gain-of-function studies, to
better understand the role of M. tuberculosis-specific genes on the behavior of the microorganism
during infection. The genes of interest in this study, Rv3377-8c, are implicated in the synthesis of
1-tuberculosinyladenosine (1-TbAd), a lipid found exclusively in M. tuberculosis.
1-TbAd makes up 1% of all lipids produced by the pathogen, is constitutively synthesized and its
absence leads to impeded growth of M. tuberculosis in macrophages, at least partially due to
problems with phagosome acidification. We hypothesize that the production of 1-TbAd will provide
M. kansasii with the means to become a more robust pathogen.
Through heterologous complementation, we have produced a clone of M. kansasii that has
successfully gained the ability to produce a denosine-labeled lipids, as s een by TLC
chromatography. We have also confirmed the identity of these lipids as 1-TbAd by liquid
chromatography–mass spectrometry. Our current efforts focus on assessing the in vitro growth of
the complemented M. kansasii strain as well as its abilities to survive and replicate in
macrophages and in vivo murine infection models.
This study provides proof-of-concept for the role of horizontal gene transfer in the emergence of M.
tuberculosis and provides a tool to study the specific contribution of M. tuberculosis- specific genes
in the pathogenesis of TB.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
101
80
Mycobacterial Genomics and Evolution
20/09/2016
Comparative genome of Mycobacterium bovis strain 1595 isolated from Korean cattle
J.M. Kim, Y.H. Jang, M.H. Hwang, N.R. Kim, H.S. Lee
Zoonosis lab, Animal and Plant Quaratine Service, Gyeongsangbuk-Do, South Korea
Mycobacterium bovis is the causative agent of bovine tuberculosis and zoonotci and both animal
and human health concerns. In this study, we isolated and performed and genomic investigation of
M. bovis strain 1595 from laryngopharyngeal lymph node of Korean native cow. M. bovis 1595 was
reveled to have SB0140 in spoligotype and 4-2-5-3-2-7-5-5-4-3-4-3-4-3 in MIRU-VNTR typing.
Combine these, strain 1595 was categorized the most frequenct patten in Korea. The complete
genome sequence of strain 1595 determined using Illumina MiSeq and PacBio single-molecular
real-time technology indicated 4,351,712 bp in size with a 65.64% G+C content. And 4,358 proteincoding genes were predicted from this assembly. In addition, we performed brief comparative
genomic analysis with genomes of Mycobacterium tuberculosis complex strains which reveled that
all genomes have similar size and G+C content. In phylogenetic analysis, all strains located within
0.1% average nucleotide identity value, and MUMmer analysis illustrated all genomes showed
positively collinear with strain 1595. Comparison based on BLASTP, the strain that has the most
completely matched proteins with M. bovis 1595 was M. bovis AF2122/97. This genome sequence
will serve as a valuable reference for improving the understanding of the disparity in the virulence
and epidemiologic traits between M. bovis genotypes in South Korea.
102
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
81
Mycobacterial Genomics and Evolution
20/09/2016
The evolution of Mycobacterium tuberculosis in HIV co-infected individuals in an HIV/TB
endemic setting
A. Koch3-5, D. Brites8-9, D. Stucki8-9, J. Evans6-5, R. Seldon6-5, M. Nicol1-5, T. Oni7, D. Warner6-5,
V. Mizrahi6-5, J. Parkhill10, S. Gagneux8-9, D. Martin4-5, R. Wilkinson3-5-12-11
1
National Health Laboratory Service 3Clinical Infectious Diseases Research Initiative 4Division of
Computational Biology 5Institute of Infectious Disease and Molecular Medicine 6Molecular
Mycobacteriology Research Unit 7School of Public Health and Family Medicine, University of Cape
Town, Cape Town, South Africa 8Swiss Tropical and Public Health Institute 9University of Basel,
Basel, Switzerland 10The Wellcome Trust Sanger Institute, Hinxton, Cambridge 11Francis Crick
Institute 12Department of Medicine, Imperial College, London, United Kingdom
Infection with HIV increases the risk of infection with tuberculosis (TB), and co-infection leads to
acceleration of both diseases. However, the biological interactions between HIV-1, Mycobacterium
tuberculosis (Mtb), and the human immune system are incompletely understood. We applied
comparative genomics to investigate the impact of HIV-1 co-infection on the evolution of Mtb strains
circulating in Khayelitsha, Cape Town, South Africa, a region with high rates of HIV-associated TB.
Evolutionary models, Fast Unconstrained Bayesian Approximation (FUBAR) and Model of Episodic
Directional Selection (MEDS), were applied to whole-genome sequence (WGS) data for 169 Mtb
strains. These phylogenetically-informed models, not previously applied to Mtb, allowed evaluation
of the relative nonsynonymous (dN) and synonymous (dS) substitution rates at individual codons.
Natural selection was evaluated by FUBAR, without compartmentilising Mtb sequences according to
HIV-1 status. Classification of resultant dN/dS values into functional gene categories revealed
differences across several categories. MEDS was then applied to evaluate relative differences in
positive selection in strains isolated from HIV-1 co-infected or uninfected individuals. The proportion
of codons showing evidence of relative differential positive selection was different in Mtb strains
isolated from HIV-1 co-infected individuals, including codons in virulence-associated and epitopeencoding genes. A lack of detailed clinical/epidemiological metadata meant that transmission
patterns and, therefore, the evolutionary history of the Mtb strains could not be determined. While
this could undermine the attribution of evolutionary differences to a particular environmental variable
(HIV-1 infection, in this case), MEDS has been shown to be relatively conservative and robust
against these uncertainties. Morevoer, the same differences were not observed in a control dataset
where HIV-1 status was randomly assigned to Mtb sequences, indicating that the data are not an
artifact of the evolutionary model applied.
These data suggest that patterns of evolution of Mtb may differ in HIV-1 co-infected individuals.
HIV-1-associated differences in epitope evolution may have important implications for the design of
Mtb vaccines intended for use in populations with high rates of HIV-1 infection. This work paves the
way both for validation of these findings on larger Mtb WGS datasets, and investigations of the
underlying causes of putative differential selection during HIV-1 co-infection.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
103
82
Mycobacterial Genomics and Evolution
20/09/2016
The true origins and evolution of tuberculosis
D.E. Minnikin
University of Birmingham, Birmingham, United Kingdom
Increasingly, genomic studies are being interpreted in various ways to suggest definitive
evolutionary pathways for tuberculosis (TB). However, such interpretations are often simply
correlations leading to unsupported estimates of the age of the disease, from 6ka to 70ka!
Considering practicalities, evolution from environmental Mycobacterium kansasii, through an
intermediate “Mycobacterium canettii” taxon, to Mycobacterium tuberculosis sensu stricto is a good
working hypothesis, supported by strong phylogenetic links in cell envelope lipid composition. The
transformation from “smooth” morphology “M. canettii” to “rough” M. tuberculosis correlates with
significant changes in cell envelope surface lipids1,2. Indeed in original studies1, it has been shown
that such differences in lipid profiles correspond to a dramatic change in overall cell hydrophobicity,
with M. tuberculosis being orders of magnitude more hydrophobic than “M. canettii”. Increased
hydrophobicity is closely linked with enhanced aerosol transmission and overall pathogenicity.
Although a coherent, plausible pathway for the bacterial aspects of tuberculosis evolution is
becoming clear, zoological and paleogeographical aspects need to be established. Using DNA
fragment amplification and lipid biomarker detection, tuberculosis in Homo sapiens has been
confirmed in skeletons from the “Fertile Crescent” back to 9-11ka BP1. Pleistocene evidence for
tuberculosis is principally confined to megafauna exhibiting specific metapodial lesions1. For a 17ka
bison metacarpal from Natural Trap Cave, Wyoming, tuberculosis was confirmed by detection of
strong lipid biomarkers and DNA amplification1. In ongoing studies, tuberculosis lipid biomarkers
have been detected in a ~40ka bison from Kent’s Cavern, Torquay and a range of ~12ka
mastodons from Buffalo, NY. Supported by micro-CT scanning, lesions are being pinpointed in
widespread bison metacarpals dating back to ~700ka. Such findings suggest a practical venue, in
megafaunal stomachs, for transforming environmental M. kansasii into “M. canettii”, the first
recognisable tubercle bacillus. Late Pleistocene dramatically enhanced hydrophobicity in tubercle
bacilli could well have produced megafaunal pandemics and extinction of bison and other
contempory taxa.
1. Minnikin et al. In: Ribón, W. (Ed), Tuberculosis –Expanding Knowledge. InTech-Open Access,
Rijeka, 2015 pp. 145-175; Tuberculosis 2015;95:S133-S139; Biofutur no 265, May 2015.
2. Boritsch et al. Nature Microbiology 2016;1:15019.
3. Rothschild & Martin Naturwissenschaften 2006;93:565-569.
104
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
83
Mycobacterial Genomics and Evolution
20/09/2016
Lineage-based analysis of Genomic Methylation and variations within Methyltransferases in
Mycobacterium tuberculosis Using Kinetic Data from Single-Molecule Real-Time (SMRT)
Sequencing Technology in Clinical Isolates
S. Modlin2, A. Elghraoui2, S. Hoffner1-2, F. Valafar2
1
Karolinska Institute, Stockholm, Sweden 2biomedical Informatics Research Center, San Diego
State University, San Diego, United States
Methylation is a type of epigenetic modification, the effects of which are poorly understood in
prokaryotes. It is well-known that many methyltransferases (MTases) combine with a cognate
restriction enzyme (RE) to create a primitive immune system termed restriction-modification (R-M)
systems. MTases that lack a cognate RE are termed “orphan” Mtases and are believed to serve
functions other than R-M-mediated immune defense. Recent work has revealed Mtb has at least
three MTases, of which two appear to be orphan Mtases. Using kinetic data, Pacific Biosciences'
SMRT -sequencing allows for detection of methylation at single nucleotide resolution concurrently
with SMRT-sequencing.
Methylation affects gene expression in Mtb in response to hypoxia, presumably allowing phenotypic
adaptation to the host-environment. Methylation-mediated phenotypic changes occur relatively
quickly allowing the bacterium to survive under the drug/immune system pressure without
chromosomal adaptation. Understanding how this capability differs within and between lineages
provides important information about lineage-specific adaptation towards resistance, persistence,
virulence, etc.
Using SMRT-sequencing we sequenced and de novo assembled genomes of 69 clinical isolates of
Mtb from 5 countries comprising 4 lineages. We compared activity of three known MTases by
examining methylation patterns in each isolate, and compared MTase sequences of each isolate to
a functional reference MTase to profile which mutations abrogated MTase activity, and which were
not deleterious. We analyzed how these were dispersed within and between lineages, revealing that
methylation activity of each MTase was consistent within lineages, but varied between them. We
found several missense mutations in each MTase, some of which abrogated methylation, while
others did not. MTase SNP patterns were also consistent within lineage, but different across
lineages, suggesting significant selective pressure on MTase functionality in Mtb. Future work will
examine methylated motif distribution in association with lineage and drug-resistance, to provide a
picture of which genes’ expression levels may be affected by methylation.
Funding: This work has been supported by grants from National Institute of Allergy and Infectious
Diseases (NIAID) grant R01AI05185 and National Science Foundation (NSF) grant 0966391.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
105
84
Mycobacterial Genomics and Evolution
20/09/2016
Mycobacterium tuberculosis population structure in Brazzaville, Republic of Congo –
identification of a new dominant H37Rv-like sublineage associated with transmission
S. Malm, L.S. Ghoma Linguissi, E.M. Tekwu, T.A. Kohl, P. Beckert, J.C. Vouvoungui, A. Sidibe,
S. Rüsch-Gerdes, I.K. Madzou- Laboum, S. Kwedi, V. Penlap Beng, M. Frank, F. Ntoumi,
S. Niemann
Molecular and Experimental Mycobacteriology Group, Research Center Borstel, Borstel, Germany
The Republic of Congo has a tuberculosis (TB) incidence of 381 cases/100,000 inhabitants and
thus is considered as a high TB burden country. Its population counts 4 million inhabitants and
approximately one fourth of the population agglomerates in its capital city, Brazzaville. Precise data
on the population structure and transmission dynamics of Mycobacterium tuberculosis complex
(MTBC) strains are key for effective TB control and have not been addressed for the Republic of
Congo yet. Thus, we characterized 74 MTBC strains isolated from patients with pulmonary TB in
Brazzaville by whole genome sequencing. The population diversity was high, with the majority of
strains belonging to the Euro-American lineage which split into LAM, Uganda I, Uganda II, Haarlem,
X-type and a new dominant sublineage termed the Congo-type (n = 26). Thirty strains were grouped
in 5 genome clusters with a maximum distance of 12 SNPs out of which 23 belonged to the newly
described Congo-type sublineage. Our study provides the first insights in the population structure of
MTBC strains in the Republic of Congo. High cluster rates and low genome diversities indicate
recent emergence and ongoing transmission of the Congo-type strains, a new Euro-American
sublineage.
106
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
85
Mycobacterial Genomics and Evolution
20/09/2016
Characterization of the Mycobacterial Integration Host Factor
N. Odermatt1, M. Lelli2, T. Herrmann2, N. Dhar1, S. Cole1
1
GHI, SV, UPCOL, EPFL, Lausanne 2ISA CRMN, Université de Lyon, Villeurbanne, Switzerland
The nucleoid associated protein (NAP) mycobacterial integration host factor (mIHF, rv1388) is one
of the most abundant proteins in Mycobacterium tuberculosis (Mtb). Although there is no sequence
similarity to the E. coli homologue, mIHF might not only be responsible for phage DNA integration,
but have a similar important role as IHF in E. coli, which acts as a global transcription factor and
DNA modelling protein.
We found that, in contrast to other NAPs, mIHF is present at the same high level in all growth
phases of Mtb. The coding sequence of the mihF gene is only about 60 % of the originally
annotated gene sequence, shrinking the protein to a size of 12 kDa. Upon construction of a
conditional knockdown mutant in Mtb, we were able to prove its essentiality and observed a severe
growth defect when mIHF was reduced to a level of below 10 % compared to the wildtype. Allowing
expression of mihF again, the batch culture recovered within three days and grew at a rate
comparable to the wildtype. Microfluidic analysis showed that depletion of mIHF led to cell death or
lysis, and in contrast to batch culture did not regain growth in permissive conditions.
To further understand the mode of DNA binding and gene regulation, we purified the mIHF protein,
which cannot be detected by UV absorption, and showed that it is not only highly soluble with a
concentration of up to 50 mg/ml but also very stable even at room temperature for several days. Its
structure, determined by nuclear magnetic resonance, showed a very high similarity to its
monomeric Streptomyces homologue, although we found some evidence that mIHF is forming
dimers under certain conditions.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
107
86
Mycobacterial Genomics and Evolution
20/09/2016
The First Population Structure and Comparative Genomics Analysis of Mycobacterium
africanum strains from Ghana Reveals Higher diversity of Lineage 5
I.D. Otchere1-2, S. Harris4, S.L. Busso4, A. Asante-Poku1, S. Osei-Wusu1, K. Koram1, J. Parkhill4,
S. Gagneux3, D. Yeboah-Manu1
1
Bacteriology, Noguchi Memorial Institute for Medical Research, University of Ghana 2Department
of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana 3Swiss TPH,
Basel, Switzerland 4Genome Campus, Wellcome Trust Sanger Institute, Cambridge University,
Cambridge, United Kingdom
Background: Mycobacterium africanum (MAF) remains an important TB causing pathogen in West
Africa however, little is known about its population structure and actual diversity which may have
implications for diagnostics and vaccines. We carried out comparative genomics analysis of
candidate Mycobacterium tuberculosis (MTB) and MAF using whole genome sequencing.
Methods: Clinical MTBC strains (187) comprising L4 (22), L5 (126) and L6 (39) isolated over 8
years from the Northern and Southern parts of Ghana were whole genome sequenced. The reads
were mapped unto a reference genome for phylogenetic and functional genomics analysis.
Maximum Likelihood tree with 100 bootstraps was constructed from the SNPs called using RAxML.
A total of 147 (18 L4, 36 L6 and 93 L5) of the genomes were de novo assembled and annotated for
comparative pan-genome analysis using Roary.
Results: The population structure of MAF showed at least 10 sub-lineages of L5 and 3 under L6.
There were year specific clusters of L5 in 2013, 2012 and 2008. We also identified a cluster of 3
MDR L5 strains from Southern Ghana in 2013. Among the global collection of MTBC, there were
two Ghana-specific L5 clusters of which one exhibited clonal expansion. From the 5947 pan genes
extracted from the collection, 3215 (54.1%) were core to all the 147 genomes whereas 719 (12.1%)
were found in single genomes. We identified the absence of some unique genes among specific
lineages and/or clades with possible clinical implications. For example, mpt64 and mlaD encoding
respectively an immunogenic protein and a mammalian cell entry protein were missing from all L6
genomes. Analysis of SNPs within some genes encoding proteins for substrate metabolism, ion
transport and secretory systems showed higher proportion of SNPs among L6 compared to L5 and
L4. We also identified a number of Lineage/sub-lineage specific SNPs that may be utilized in rapid
PCR based genotyping of MTBC.
Conclusion: Contrary to the notion that MAF is an ancient species and may show less diversity as
compared to MTBSS, MAF especially L5 displayed an extensive diversity similar to what is known
about L4. The diversity observed has implications for current and future diagnostic and vaccine
prospects.
108
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
87
Mycobacterial Genomics and Evolution
20/09/2016
Understanding the regulation of cytochrome P450s: core enzymes involved in M.
tuberculosis metabolism
A. Otter, S. Kendall
Dept. of Pathology & Pathogen Biology, Centre for Emerging, Endemic and Exotic Diseases, Royal
Veterinary College, London, United Kingdom
The genome of Mycobacterium tuberculosis encodes for 161 helix-turn-helix DNA binding
transcriptional regulators. The most abundant of these (32%) belong to the TetR family. This family
of regulators were first described in Escherichia coli and are best known for their control of efflux
pumps. However, more recent understanding derived in both Streptomyces and mycobacteria
indicates that this family are frequently regulators of oxidoreductases.
In M. tuberculosis and Streptomyces, 36-40% of genes located next to TetR regulators are
oxidoreducatase enzymes and not membrane proteins as previously thought, with membrane
proteins only representing 10-24% of TetR associated genes.. These insights from both genomes
suggest that the majority of TetR regulators may be crucial regulators of metabolic pathways [1, 2].
In M. tuberculosis, TetRs have been identified to control enzymes required for branched chain
amino acid catabolism and cholesterol catabolism, a pathway that has an established role in
virulence [3-5].
TetR regulators often regulate nearby genes and bioinformatic analysis of the M. tuberculosis
genome shows that 11 TetR regulators are within close proximity to gene(s) encoding Cytochrome
P450s (CYPs). CYPs are absent in many bacteria, but mycobacteria are an exception, with 20 CYP
genes located throughout the genome of M. tuberculosis [6]. CYPs have been found to play key
roles in metabolism, sterol transformation and synthesis of host lipids, providing attractive drug
targets as previous research has shown antifungal drugs, particularly azole based compounds, to
be potent CYP inhibitors. Though numerous CYPs have been studied for their function and role in
M. tuberculosis, very little is known about their regulation.
To further understand TetRs and their role in CYP regulation, we are expressing and purifying
candidate TetRs to demonstrate binding to CYP promoters, identifying regulatory motifs within CYP
promoters using MEME and searching for motifs on a genome wide scale using FIMO. In the future
we will also make deletion mutants of both the TetRs and CYPs in order to better understand their
regulation and physiological role in M. tuberculosis.
1. Ahn SK, Cuthbertson L, Nodwell JR: Genome Context as a Predictive Tool for Identifying
Regulatory Targets of the TetR Family Transcriptional Regulators. PLoS One 2012, 7(11).
2. Balhana RJC, Singla A, Sikder MH, Withers M, Kendall SL: Global Analyses of TetR Family
Transcriptional Regulators in Mycobacteria Indicates Conservation across Species and Diversity
in Regulated Functions. BMC Genomics 2015, 16.
3. Balhana RJ, Swanston SN, Coade S, Withers M, Sikder MH, Stoker NG, Kendall SL: bkaR is a TetRtype Repressor That Controls an Operon Associated with Branched-Chain Keto-Acid
Metabolism in Mycobacteria. FEMS Microbiology Letters 2013, 345(2):132-140.
4. Kendall SL, Burgess P, Balhana R, Withers M, Ten Bokum A, Lott JS, Gao C, Uhia-Castro I, Stoker NG:
Cholesterol Utilization in Mycobacteria is controlled by Two TetR-Type Transcriptional
Regulators: kstR and kstR2. Microbiology 2010, 156(Pt 5):1362-1371.
5. Kendall SL, Withers M, Soffair CN, Moreland NJ, Gurcha S, Sidders B, Frita R, Ten Bokum A, Besra
GS, Lott JS et al: A Highly Conserved Transcriptional Repressor Controls a Large Regulon
Involved In Lipid Degradation in Mycobacterium smegmatis and Mycobacterium tuberculosis.
Molecular Microbiology 2007, 65(3):684-699.
6. McLean KJ, Belcher J, Driscoll MD, Fernandez CC, Le Van D, Bui S, Golovanova M, Munro AW: The
Mycobacterium Tuberculosis Cytochromes P450: Physiology, Biochemistry & Molecular
Intervention. Future Medicinal Chemistry 2010, 2(8):1339-1353.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
109
88
Mycobacterial Genomics and Evolution
20/09/2016
Comparative genomic analysis of hypervirulent Mycobacterium tuberculosis strains and
closely related hypovirulent strains reveals novel mutations associated with enhanced
growth during macrophage infection
R. Rajwani, G. Siu, W.C. Yam
Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
Background: Among clinical strains of Mycobacterium tuberculosis (MTB), the relationship
between genetic diversity and disease severity remains inadequately understood which hinders
development of new and improved vaccine against adult tuberculosis (TB). We had previously
isolated two hypervirulent MTB strains (H107 and H112) from immunocompetent patients suffering
from tuberculous meningitis. Both strains demonstrated significant higher bacillary growth in ThP1
macrophage and lower T-cell helper cell 1 (Th1) response when compared to virulent reference
strain M. tuberculosis H37Rv and other ordinary pulmonary clinical strains. Here, we describe the
whole genome sequencing results of the hypervirulent strains in comparison with other two
hypovirulent pulmonary strains (H54 and H83) of same lineage.
Methods: Genomic DNA was extracted from late-log phase cultures of MTB strains. 20-Kb single
molecule real-time (SMRT)-bell libraries were prepared and sequenced using P6-C4 chemistry. The
resulting reads with an average length of 8Kb were assembled through hierarchical genome
assembly process (HGAP). Fully closed circular genomes were obtained for all strains which were
annotated using Glimmer3. For genome comparisons, raw reads were aligned against reference
Mycobacterium tuberculosis H37Rv genome (refseq accession no. NC_000962.3) and variants
were called using Quiver. Variants were annotated using ensembl variant effect predictor (VEP).
Further analysis and comparisons were performed in SAMtools./BCFtools. Literature was reviewed
to identify variants associated with hypervirulence.
Result: A total of 1297 single nucleotide polymorphisms and 1004 indels that are specific to
hypervirulent strains were identified, in which two frame-shift deletions and seven missense variants
in genes previously shown to confer hypervirulence in MTB. These include mutations in mammalian
cell entry 1 (mce1) operon including a frameshift deletion in mce1D (H107) and misense mutations
in lipoprotein LprK (H107) and integral membrane protein YrbE1B (H112). We also identified a
glutamic acid to lysine amino acid substitution in sensor kinase domain of KdpD (H112).
Conclusion: The current study identifies possible mechanisms of enhanced virulence in H107 and
H112. In our laboratory, we are developing allele complementation based functional assays to prove
that these variants are indeed the underlying reason of hypervirulence in these strains.
110
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
89
Mycobacterial Genomics and Evolution
20/09/2016
M. tuberculosis whole genome sequence analysis reveals cell wall-associated genes as
targets of convergent evolution in tuberculous meningitis
C. Ruesen4, L. Chaidir2, A. Van Laarhoven4, S. Dian1, A. Rizal Ganiem1, M. Huijnen3,
B. Alisjahbana2, B. Dutilh3, R. Van Crevel4
1
Department of Neurology, Faculty of Medicine, Padjadjaran University/Hasan Sadikin
Hospital 2Health Research Unit, Faculty of Medicine, Padjadjaran University/Hasan Sadikin
Hospital, Bandung, Indonesia 3Centre for Molecular and Biomolecular Informatics, Radboud
university medical center 4Department of Internal Medicine, Radboud university medical center
Nijmegen, Nijmegen, The Netherlands
Background: Meningitis is the most severe manifestation of tuberculosis. It is largely unknown why
certain people develop pulmonary TB (PTB) and others TB meningitis (TBM), but we hypothesize
that genetic diversity of infecting M. tuberculosis strains plays a role.
Methods: M. tuberculosis strains isolated from HIV-negative patients, 102 from cerebrospinal fluid
of TBM patients, and 176 from sputum of PTB patients were whole-genome sequenced using
Illumina HiSeqTM2000 and aligned to the reference genome M. tuberculosis H37Rv. A maximum
likelihood phylogenetic tree was constructed based on multiple alignment of the common variable
positions. M. tuberculosis genetic variation was linked to clinical TB phenotype by looking for
phylogenetic convergence: the repeated appearance of phenotype-related changes at specific loci
or genes in phylogenetically unrelated branches. Enrichment scores for the TB phenotype with
associated p-values were calculated for each SNP. Candidate SNPs were validated in an
independent set of isolates.
Results: Strains belonged to the East-Asian Beijing lineage (35.6%), East-Asian non-Beijing
lineage (0.7%), Euro-American lineage (61.2%), and Indo-Oceanic lineage (2.5%). We found no
association between lineage and phenotype (Chi-square = 5.095; p=0.165). Large genomic
differences were observed between the isolates; the minimum genetic distance varied from 5 to 952
SNPs. The phylogenetic tree, based on 25198 common variable positions, revealed 104 isolates in
terminal branch pairs with distinct phenotypes, with 38 to 544 SNPs difference between paired
isolates. Mutations in Rv1396c (PE-PGRS25) and Rv1899c (LppD) were enriched for TBM in paired
isolates (p=0.025 and p=0.045 respectively), and validated in the set of 174 unpaired isolates
(p=0.025 and p=0.011).
Conclusion: Using phylogenetic convergence analysis we identified mutations in 2 possibly
immunoregulatory genes that might be important in TBM pathogenesis. These cell wall-associated
genes play a role in host-pathogen interaction, possibly through pathogen recognition and antigen
presentation. Further validation may come from independent cohorts, or experimental or in-vitro
studies.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
111
90
Mycobacterial Genomics and Evolution
20/09/2016
Dynamics of Bovine Tuberculosis Disease Transmission in Egypt: The “Real” Jewel of the
Nile
A. Talaat, H. Abdelaal
Pathobiological Sciences, University of Wisconsin-Madison, Madison, United States
Bovine tuberculosis (BTB) is a chronic infectious disease characterized by the formation of
granulomatous lesions in organs, mainly lungs and lymph nodes. BTB is caused by slowly growing
bacilli, mainly, M. bovis. In the developing countries, BTB represent a major problem with a
prevalence that could reach up to 10-15% of cattle herds in some parts of Africa. In Egypt, BTB
transmission among animals and from animals to humans represents a major problem because of
the complexity of animal husbandry and animal trade. In this project, our teams in both USA and
Egypt used Single Intradermal Comparative Skin Test (SICST) and whole genome sequence
analysis (WGSA) to examine the dynamics of BTB transmission among 5 different regions within the
Nile Delta of Egypt. During the years of 2012-2015, several farms were visited and M. bovis isolates
from the same herds were analyzed. As expected, farms with a known history of risky management
behavior were associated with high level of BTB prevalence that reached up to 45% in some herds.
Interestingly, WGSA was able to trace dynamics of BTB transmissions among herds when source
and timing of infection were verified with available historical data. Moreover, isolates belong to M.
bovis BCG lineage or those with drug resistance phenotypes were identified. In this presentation,
we will discuss the key outcomes of this project highlighting the importance of ecological combined
with genomic (EcoGenomic) analyses of BTB, especially in enzootic countries. Currently, we are
investigating the virulence of M. bovis isolates with different genomotypes.
112
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
91
Mycobacterial Genomics and Evolution
20/09/2016
Prevalence of PZase, PanD, and RpsA mutations in multidrug- and extensively-drug
resistant Mycobacterium tuberculosis Clinical Isolates
S. Ramirez-Busby2, L. Fink2, T. Rodwell3, M. Pettigrove3, L. Jackson3, D. Catanzaro1,
A. Goodmanson2, A. Amallraja2, R. Shanmugan2, A. Catanzaro3, F. Valafar2
1
Department of Biology, University of Arkansas, Fayetteville 2Biomedical Informatics Research
Center, San Diego State University 3Medicine, University of California, San Diego, San Diego,
United States
Pyrazinamide (PZA) is an important first-line drug recommended by the World Health Organization
(WHO) and included in all new treatment regimens to treat the highly infectious M. tuberculosis.
However, prevalence of resistance to PZA continues to rise while reliable phenotyping has yet to be
established. Three tests were performed on our isolates: DST, Wayne’s enzymatic assay, and
whole-genome sequencing. Coding and promoter regions of three genes—pncA, rpsA, panD—were
investigated for markers of PZA resistance. Enzyme activity was used when there was phenotypegenotype discordance. Two hundred and thirty-five resistant (PZAR) and 80 susceptible (PZAS)
isolates were included in this study. High concordance was observed between DST and enzymatic
assay but less so with the genotype. 86% of PZAR and 30% of PZAS isolates had a mutation in
PZase or its promoter. Fourteen percent of PZAR isolates lacked a genetic explanation for the
observed resistance phenotype. In our population, only 6 (2%) PZAR isolates did not have a PZase
mutation but had a mutation in RpsA or PanD. Assuming that all RpsA and PanD mutations cause
resistance, the combination of PZase, RpsA, and PanD potentially offer a combined sensitivity of
88% for prediction of PZA resistance phenotype. Overall, this highlights the need for discovery of
alternative mechanisms of resistance for PZA, at least in resistant isolates that do not have a PZase
mutation.
Funding: This work has been supported by grants from National Institute of Allergy and Infectious
Diseases (NIAID) grant R01AI05185 and National Science Foundation (NSF) grant 0966391.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
113
92
Mycobacterial Genomics and Evolution
20/09/2016
MIRU-Heuristics for Evaluation of Repeats and their Ordinal (MIRU-HERO): MIRU analysis
on Genomic Sequencing Data
L. Fink, F. Valafar
biomedical Informatics Research Center, San Diego State University, San Diego, United States
A technique regularly used by public health agencies to distinguish between strains of
Mycobacterium tuberculosis (Mtb) in an outbreak is MIRU-VNTR typing. While the technique is
relatively fast, it does not offer the resolution that whole genome sequence (WGS) analysis
provides. Since WGS technologies are quickly becoming more accessible and less costly, it is
probable that they will eventually be commonly used in contact tracing and analysis of Mtb
outbreaks. In an effort to correlate the public health efforts of the PCR era to those of the WGS era,
MIRU-HERO has been developed as a stand-alone, command-line accessible program for Linuxbased systems to quickly and efficiently analyze 24-locus MIRU-VNTR data in reference-based or
de novo-assembled genomes. The benefits of this system are three-fold: privacy, transparency and
resolution.
MIRU-HERO uses a set of subject MIRU-VNTR sequences to BLAST against a query genome in
fasta format. Sequences in the genome which match up to 97% identity of a subject sequence are
appended to a list. Sequential matches from the list are correlated to a MIRU region, and the results
are output to a file including the miru name, number of copies detected, and the specific regions of
detection in the genome (denoted by base pair position number).
The program was run successfully on a set of 75 in-house denovo-assembled genomes with 97.6%
correlation to laboratory conducted MIRU-typing.
MIRU-HERO is both functional and applicable in the determination of MIRU-VNTR information for
mycobacterial genomes. MIRU-HERO resolves privacy issues that web-based programs face as it
is a locally installed program not needing to transmit sensitive medical information over the internet.
Additionally, compared to conventional PCR based approaches in MIRU typing, there is an
increased level of detail which can be used to resolve differences between mycobacterial strains.
Funding: This work has been supported by grants from National Institute of Allergy and Infectious
Diseases (NIAID) grant R01AI05185 and National Science Foundation (NSF) grant 0966391.
114
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
93
Mycobacterial Genomics and Evolution
20/09/2016
Whole genome phylogenetic analysis of clinical isolates from high-burden countries
C. Chan2, R. Shanmugan2, S. Hoffner1, F. Valafar2
1
Karolinska Institute, Stockholm, Sweden 2biomedical Informatics Research Center, San Diego
State University, San Diego, United States
Lineage-specific differences in MTBC play an important role in virulence and emergence of drugresistance. Most recent research indicates that genome-wide single nucleotide polymorphisms
(SNPs) are good markers for lineage analysis compared to previous systems (like spoligo/MIRU).
This study includes 368 mostly MDR/XDR clinical isolates collected from India, Moldova, South
Africa, the Philippines, and 7 mono-PZA resistant isolates from Sweden. We used Pacific
Biosciences’ single molecule sequencing technology for our WGS approach. Based on our analysis,
we identified 36,634 unstable loci in Mtb genomes that could harbor a SNP. Nearly 12K of these
positions are more prone to indicate heterogeneity. RAxML was used to create a high-resolution
phylogenetic tree. Using the clustering patterns obtained from our tree, five isolates were identified
as discordant with lineages predicted using MIRU12/spoligo patterns. Bootstrap values for these
isolates were high indicating high-confidence grouping, and their lineages confirmed using PhyTB.
Using our tree, we were also able to resolve some sub-lineages within the four discovered lineage
groups. Ancestral reconstruction of SNPs identified 6247 potential lineage markers and 498 genes
under convergent selection. Comparison of our lineage-associated SNPs with those determined by
Coll et al. (2014), identified 2028 common SNPs, while 11 SNPs were associated with different
lineages than those reported by Coll et al. Six of the 11 SNPs are harbored by PE/PPE genes. 236
SNPs identified by Coll et al. as associated with a single lineage were associated with multiple
lineages in our isolates. On examining convergent evolution patterns in the tree, it was noted that
almost a third of the SNPs indicative of convergent evolution occurred within PE/PPE genes. This
suggests the constantly changing nature of the PE/PPE gene family and the need for further
investigation of PE/PPE genes, which may only be elucidated using sequencing with longer reads.
Funding: This work has been supported by grants from National Institute of Allergy and Infectious
Diseases (NIAID) grant RO1AI05185 and National Science Foundation (NSF) grant 0966391.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
115
94
Mycobacterial Genomics and Evolution
20/09/2016
Current perspectives on the glycoside hydrolases of Mycobacterium tuberculosis: their
importance and prospects on assigning function to unknowns
N. Van Wyk3, L. Kremer3-4, M. Drancourt2, B. Henrissat1-5
1
Architecture et Fonction des Macromolécules Biologiques (AFMB), Architecture et Fonction des
Macromolécules Biologiques (AFMB), UMR 7257 CNRS, Université Aix-Marseille 2Unité de
Recherche sur les Maladies Infectieuses et Tropicales Emergentes, Centre National de la
Recherche Scientifique (CNRS), Institut de Recherche pour le Développement, Faculté de
Médecine, Aix-Marseille Université, Marseille 3Centre d'études d'agents Pathogènes et
Biotechnologies pour la Santé (CPBS), Centre National de la Recherche Scientifique FRE3689,
Université de Montpellier 4INSERM, Montpellier, France 5Department of Biological Sciences, King
Abdulaziz University, Jeddah, Saudi Arabia
Glycoside hydrolases (GHs) are the enzymes that catalyze the hydrolysis of glycosidic bonds in
glycoconjugates, oligo- and polysaccharides. A classification of these enzymes based on conserved
sequence and structure motifs initiated by the Carbohydrate Active Enzyme (CAZy) database has
proven useful in the systematic groupings of similar enzymes into families. The human pathogen
Mycobacterium tuberculosis employs 30 GHs to perform a variety of different functions which can
be divided into four broad categories: alpha-glucan metabolism, peptidoglycan remodelling, betaglycan hydrolysis and α-demannosylation. The poster presented here provides an overview of how
the GHs that have been characterized play a role in each category. Expanding the genomic analysis
of GH presence to other Mycobacterium species has highlighted the importance of certain families –
most notably GH13 and GH23 – in the general genomic make-up of mycobacteria. Since many GHs
are still uncharacterized and considered as “conserved hypothetical”, the grouping of them into
respective families provides a strong prediction on their putative biological functions.
116
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
95
Mycobacterial Biology & Physiology
20/09/2016
Development of an innovative Point-Of-Care (POC) device for Tuberculosis Diagnosis
E. Ressami2, B. Lakssir2, M. Abid3, H. Ait Benhassou1
1
Medical Biotechnology Center 2Microelectronics Packaging Center, Moroccan Foundation for
Advanced Science, Innovation and Research, Rabat 3Laboratoire de Génétique Mycobactérienne,
Institut Pasteur du Maroc, Tangier, Morocco
Tuberculosis (TB) is one of the deadliest infectious diseases worldwide. It is caused by bacteria
called Mycobacterium tuberculosis that infects a new person every second in the world. Each year,
about 1% of the world population is infected and about 9 million people develop the disease,
resulting in approximately 2 million deaths per year.
In Morocco, as in most developing countries in North Africa and the Middle East, where the timely
and accurate diagnosis of TB remains a great challenge, TB constitutes a major public health threat
with 30,000 new cases each year. Recently, Morocco and other countries in the MENA region have
become a primary destination of new waves of refugees from Syria and Sub-Saharan countries,
which increased the difficulty in the control and the management of TB. Currently, the conventional
methods routinely used to diagnose TB such as smear microscopy and bacterial culture are
complex, unreliable, technically challenging and time-consuming at point-of-care settings.
The goal of this project is to contribute in reinforcing the implemented national and regional public
health strategy in the effective management of tuberculosis among local populations, Syrian and
Sub-Saharan refugees living in Morocco and in the MENA region. Indeed, through this proposal, we
aim to reduce susceptible and resistant tuberculosis transmission, morbidity, and mortality by
enabling early TB screening and diagnosis.
The idea of the project is to establish an innovative platform combining new advanced high-tech
approaches, tools and techniques in Molecular biology, Microelectronics and Photonics. Thus, we
developed, a mobile phone-based, easy-to-use readout device to perform a LAMP (< $US 2.0) in
less than 15 min reaction. This LAMP assay have several advantages that are relevant and
attractive as a diagnostic platform for resource-poor settings: it is specific, highly sensitive, fast and
generates a result that can be directly detected by an automated phone camera and remotely
transmitted to clinical centers.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
117
96
Mycobacterial Biology & Physiology
20/09/2016
DrrS, a DosR regulated small RNA in Mycobacterium tuberculosis
A. Moores3, A. Riesco3, S. Waddell1, D. Young2, K. Arnvig3
1
University of Sussex, Brighton 2Francis Crick Institute 3Structural and Molecular Bilogy, University
College London, London, United Kingdom
M. tuberculosis (Mtb) depends on an ability to adjust to stresses delivered by a range of host
environments, adjustments that require significant changes in gene expression. Small RNAs
(sRNAs) play an important role as post-transcriptional regulators of prokaryotic gene expression,
where they are associated with stress responses and, in the case of pathogens, adaptation to the
host environment.
Our aim is to elucidate this aspect of Mtb gene regulation by applying a multidisciplinary approach
complementing an RNA-seq systems level analysis with more traditional techniques including
Northern blotting, qRT-PCR and 5’/3’ RACE. We have previously identified several sRNAs in Mtb,
and their sheer abundance, and the fact that many are part of pathogenesis-associated regulons
suggest that they make a significant contribution to the regulation of gene expression during
infection. This study is focussed on the sRNA DrrS, which is regulated by the dormancy master
regulator DosR. We have investigated DrrS expression under different growth conditions and used
reporter gene assays to define its promoter elements. We have found that DrrS is highly stable and
generated by rapid processing of a longer transcript, DrrS+. Moreover, we have investigated the
turnover of the mature DrrS and identified RNA determinants involved in its extraordinary stability. In
order to elucidate the biological function of DrrS, we have combined reverse genetics with
microarray analysis and identified >200 genes that are differentially expressed in a ΔdrrS strain
compared to the parental Mtb H37Rv with significant enrichment of genes belonging to information
pathways. By subsequently interrogating this data using in silico target prediction, we have identified
a small subset of genes that are potentially direct targets of DrrS; among these are genes
associated with information pathways, amino acid biosynthesis and two major secretion pathways in
Mtb.
In summary, our study is a comprehensive analysis of the Mtb sRNA, DrrS in terms of expression,
processing, stability and target identification, which makes an important contribution to our growing
knowledge of Mtb RNA biology.
118
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
97
Mycobacterial Biology & Physiology
20/09/2016
The identification of novel proteins involved in iron-sulphur cluster biogenesis in
mycobacteria
J. Arries, S. Sampson, R. Warren, M. Williams
Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
Mycobacterium tuberculosis remains a public health threat. Given the emergence of drug
resistance, it is imperative that we improve our understanding of the pathogenesis of Mtb, in order
to develop new anti-TB drugs and strategies for shortening drug treatment.
Iron-sulphur clusters are ubiquitous cofactors required for the maturation of various proteins, many
of which are involved in essential biological processes. Multiprotein complexes are required for the
in vivo assembly of Fe-S clusters, and the SUF system, encoded by the Rv1460-Rv1461-Rv1462Rv1463-csd-Rv1465-Rv1466 operon in Mtb, is thought to be the major Fe-S cluster biogenesis
machinery. This process is poorly understood in mycobacteria, and it is currently unclear if proteins
outside of this operon are involved in Fe-S cluster biogenesis.
In this study, we sought to identify novel proteins involved in Fe-S cluster assembly. We employed
affinity purification and mass spectrometry to identify proteins that interact with the SUF machinery.
Utilising this method, we were able to demonstrate affinity enrichment for Fe-S cluster assembly
proteins, and Fe-S cluster-containing enzymes, as well as identify potential novel interacting
partners. This work has established the methodology for identifying novel protein-protein
interactions, and lays the foundation for elucidating the process of Fe-S cluster assembly in
mycobacteria.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
119
98
Mycobacterial Biology & Physiology
20/09/2016
Ergothioneine is essential for Mycobacterium tuberculosis growth during starvation and
long term infection
M. Richard-Greenblatt, A. Steyn, H. Bach, Y. Av-Gay
Infection and Immunity Research Centre, University of British Columbia, Vancouver, Canada
Ergothioneine (EGT), an antioxidant obtained from the diet in mammalian systems, is synthesized
by M. tuberculosis (Mtb). EGT is derived from histidine and contains sulfur bound to its imidazole
ring, imparting unique chemical properties to this amino acid. Unlike other sulfur-containing
molecules, such as the low molecular weight (LMW) thiol, and glutathione (GSH), EGT exists
primarily as a thione under physiological conditions. The chemical structure of EGT, which provides
the molecule with enhanced stability to oxidation. Nonetheless, the knockdown of the EGT
transporter in mammalian cells leads to augmented oxidative damage and cell death in the
presence of exogenous stressors.
Similar to other microorganisms, the physiological role and regulation of EGT biosynthesis in Mtb is
unknown. We identified an Mtb gene (Rv3701c) to encode for EgtD, the enzyme that catalyzes the
methylation of the amino acid histidine. Through the generation of an ΔegtD mutant we found that
this methyltransferase is essential for EGT biosynthesis. We further demonstrated that EgtD activity
is regulated by an Mtb protein-serine/threonine kinase in response to nutrient starvation. Under
starvation, the Mtb ΔegtD mutant was unable to maintain viability compared to its parental wild type
strain. As, starvation is associated with induction of a non-replicative state in Mtb, these findings
indicate that EGT plays a role in mediating persistent infection or disease latency. Indeed,
macrophage infection showed minimal impact on the survival of the ΔegtD mutant compared to its
parental strain during initial stages (replicative phase) of intracellular infection. Further metabolic
analysis identified Mtb intracellular EGT levels to be directly correlated with carbon source type and
availability pointing to a role for EGT in energy storage.
120
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
99
Mycobacterial Biology & Physiology
20/09/2016
Identifying mechanisms for environmental adaptation in CMNR bacteria: a systems
analysis of gene regulation of alternative substrate utilization and cell envelope
biosynthesis
R. Bailo Vergara1, E. Peterson2, A. Singh1, N. Baliga2, A. Bhatt1
1
IMI - Institute of Microbiology and Infection, University of Birmingham, Birmingham, United
Kingdom 2Institute for Systems Biology, Seattle, United States
The genera Corynebacterium, Mycobacterium, Nocardia, and Rhodococcus form the so-called
CMNR group that belongs to the order Actinomycetales. The core common characteristic of this
group is the presence of a waxy cell envelope which is mainly composed of peptidoglycans,
arabinogalactans, and mycolic acids. CMNR bacteria are pathogens, soil inhabitants, plant
commensals, and industrial microbes that are of pharmaceutical, environmental, and
biotechnological interest.
The diversity of habitats colonized by CMNR bacteria is based on their ability to adapt to new
environments by dynamically altering the composition of mycolic acids in their cell envelope. The
hypothesis of our research is that depending on the environment and available substrates, the
CMNR bacteria selectively alter the composition of their cell envelope by using expanded families of
uptake and biosynthesis enzymes in different combinations.
Our initial experiments are being carried out using M. smegmatis because it is a model organism
that maintains the unique and complex cell envelope organization that is common to the CMNR
bacteria. Later, we will use other model representatives of this group such as Corynebacterium
glutamicum, Rhodococcus jostii RHA1. So, firstly, we are phenotypically characterising changes in
the cell envelope composition of M. smegmatis during growth on ecologically-relevant substrates
and environments monitoring the OD600 and analysing its lipid content by 2-D chromatography. We
will then characterise the growth dynamics and cell envelope changes during transitions between
selected growth conditions. Once we have revealed those transitions in which M. smegmatis
underwent significant changes in its cell-envelope composition, we will measure genome-wide gene
expression changes over a time course during these adaptations by RNA-seq. The compendium of
transcriptome measurements along with publically available gene expression data from microarray
experiments will be used to generate our gene regulatory network model. Finally, we will develop
algorithms to interrogate the model to discern modular architecture (i.e., conditional co-regulation of
specific combinations of genes) and topology (i.e., promoter architecture and regulatory influences
of transcription factors) that are conserved or unique across the CMNR bacterial genomes. Model
predictions will be tested by analysing consequences of the deletion of specific genes related to the
cell envelope composition of representative CMNR bacteria under relevant environmental
transitions.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
121
100 Mycobacterial Biology & Physiology
20/09/2016
Understanding the role of malic enzyme in Mycobacterium tuberculosis infections
P.S. Basu, R. Balhana, I. Gobe, D. Beste
Microbial Sciences, FHMS, University of Surrey, Guildford, United Kingdom
The PEP-pyruvate-oxaloacetate or anaplerotic node represents a critical metabolic cross-road for
central metabolism controlling the distribution of flux between anabolism, catabolism and energy
supply to the cell. In Mycobacterium tuberculosis this node consists of the enzymes PEP
carboxykinase (PCK), pyruvate carboxylase (PCA), malic enzyme (MEZ) and pyruvate phosphate
dikinase (PPDK). Aside from PCK these enzymes remain uncharacterised. Here we show that mez
encodes a functional malic enzyme which has a high affinity for malate and therefore preferentially
functions in the gluconeogenic direction, however this enzyme can also function glycolytically. M.
tuberculosis strains deficient in MEZ produce small shiny colonies with a very sticky texture
suggesting changes in the cell wall lipids. MEZ was not required for growth on a range of glycolytic
and gluconogenic substrates in vitro. However, we demonstrate that MEZ in collaboration with
PPDK provide M. tuberculosis with an alternative gluconeogenic route in the absence of PCK.
Although MEZ was not required for intracellular growth of M. tuberculosis in macrophages this strain
was attenuated for the initial invasion. We are currently investigating the hypothesis that that this is
due to alterations in the cell wall lipids. Our studies further demonstrate the metabolic plasticity of
central carbon metabolism in M. tuberculosis.
122
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
101 Mycobacterial Biology & Physiology
20/09/2016
Aspartate amino acid biosynthesis in Mycobacterium tuberculosis - characterization of a
new drug target space
M. Berney1, L. Berney-Meyer1, E. Hasenoehrl1, D. Rae Sajorda1, W.R. Jacobs1-2, F. Wang2
1
Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx 2California
Institute for Biomedical Research (CaliBR), La Jolla 3Howard Hughes Medical Institute, Maryland,
United States
A major constraint for developing new TB drugs is the limited target space. We are in the process of
exploiting a largely untapped drug target space that is unique to M. tuberculosis. Intracellular
pathogens are faced with a host-induced process called nutritional immunity that depletes essential
nutrients from the pathogen-containing vacuole. Emerging evidence suggests that many
intracellular pathogens manipulate the phagocytes to regain access to those nutrients. In contrast,
M. tuberculosis is not able to scavenge essential amino acids in the host and relies on its own
biosynthetic machinery. We have recently shown that inactivation of methionine and Sadenosylmethionine biosynthesis leads to unusually rapid culture sterilization and complete
clearance from host tissues (Berney et al., 2015, PNAS). The underlying unprecedented multi-target
inhibition mechanism is optimal for drug development. We have now expanded our focus to
characterize the regulation and metabolic flux in the whole aspartate amino acid biosynthesis
pathway. Using metabolomics, transcriptomics, animal experiments and conditional knockdowns we
could identify several new vulnerable enzymes in the aspartate pathway that have the potential to
become new drug targets.
Berney, M., Berney-Meyer, L., Wong, K.W., Chen, B., Chen, M., Kim, J. et al. (2015) Essential roles
of methionine and S-adenosylmethionine in the autarkic lifestyle of Mycobacterium tuberculosis.
Proc Natl Acad Sci U S A 112: 10008-10013.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
123
102 Mycobacterial Biology & Physiology
20/09/2016
13
C isotopomer analysis identifies an essential pathway for the intracellular survival of
Mycobacterium tuberculosis
P.S. Basu2, K. Noh1, R. Balhana2, T. Mendum2, I. Gobe2, J. Mcfadden2, D. Beste2
1
Institute of Bio- and Geosciences 1: Biotechnology 2, Forschungszentrum Jülich, Jülich, Germany
2
Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
Metabolic reprogramming in response to the stresses of the host niche during both the acute and
chronic phase of tuberculosis infections is a crucial determinant of virulence. Experimental evidence
has identified central carbon metabolism as instrumental in this pathogenic strategy. Using mutant
strains, 13C isotopomer analysis and mathematical modelling we demonstrate that intracellular M.
tuberculosis uses a novel pathway which requires the enzymes phosphate dikinase (PPDK), PEP
carboxykinase (PCK). Disruption of this pathway impacts on the intracellular survival of M.
tuberculosis. Strains deficient in either pyruvate phosphate dikinase (PPDK) or PEP carboxykinase
(PCK) are similarly attenuated for intracellular growth and M. tuberculosis lacking both pyruvate
carboxylase (PCA) and PCK do not survive within human macrophages. As PCA, PPDK and PCK
were also required for optimal survival in the presence of reactive nitrogen intermediates we
hypothesise that this pathway generates pyruvate to thwart nitrosative stress and generates the
high-energy metabolite phosphoenolpyruvate (PEP) which allows for ATP synthesis via substratelevel phosphorylation. This work identifies a novel pathway required for intracellular survival which
could be targeted with therapeutics.
Funding: MRC -MR/K01224X/1.
124
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
103 Mycobacterial Biology & Physiology
20/09/2016
Proteomic Analysis of Mycobacterium tuberculosis Membrane Vesicles
A. Birhanu2, S. Yimer1, T. Riaz1, T. Tønjum1-2
1
Microbiology, Oslo University Hosiptal 2Microbiology, University of Oslo, Oslo, Norway
Secreted membrane vesicles (MVs) are abundant in Mycobacterium tuberculosis (Mtb) and are
suggested to have important roles in mycobacterial physiology and pathogenesis. MVs may contain
varied cargo, including nucleic acids, toxins, lipoproteins and enzymes. In particular, MVs have
been shown to serve as a vehicle for the selective packaging and release of virulence factors, such
as toxins and immunomodulatory molecules. However, how and why MVs escape the thick cell
walls of mycobacteria is still unknown.
The aim of this project is characterize the proteomic profile of MVs from selected Mtb lineages and
to unveil the role of MVs in mycobacterial adaptation to environmental factors.
Mtb cells were cultured with and without oxidative and nitrosative stress. The Mtb cell free culture
supernatant was used as a source to isolate enriched MVs by density gradient ultra-centrifugation.
The purified membrane vesicles were investigated by mass spectrometry. The MaxQuant and
Perseus softwares were used for peptide search, data analysis and quality control, respectively.
The functional categories of the proteins identified were grouped using the KEGG pathway
database.
The mass spectrometry proteomic data revealed that MVs contain enriched packaging of virulence
associated mycobacterial proteins. The majority of the MV-specific components detected were
secreted proteins which are reported to be major virulence factors (antigen-85 complexes, ESAT-6
and PE/PPE, etc). Based on the KEGG pathway functional analysis, the identified proteins were
found to be involved in two-component systems, ABC transporters, amino acid and fatty acid
metabolism, as well as catalytic and hydrolase activity and hypothetical proteins.
This study generated new knowledge on the proteomic profile of Mtb MVs. We have characterized
Mtb membrane vesicles, which are enriched with surface/secreted antigenic proteins and virulence
factors. Further in silico and in vitro/in vivo studies are required to investigate the adaptive and
clinical relevance of the MV proteins identified, including the newly discovered un-characterized
proteins.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
125
104 Mycobacterial Biology & Physiology
20/09/2016
The rv0805 gene in M. tuberculosis encodes a metallophosphoesterase with unique
structural features that determine its biological functions
P. Biswas1, N. Matange1, G. Larrouy-Maumus2, S. Visweswariah1
1
Department of Molecular Reproduction Development and Genetics, Indian Institute of Science,
Bangalore, India 2Department of Life Sciences, Imperial College, London, United Kingdom
Cyclic AMP-mediated signalling in mycobacteria plays an important role in the physiology of the
organism and survival of pathogenic mycobacteria within host macrophages. Rv0805 was initially
identified as a class III cNMP PDE, and orthologs are found only in slow growing mycobacteria.The
crystal structure of Rv0805 reveals that it has a β𝛂β𝛂β MPE core fold with Fe3+ and Mn2+ present at
its active site which are co-ordinated via aspartate, histidine and asparagine residues. These metal
ions contribute to dimerization of Rv0805. The C-terminal cap domain of Rv0805 modulates its
expression, and helps in its cell envelope localization. Interestingly, a strain of M. bovis BCG
deleted for the rv0805 gene shows a stark growth defect when glucose or propionate was provided
as the sole carbon source. The growth defect could be complemented by expression of the full
length protein but not following deletion of the C-terminal 40 residues.Acetate, succinate, glutamate
or citrate supplementation did not rescue the growth defect observed in glucose. In contrast,
metabolomic analysis indicated differences in aspartate and ⍺-ketoglutarate levels when the
bacteria were grown in propionate as the sole carbon source. This observation suggests a role for
Rv0805 in the biosynthesis of intermediates involved in cell wall generation in slow-growing
mycobacteria under conditions where glucose or propionate serve as the primary carbon source.
126
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
105 Mycobacterial Biology & Physiology
20/09/2016
Building genome-wide mutant resources in slow-growing mycobacteria by Cartesian
Pooling-Coordinate Sequencing (CP-CSeq)
K. Vandewalle3, K. Borgers3, N. Festjens3, E. Plets3, M. Vuylsteke1-3, Y. Saeys2, N. Callewaert3
1
Gnomixx 2IRC, Unit for Immunoregulation and Mucosal Immunology 3MBC, Unit for Medical
Biotechnology, VIB - Ghent University, Ghent, Belgium
Scientific mycobacterial research requires the availability of efficient molecular and genetic tools.
The genetic manipulation of slow-growing mycobacteria has long been problematic due to their slow
growth, the low efficiency of classical transformation methods and the low frequency of homologous
recombination. The introduction of conditionally replicating mycobacteriophages changed the field:
specialized transduction has proven to be useful for generating deletion mutants in most
mycobacteria. But the slow growth of mycobacteria remains a problem and applying these new
methods in a gene-by-gene approach is laborious and costly. However, transposon mutagenesis
makes it possible to make a wide variety of mutants in an isogenic mycobacterial background in a
quick and efficient manner.
Our lab developed a method to create characterized ordered transposon insertion mutant libraries,
namely Cartesian Pooling-Coordinate Sequencing or CP-CSeq, combining pooling steps of
transposon mutants along Cartesian coordinates with multiplex transposon sequencing of the
resulting pools. In this manner a database containing positional information about each individual
mutant present in a sequence-tagged mutant library can be obtained. We demonstrated this
approach in the Mycobacterium bovis BCG vaccine strain, providing the largest resource of mutants
in any strain of the M. tuberculosis complex. The ordered library contains 9.216 clones (96 x 96-well
plates) for which 77% of the mutants could be positioned in the library, targeting 64% of the nonessential genes for in vitro growth.
CP-CSeq has the potential to dramatically speed up hypothesis testing in Mycobacterium biology
and can be extended to any entity with difficult genetics and/or slow growth for which sequencetagged identification is possible.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
127
106 Mycobacterial Biology & Physiology
20/09/2016
Key experimental evidence of chromosomal DNA transfer among selected tuberculosiscausing mycobacteria
E. Boritsch7, V. Khanna4, A. Pawlik7, N. Honoré7, V.H. Navas5, L. Ma6, C. Bouchier6, T. Seemann2,
P. Supply3, T.P. Stinear1, R. Brosch7
1
Department of Microbiology and Immunology, University of Melbourne, Parkville 2Monash
University, Clayton, Victorian Bioinformatics Consortium, Victoria, Australia 3Center for Infection
and Immunity, Inserm U1019, CNRS UMR8204, Université de Lille, Institut Pasteur de Lille,
Lille 4Hub Bioinformatique et Biostatistique, C3BI, USR 3756 IP CNRS 5Lymphocyte Cell Biology
Unit 6PF1-Plate-Forme Génomique 7Unit for Integrated Mycobacterial Pathogenomics, Institut
Pasteur, Paris, France
Horizontal gene transfer (HGT) is a major driving force of bacterial diversification and evolution. For
tuberculosis-causing mycobacteria, the impact of HGT in the emergence and distribution of
dominant lineages remains a matter of debate. Here, by using fluorescence-assisted mating assays
and whole genome sequencing, we present unique experimental evidence of chromosomal DNA
transfer between tubercle bacilli of the early-branching Mycobacterium canettii clade. We found that
the obtained recombinants had received multiple donor-derived DNA fragments in the size range of
100 bp to 118 kbp, fragments large enough to contain whole operons. Even though the transfer
frequency between M. canettii strains was low and no transfer could be observed among classical
Mycobacterium tuberculosis complex (MTBC) strains, our study provides the proof of concept for
genetic exchange in tubercle bacilli. This outstanding, now experimentally validated phenomenon
presumably played a key role in the early evolution of the MTBC towards pathogenicity. Moreover,
our findings also provide important new information for the risk-evaluation of potential transfer of
drug resistance and fitness mutations among clinically-relevant mycobacterial strains.
128
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
107 Mycobacterial Biology & Physiology
20/09/2016
A cytoplasmic peptidoglycan amidase homologue controls mycobacterial cell wall
synthesis
C. Boutte2, C. Baer3, C. Sassetti3, T. Ioerger1, E. Rubin2
1
Texas A&M University, College Station 2Immunology and Infectious Disease, Rubin Lab, Harvard
TH Chan School of Public Health, Somerville 3UMass Worcester Medical School, Worcester,
United States
Regulation of cell wall assembly is essential for bacterial survival and contributes to pathogenesis
and antibiotic tolerance in Mycobacterium tuberculosis (Mtb). However, little is known about how the
cell wall is regulated in stress. We found that CwlM, a protein homologous to peptidoglycan
amidases, coordinates peptidoglycan synthesis with nutrient availability. Surprisingly, CwlM is
sequestered from peptidoglycan (PG) by localization in the cytoplasm, and its enzymatic function is
not essential. Rather, CwlM is phosphorylated and associates with MurA, the first enzyme in PG
precursor synthesis. Phosphorylated CwlM activates MurA ~30 fold. CwlM is dephosphorylated in
starvation, resulting in lower MurA activity, decreased cell wall metabolism, and increased tolerance
to multiple antibiotics. We describe a system that controls cell wall metabolism in response to
starvation, and show that this regulation contributes to antibiotic tolerance.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
129
108 Mycobacterial Biology & Physiology
20/09/2016
Differences in the expression profile of the phoP locus among laboratory Mycobacterium
tuberculosis strains in acidic stress response
N. Bresciani, D.M. Cirillo, P. Miotto
Emerging bacterial pathogens unit, Division of mmunology, Transplantation and Infectious
Diseases, San Raffaele Scientific Institute, Milan, Italy
The phoPR two-component system is involved in the regulation of relevant pathogenic features in
M. tuberculosis (MTB). Despite its relevant role, the environmental signals triggering the phoPR
machinery are still unclear. In addition, small regulatory RNAs (sRNAs), that regulate several key
bacterial processes, control stress response and virulence even if their role in tuberculosis (TB)
pathogenesis remains poorly understood.
The aim of this study is to characterize the expression profile of ncRv0757c, a recently identified
mycobacterial sRNA cis-encoded to the phoP gene, in two MTB laboratory strains (CDC1551 and
H37Rv) during acidic pH stress response in vitro.
We compared strains in early logarithmic growth phase grown at pH 7.0 with bacteria grown at pH
5.0. At selected time points (24 and 48 hours), we extracted the total RNA using the mirVana
miRNA isolation kit (Thermo Fisher Scientific) and checked the RNA purity on Agilent 2100
Bioanalyzer (Agilent Technologies). We assessed the expression profile of ncRv0757csRNA, phoP
and phoR genes by qRT-PCR using SYBR Green RT-PCR reagents kit (Life Technologies) and rrs
gene as endogenous control.
The sRNAncRv0757c was found to be significantly downregulated at 24 and 48 hours in CDC1551
at pH 5 compared to the reference strain at pH 7: its expression resulted 5-fold change lower than
H37Rv (p-value <0.001). At the same time, the phoP (p-value24H <0.05; p-value48H<0.01) and phoR
(p-value24H <0.01; p-value48H<0.001) genes are downregulated more than 5-fold at pH 5. The phoP
gene and ncRv0757c showed no response to acidic pH in H37Rv.
The phoP locus was modulated in response to acidic stress only in in the CDC1551 strain, but not in
H37Rv, thus suggesting that the response to acidic pH is more complex than thought and additional
factors might be involved. Despite the sRNA ncRv0757c could play a role in the regulation of the
phoP gene, as suggested by a similar expression profile, our results do not support its direct
involvement in the differential modulation of the phoP gene during acidic conditions observed
between CDC1551 and H37Rv strains.
130
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
109 Mycobacterial Biology & Physiology
20/09/2016
THPP target assignment reveals EchA6 as an essential fatty acid shuttle in mycobacteria
J. Cox3, K. Abrahams3, D. Barros2, K. Futterer3, L. Ballell2, G. Drewes1, G. Besra3
1
Cellzome - a GSK Company, Heidelberg, Germany 2GSK, Tres Cantos, Spain 3University of
Birmingham, Birmingham, United Kingdom
Phenotypic screens for bactericidal compounds against drug-resistant tuberculosis are beginning to
yield novel inhibitors.
However, reliable target identification remains challenging. Here, we show that
tetrahydropyrazo[1,5-a]pyrimidine-3-carboxamide (THPP) selectively pulls down EchA6 in a
stereospecific manner, instead of the previously assigned target M. tuberculosis MmpL3. While
homologous to mammalian enoyl-coenzyme A (CoA) hydratases, EchA6 is non-catalytic yet
essential and binds long-chain acyl-CoAs. THPP inhibitors compete with CoA-binding, suppress
mycolic acid synthesis, and are bactericidal in a mouse model of chronic tuberculosis infection. A
point mutation, W133A, abrogated THPP binding and increased both the in vitro minimum inhibitory
concentration and the in vivo effective dose 99 in mice.
Surprisingly, EchA6 interacts with selected enzymes of fatty acid synthase II (FAS-II) in bacterial
two-hybrid assays, suggesting essentiality may be linked to feeding long-chain fatty acids to FAS-II.
Finally, our data show that spontaneous resistance-conferring mutations can potentially obscure the
actual target or alternative targets of small molecule inhibitors.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
131
110 Mycobacterial Biology & Physiology
20/09/2016
Several Mycobacterial species are capable of forming spore-like state
J. Dahl
Department of Biology, University of Minnesota, Duluth, United States
Recent reports have argued both for and against the capability of mycobacteria to form heatresistant spores. The existence of a spore-like state would help explain mycobacteria persistence
and dormancy in infected hosts and in the environment. Our laboratory has recently identified
several mycobacterial species that can survive conditions exceeding classic pasteurization. Two of
these mycobacterial species remain viable even after 6 hours exposure to wet heat of 65oC.
Production of the heat-resistant state does not appear to be dependent upon production of
dipicolonic acid nor upon the action of the stringent response. This is in spite of these two features
being vital for gram-positive bacterial sporulation. Heat resistance of these two mycobacteria
correlates with an ability to survive in a desiccated state for upwards to 3 years. Collectively our
data shows that mycobacterial are capable of much more stress-resistant physiological states than
was previously realized.
132
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
111 Mycobacterial Biology & Physiology
20/09/2016
Mycobacterium tuberculosis cyclophilins show chaperone like activity in-vitro and in-vivo
S. Pandey1-3, D. Tripathi2, A. Sharma2, T.K. Chaudhary2, S.E. Hasnain2, N.Z. Ehtesham1
1
Inflammation Biology and Cell Signaling Laboratory, National Institute of Pathology,
Allahabad 2Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi 3School of
Life Sciences, University of Hyderabad, Hyderabad, India
Tuberculosis, named after its causative agent the tubercle bacillus or Mycobacterium tuberculosis is
one of the major causes of mortality and morbidity all over the world. Approximately 2 million people
die of this deadly disease every year.
PPIase peptidyl-prolyl cis-trans isomerases commonly known as cyclophilins are ubiquitously
expressed enzymes that (PPIase, EC 5.2.1.8) assist in protein folding by isomerization of Xaa-Pro
peptide bonds, which otherwise can act as folding bottleneck and will limit the rate of final folding
steps. Mycobacterium tuberculosis (M.tb) is known to possess two cyclophilins (Ppiases), PpiA
(Rv0009) and PpiB (Rv2582). This study describes chaperone-like activity of mycobacterial Ppiases
and their role in stress adaptation. We show that recombinant rPpiA and rPpiB can bind to nonnative proteins in vitro and can prevent their aggregation. Purified rPpiA and rPpiB exist in
oligomeric form as evident from gel filtration chromatography. E. coli cells overexpressing PpiA and
PpiB of M.tb could survive thermal stress as compared to plasmid vector control. HEK293T cells
transiently expressing M.tb PpiA and PpiB proteins show increased survival as compared to control
cells in response to oxidative stress and hypoxic conditions, thereby pointing to their likely role in
adaption under host generated oxidative stress and conditions of hypoxia. The chaperone-like
function of these M.tuberculosis cyclophilins may possibly function as a stress responder and
consequently contribute to virulence.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
133
112 Mycobacterial Biology & Physiology
20/09/2016
The role of glycolysis in the activation of human monocyte-derived dendritic cells following
exposure to bcg
K. Elayati, D. Triglia, S. O'leary, J. Keane, M. O'sullivan
Clinical Medicine, TCD, Dublin, Ireland
Background: Dendritic cells(DCs) play an essential role in both innate and adaptive immunity.
Recent advances have indicated a role for monocyte-derived DCs (moDCs) in the production of IL1α and IL-1β in a murine model of Mycobacterium tuberculosis (Mtb) infection. In addition, moDCs
are essential for transport of live mycobacteria from the lung to the draining lymph nodes to initiate
an adaptive immune response. A metabolicswitch from oxidative phosphorylation to glycolysis
occurs in DCs stimulated with TLR agonists and is required for activation. The aim of this study is to
investigate the role of glycolysis in human moDCs activated with mycobacteria.
Methods: Human monocyte-derived DCs were infected with different ratios of BCG-GFP (MOI 50 –
250) in the present and absence of glycolysis inhibitor 2-deoxyglucose (2DG) for 24 hr. Changes in
expression of maturation markers, chemokine receptors and cell viability were assessed by flow
cytomety. Statistical analysis was performed using standard statistical software (Prism
5.0).Significance level was set at p˂0.05.
Results: Infection of moDCs resulted in increased expression of the maturation marker CD86.There
was no change in the percentage of HLA-DR positive cells but surface expression was increased in
DCs infected at an MOI of 100 compared to uninfected cells.There was a decrease in surface
expression of CCR7(% and MFI) in cells infected at an MOI of 100 compared to uninfected but this
did not reach statistical significance.Additionally, there was a dose-dependent increase in cell death
of infected moDCs. Inhibition of glycolysis had no significant effect on expression of CD86, HLA-DR
or CCR7. However, there was a significant increase in the viability of moDCs infected at MOI 100
and 250 BCG-GFP following treatment with 2DG.Annexin V staining revealed that DCs did not
undergo apoptosis with or without 2DG treatment.
Conclusion: BCG-GFP infection of moDCsmay cause a metabolic switch towards glycolysis and
away from oxidative phosphorylation.However, aerobic glycolysis does not appear to be essential
for DC maturation following infection with BCG andappears to compromise cell viability.Future
experiments will investigate whether DCs overcome blocking of glycolysis by generating energy
through alternate methods.
134
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
113 Mycobacterial Biology & Physiology
20/09/2016
A new mechanism of programmed cell death in M. tuberculosis by a TA toxin hydrolyzing
intracellular NAD+
D. Freire3, C. Gutierrez1, A. Grabowska4, V. Pogenberg3, T. Schneider3, M. Cianci3, A. Sala2,
K. Nukdee2, P. Genevaux2, M. Wilmanns3, O. Neyrolles1, A. Parret3
1
Institute of Pharmacology and Structural Biology 2Laboratory of Microbiology and Molecular
Genetics, University of Toulouse, Toulouse, France 3European Molecular Biology Laboratory,
Hamburg, Germany 4Department of Pathogen Molecular Biology, London School for Hygiene and
Tropical Medicine, London, United Kingdom
Toxin-antitoxin (TA) modules are two-gene operons that are widely prevalent in bacterial genomes.
They are essential to various cellular processes, such as cell wall synthesis and gene regulation, as
well as protection against phages and programmed cell death. TA modules encode a toxic protein
which infers growth arrest of the producer cell by interfering with cellular replication and translation
processes in a similar way as antibiotics do. The antidote is provided in the form of an antitoxin
which very efficiently inhibits the toxic activity in normal growth conditions. In M. tuberculosis (Mtb),
TA systems are found in usually high numbers and are associated with pathogenesis and persistent
infections. The vast majority of TA systems in Mtb belong to class II, systems, where the antitoxin is
a labile protein which inhibits toxin activity through complex formation.
Here, we present the structure-function analysis of a new type of class II TA system from M.
tuberculosis H37Rv encoded by Rv1990c-Rv1989c. The crystal structure of the TA complex reveals
unexpected structural homology of the toxin Rv1989c to bacterial exotoxins with NAD+ hydrolase
activity. The antitoxin inactivates the toxin by occupying the active site through substrate mimicry.
This TA complex forms a compact heterododecameric complex in which the hypothetical DNAbinding domain of each antitoxin moiety is sterically blocked. We established NAD+-hydrolysis
activity in cellular extracts of M. smegmatis and E. coli upon induction of the toxin. Expression of
active site mutants in M. tuberculosis no longer conferred the killing phenotype. Our findings
suggest that this TA module represents a new subclass of type II TA systems where the cognate
toxin directly confers bactericidal activity by rapid depletion of cellular NAD+. As a result, constitutive
activation of Rv1989c by blocking interaction with Rv1990c would trigger bacterial suicide. These
findings open up new avenues for the development of highly effective antituberculosis drugs.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
135
114 Mycobacterial Biology & Physiology
20/09/2016
Mycobacterium tuberculosis Dormancy proteins: Key players in cell wall integrity, stress
tolerance and virulence
R. Garg3-4, D. Tripathi3, S. Kant2, V. Gupta3, A. Seth1, R. Bhatnagar3, N. Banerjee3
1
Department of Bio-Science and Biotechnology, Banasthali University, Banasthali,
Rajasthan 2International Centre for Genetic Engineering and Biotechnology 3BSL3 Laboratory,
School of Biotechnology, Jawaharlal Nehru University, Delhi 4Amity Institute of Biotechnology,
Amity University Gurgaon Haryana, New Delhi, India
Dormancy is a physiological phenomenon of Mycobacterium tuberculosis, which has still remained
an enigma for scientists across the globe. M. tuberculosis (Mtb) has two stages of disease
pathogenesis, the first being active stage involves replication of bacteria in the macrophages and
the second is the dormancy stage, which the present drug regimen fails to target. It has been seen
that a two component system DosRS is responsible for inducing 48 member dormancy regulon in
Mtb, using signals like hypoxia, NO and CO. Diversity of functions associated with dormancy
regulon genes makes it important to characterize them, so that new avenues in drug research can
be opened. In an attempt to discover role of various dormancy genes, we studied locus Rv0574c
which codes for polyglutamate synthase like protein, contributing to virulence of Mtb, by virtue of
synthesizing poly-α-L-glutamine layer in mycobacterial cell wall. Rv0574c gene showed highest
expression in late log phase with concomitant accumulation of poly-α-L-glutamine in the cell wall.
Rv0574c was activated under conditions prevalent in the tubercular granuloma e.g. hypoxia, nitric
oxide and CO2. For functional characterization, a deletion mutant of the gene was produced by
allelic exchange. The mutant produced lower amounts of poly-α-L-glutamine in the cell wall
compared to the wild type bacteria. Additionally, increased sensitivity of the mutant to antitubercular drugs, SDS, lysozyme and mechanical stress was accompanied with drastic reduction in
ability to form biofilm. Growth of the ΔRv0574c strain was normal under in vitro conditions but was
retarded in THP-1 macrophages and in the lungs and spleen of BALB/c mice. This was in
agreement with histopathology of the lungs showing slow growth and less severe pathology than
the wild type strain. Therefore, Rv0574c locus by virtue of modulating PLG content in the cell wall,
helps in maintaining cellular integrity under hostile host environment. The second gene which we
are studying is cation transporter protein F (ctpF), which belongs to the under explored P-type
ATPase family in Mycobacteria. ctpF gene expresses maximally in stationary phase and shows
upregulation in acidic conditions, NO stress, CO2 as well as in hypoxia. Creation of a deletion
mutant will help us further assess the contribution of this gene in virulence of Mtb. These two
proteins as well as other conserved but functionally uncharacterized dormancy proteins pose
themselves as attractive anti-tubercular drug targets.
136
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
115 Mycobacterial Biology & Physiology
20/09/2016
Discovery and characterization of a model mycobacterial sRNA involved in the iron-sparing
response
E. Gerrick, S. Fortune
IID, Fortune Lab, Harvard School of Public Health, Boston, United States
An emerging theme in bacteria is a reliance on small regulatory RNA molecules (sRNAs) for rapidly
modulating gene expression. sRNAs, which in most bacteria rely on the sRNA accessory factor
protein Hfq to bind to their target mRNAs, play a vital role in virtually all stages of growth. However,
despite the increasingly well-recognized importance of bacterial sRNAs, very little is known about
these regulators in the important human pathogen Mycobacterium tuberculosis (Mtb) or any of its
relatives. While a number of putative sRNAs have been identified in Mtb, not a single sRNA target
has been experimentally validated and mycobacteria lack every known sRNA accessory factor
protein. In order to remedy this, we have performed bacterial sRNA sequencing (bsRNA-Seq) on
Mtb grown under iron starvation conditions to identify potential iron-sparing sRNAs. We then
created a computational pipeline to identify novel sRNAs within bsRNA-Seq datasets and
discovered one novel sRNA that is highly abundant only during iron starvation. This sRNA, which
we have named MyhB, is highly conserved across all mycobacterial lineages, and we have
confirmed its role in the iron sparing response in M. smegmatis. We then used RNA-sequencing to
identify 14 genes as putative members of the MyhB regulon and confirmed regulation of one target,
bfrA, by RT-qPCR and using a luciferase reporter fusion. In addition, we have identified a 6nt
putative seed region of MyhB, which is predicted to bind to its mRNA targets. These results
represent the most in-depth characterization of a mycobacterial sRNA to date and provide valuable
insights into mycobacterial sRNA-mediated riboregulation.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
137
116 Mycobacterial Biology & Physiology
20/09/2016
The role of nitric oxide in the production of Rpf-dependent Mycobacteria
S. Glenn1, O. Turapov1, B. Kana3, P. Andrew1, V. Makarov2, G. Mukamolova1
1
Department of Infection, Immunity and Inflammation, University of Leicester, Leicester,
France 2Bach Institute of Biochemistry, Russian Academy of Sciences, Moscow, Russia 3DST/NRF
Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the
Witwatersrand, Johannesburg, South Africa
Tuberculosis (TB) is a major global health problem, causing 1.5 million deaths annually. One third of
the world’s population are thought to be latently infected with Mycobacterium tuberculosis (Mtb)
which has specialised mechanisms for long-term persistence in vivo and in vitro. Past studies have
shown that a high number of Mtb bacilli isolated from human TB sputum or from murine infected
tissues are only able to grow in the presence of resuscitation promoting factors (Rpf). Furthermore
Rpf-dependent Mtb are more tolerant to first line tuberculosis drugs rifampicin and isoniazid and
apparently enriched in sputum of treated tuberculosis patients, indicating the clinical importance of
these bacilli for TB management and treatment. The mechanisms behind the formation of these
Rpf-dependent bacilli are still poorly understood at both a host and pathogen level, and the lack of a
reliable model has made investigations into this problematic. We hypothesised that nitric oxide, an
important component of the host immune defences, produced in vivo might be responsible for the
generation of Rpf-dependent bacilli. Using several novel NO donors, we investigated the induction
of Rpf-dependency in mycobacteria in vitro. Treatment of mycobacteria with novel NO donor
resulted in rapid generation of Rpf-dependent bacilli, which maintained integrity of cellular
membrane and could be resuscitated by addition of Rpf-containing culture supernatant. By
application of DAF-FM staining we confirmed that the NO donors release a low level of nitric oxide,
which we hypothesise stimulates signaling pathways within the bacteria, resulting in Rpfdependency. Current investigation of transcriptomics and proteomics profiles of NO-treated Mtb will
aid establishment of molecular mechanisms underlying generation of RPF-dependent mycobacteria
in vivo.
138
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
117 Mycobacterial Biology & Physiology
20/09/2016
Evaluation of selective translation in Mycobacteria using reporter strains
A. Grabowska, N. Andreu, T. Cortes
Department of Pathogen Molecular Biology, London School of Hygiene & Tropical Medicine,
London, United Kingdom
Mycobacterium tuberculosis differs from other bacterial pathogens in expressing an unexpected
number of leaderless mRNA transcripts. Leaderless mRNAs lack a 5’ untranslated region (UTR)
that usually harbours the Shine-Dalgarno (SD) sequence, the dominant signal for canonical
translation initiation in bacteria. In Escherichia coli, only a few leaderless transcripts have been
described and they are translated with low efficiency by E. coli ribosomes, when the 70S monosome
directly binds to the ATG start codon. The scenario changes during stress conditions, where a
translational reprogramming takes place generating specialised ribosomes that selectively translate
leaderless transcripts. We have previously demonstrated that proteins with secondary adaptive
functions are generally leaderless in M. tuberculosis. Furthermore, in a starvation model of growth
arrest, we detected a significant increase in the overall ratio of leaderless to SD transcripts. Here,
we have engineered bioluminescent mycobacterial reporter strains representative of leaderless and
SD transcripts to study differences in their translational efficiencies. First, reporter constructs were
assessed in E. coli. As expected, we detected basal levels of luminescence for the leaderless
reporter in E. coli during exponential growth. In contrast, leaderless and SD transcripts were
translated with similar efficiencies in exponentially growing mycobacteria, suggesting the
mycobacterial ribosome differs from the E. coli model in being able to translate both types of
transcripts. Secondly, reporter strains were used to study changes in translation efficiencies during
different phases of growth. Whereas translation of leaderless and SD transcripts was maintained at
similar rates during exponential growth, upon entrance into growth arrest, translation of leaderless
transcripts was maintained whilst translation of SD transcripts significantly declined. Overall, our
results suggest that preferential translation of leaderless transcripts may have particular importance
in the physiology of non-replicating M. tuberculosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
139
118 Mycobacterial Biology & Physiology
20/09/2016
Mycobacterium tuberculosis H37Rv encoded ParE2 toxin induces persister cell formation
through SOS response
M. Gupta1-2, R. Sitaraman2, R. Bhatnagar1, N. Banerjee1
1
Molecular and Cell Biology Laboratory, Jawaharlal Nehru University 2Department of
Biotechnology, TERI University, New Delhi, India
Mycobacterium tuberculosis H37Rv (Mtb) escapes host-generated stresses and antibiotics by
entering a dormant, persistent state. Activation of toxin-antitoxin modules is one of the mechanisms
known to trigger such a non-replicative state. We have earlier investigated the parDE2 operon of
Mtb, which encoded the MParE2 toxin, an inhibitor of its own DNA gyrase. Here we scrutinize the
toxicity of MParE2 in RecA1 and RecA+ E. coli backgrounds to study the role of SOS response as a
survival strategy and examine different phenotypes generated due to MParE2 expression. Ectopic
expression of parE2 affected growth and viability of both the E. coli strains to different degrees.
Live-dead staining (SG-I/PI) revealed that in a short span of 4h, the toxin killed ~54% of the RecA1
strain compared to ~27% cells of the RecA+ cells. In both cases, the majority of the live cells
~99.99% were viable but non-culturable (VBNCs) and only 0.01% population were colony formers.
The results suggested that though more cells survived from the toxic effect in the RecA+ strain,
VBNC formation was not solely dependent on SOS regulation. MParE2 toxicity caused DNA
damage, triggering activation of SOS response, which led to inhibition of cell division and formation
of multi-nucleoid, long filamentous cells. A massive increase in sulA and tisB transcript levels, and
loss in membrane potential in the RecA+ cells and not in the RecA1 strain, all pointed to SOS
mediated regulation of these phenotypes. Severe morphological aberrations observed under the
electron-microscope in the parE2 expressing RecA1 E. coli strain were largely mitigated when the
toxin was expressed in the RecA+ cells. The latter cells were extensively filamentous with smooth
and intact membranes compared to the RecA1 cells, which were wrinkled and corrugated with
hyper-hydrated periplasmic space and large electron-lucent ‘vacuoles’, reiterating the SOSmediated rescue of MParE2-toxicated E. coli cells. As the effect of toxin waned with time, the cells
resumed division and recovered in colony forming ability. The MParE2 expressing RecA+ cells
produced significantly higher number of persisters than the RecA1 strain, upon exposure to different
antibiotics. The MParE2 triggered VBNC phenotype and persistence are hallmarks of dormancy,
potentially relevant in Tuberculosis.
140
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
119 Mycobacterial Biology & Physiology
20/09/2016
Regulatory circuits in the mycobacterial Pup-proteasome system
Y. Elharar, M. Korman, S. Schlussel, E. Gur
Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
Pup is a bacterial ubiquitin analog that is conjugated to lysine side chains of protein targets, thereby
tagging them for proteasome-mediated degradation. The Pup-proteasome system (PPS) was
discovered in Mycobacterium tuberculosis, where it was shown to be essential for both long term
persistence under starvation conditions, and for resistance to reactive nitrogen intermediates.
Indeed, PPS-defective M. tuberculosis mutants present attenuated virulence as compared to the
wild type strain. Being an intracellular proteolytic system, the PPS is tightly regulated to prevent
excessive protein degradation and unnecessary destruction of proteins. However, the mechanisms
that control PPS function and are likely to be targets for the development of anti-TB medicine, are
currently poorly characterize. In the PPS, the single Pup-ligase, PafA, shows exceptionally broad
specificity in tagging hundreds of protein targets for degradation. This promiscuity of PafA target
selection is well suited to the role of the PPS as an amino acid recycling factory under starvation
conditions. However, such PafA promiscuity also poses a threat to proper cell function, as
irreversible damage would be caused by excessive non-specific protein degradation without strict
control of the PPS. Our studies indicate that the PPS is, indeed, highly regulated, such that at the
maximum, protein pupylation only affects about 2% of the cell’s protein mass. While our previous
findings indicated that the PPS is auto-regulated by the tagging and degradation of its own
components, the mechanisms that control the expression of PPS genes remained unknown.
Furthermore, how these levels are coordinated at the system level remains poorly understood. We
now report that the level of PafA transcription is negatively regulated by PafB and PafC, two PPS
components encoded by the paf operon whose functions were unknown until now. Translational
control allows high PafA expression under starvation conditions, yet maintains low levels of this
enzyme when nutrients are in excess. We also find that levels of pupylated proteins in the cell are
limited by the concentration of Pup. Finally, our current understanding of the regulatory circuits that
control the PPS allow us to now to simulate the levels of PPS components and their effects on the
level of pupylated proteins, with the results of such simulation being consistent with experimental
observations made in both exponentially growing and starving cells.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
141
120 Mycobacterial Biology & Physiology
20/09/2016
Single-cell mycobacteria analysis reveals variability in the division time inherited
J.E. Hernandez, N. Ma, S. Hingley-Wilson, J. Mcfadden
University of Surrey, Guildford, Surrey, United Kingdom
Introduction: Variable outcomes observed in tuberculosis treatment could be attributed to
mycobacteria variable growth and division patterns that create deterministic population diversity at a
very high frequency [1]. Advances in microfluidics technology have allowed to study cell at singlecell level, and persistence phenomenon of bacterial populations has been analysed [2].
Tuberculosis treatment was unsuccessful in 14% of cases for 2013 [3] that suggest that new drugs
are necessaries to achieve the infection control. Studies of population heterogeneity can contribute
to find new therapeutic targets
Aim: To examine growth and division in mycobacteria using microfluidics and time-lapse
microscopy, and to track these cells to analyse division time
Material and Method: We studied Mycobacterium smegmatis mc2 155 at single cell level by the use
of microfluidics technology CellASIC ONIX Microfluidic Platform with B04A Microfluidic Bacteria
Plate (Millipore Corporation). Media flow was controlled via the CellASIC system at a flow rate of
~10 µl/hr. Imaging was performed under a Nikon confocal microscope (Nikon A1M on Eclipse Ti-E)
equipped with an environmental chamber, motorized stage and perfect focus system.
Automated multi area imaging was carried out using a 40X air objective lens (Nikon Apo λ) with a
numerical aperture of 0.95 Time-lapse videos of growing cells were recorded. Nikon Elements
software was used to obtain crops from the videos, and cells were manually tracked until 5st
generation with. Division time was calculated, and Pearson correlation between mother-daughters
and sisters-sisters division times were established
Results: Time-lapse video of M. smegmatis growth were obtained. After three independent
experiments we tracked an average 1000 cells each time. Correlation coefficient for mother and
daughters division time was around -0.2 which suggest non-correlation between them. Interestingly,
a positive correlation, coefficient correlation around 0.5 was observed between sisters.
Conclusion: Inheritance of division on time in M. smegmatis suggested that a factor inherited at
birth determines the division time and that this factor is rearranged during each generation.
Additional studies to identify factor(s) responsible of that behaviour are required to design new
strategies that reduce the variability in the mycobacteria population
References
1. Bree B. Aldridge, M.F.-S., Danielle Heller,Vijay Ambravaneswaran, and M.T.a.S.M.F. Daniel
Irimia, Asymmetry and Aging of Mycobacterial Cells Lead to Variable Growth and Antibiotic
Susceptibility. Science. 335(100): p. 4.
2. Balaban N. Q., J Merrin, J., Chait R., Kowalik L. and Leibler S., Bacterial Persistence as a
Phenotypic Switch. Science, 2004. 305: p. 4.
3. WHO, Global Tuberculosis report. 2014
142
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
121 Mycobacterial Biology & Physiology
20/09/2016
The ParA-Wag31 interaction provides the link between chromosome segregation and other
cell cycle processes in Mycobacteria
M. Pióro1, T. Małecki2, K. Ginda3, D. Jakimowicz2-1
1
Institute of Immunology and Experimental Therapy 2Faculty of Biotechnology, University of
Wroclaw, Wroclaw, Poland 3University of Oxford, Oxford, United Kingdom
Bacterial chromosome segregation depends on the ParA and ParB proteins. ParB binds DNA and
forms large nucleoprotein complexes which organise newly replicated chromosomes. The ParB
complexes are positioned in the specific locations of the cell due to interactions with dynamic
ATPase, ParA.
It was suggested that ParA is essential in M. tuberculosis (1).We have shown that elimination of
ParA inhibits growth of Mycobacterium smegmatis and disturbs chromosome segregation (2). The
dynamic structures of ParA segregate ParB complexes soon after the initiation of the chromosome
replication (3). Moreover, we have shown that mycobacterial ParA interacts with polar growth
determinant Wag31, the homologue of DivIVA protein (2). This interaction is unique to Mycobacteria
and taking into account indispensability of both proteins in M. tuberculosis it provides a potential
drug target.
Here, we further explored interaction between ParA and Wag31 using M. smegmatis as model
species. To determine the biological significance of the interaction, we have identified the mutant
ParA protein that does not interact with Wag31. We have constructed the M. smegmatis strain
expressing mutated parA and analysed its growth in different conditions. We have also analysed the
localisation of mutated ParA and its function in chromosome segregation.
Our studies indicate that interaction between ParA and Wag31 provides the link between the
chromosome segregation and the other processes of cell cycle. We suggest this interaction allows
the coordination of the cell cycle and may be crucial during persistence in M. tuberculosis.
1.
Sassetti CM, Boyd DH, Rubin EJ (2003) Genes required for mycobacterial growth defined by
high density mutagenesis. Molecular microbiology 48(1):77–84.
2.
Ginda K, et al. (2013) ParA of Mycobacterium smegmatis co-ordinates chromosome
segregation with the cell cycle and interacts with the polar growth determinant DivIVA.
Molecular microbiology 87(5):998–1012.
3.
Trojanowski D, et al. (2015) Choreography of the Mycobacterium Replication Machinery
during the Cell Cycle. 6(1):1–12.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
143
122 Mycobacterial Biology & Physiology
20/09/2016
Biosynthesis of mycobacterial lipoarabinomannan: disruption of aftB results in complete
loss of terminal β(1→2) arabinofuranosyl residues
M. Jankute3, L. Alderwick3, S. Noack2, N. Veerapen3, J. Nigou1, G. Besra3
1
Institute of Pharmacology and Structural Biology, The French National Centre for Scientific
Research, Toulouse, France 2Institute for Bio- and Geosciences: Biotechnology, Research Centre
Jülich, Jülich, Germany 3Institute of Microbiology and Infection, University of Birmingham,
Birmingham, United Kingdom
Mycobacterium tuberculosis, the etiological agent of tuberculosis, remains a highly successful
bacterial pathogen. Its persistence is associated with the thick, carbohydrate and lipid rich cell wall
with distinct lipoglycans that is highly impermeable to hydrophilic drugs. This highly complex and
unique structure is crucial for the growth, viability and virulence of M. tuberculosis, thus representing
an attractive target for vaccine and drug development. It contains a large macromolecular structure
known as the mycolyl–arabinogalactan– peptidoglycan complex (mAGP), as well as phosphatidylmyo-inositol derived glycolipids with potent immunomodulatory activity, notably lipomannan and
lipoarabinomannan (LAM). AG and LAM (lipo)polysaccharides share similar arabinan domains with
a well-defined structure, however, the biosynthesis of both remains somewhat incomplete.
Mycobacterial glycosyltransferases with dual functionalities have been previously identified. Here,
we have investigated the potential role of known arabinosyltransferase AftB in the biosynthesis of
LAM. Deletion of aftB in Mycobacterium smegmatis could only be achieved in the presence of a
rescue plasmid carrying a functional copy of a gene, strongly suggesting that aftB is essential. Lipid
analysis of the conditional mutant strain demonstrated an increase in cell wall associated lipids and
a significant decrease in cell-wall bound mycolic acid methyl esters. A monoclonal antibody
generated against arabinan motif of LAM no longer recognized the LAM structure extracted from the
conditional mutant strain. Subsequent structural characterization confirmed a complete lack of
β(1→2) arabinofuranosyl residues. Furthermore, we demonstrated that truncated LAM display
proinflammatory activity, which is due to its ability to activate Toll-like receptor 2. Altogether, our
results indicate that Ms-AftB is an essential mycobacterial ArafT that carries dual functionality and is
responsible for the synthesis of the arabinan domain of both LAM and AG.
144
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
123 Mycobacterial Biology & Physiology
20/09/2016
Mycolic acid processing in Mycobacterium tuberculosis
A. Javid, A. Bhatt, R. Kalscheuer
University of Birmingham, Birmingham, United Kingdom
Mycobacterium tuberculosis the causative agent of tuberculosis (TB) infects upto 9 million people
per year resulting in approximately 1.5 million deaths due to the disease. With the emergence of
multi drug resistant (MDR) and extremely drug resistant (XDR) strains arises the need for novel
targets for anti-TB therapy. Mycolic acids are essential components of the unique, lipid rich cell wall
of M. tuberculosis. However, enzymes involved in the biosynthesis of mycolic acids remain under
exploited as drug targets despite one of the early and hallmark anti-TB drug isoniazid which inhibits
mycolate biosynthesis. Previous studies from our laboratory identified mycolate processing
enzymes and transporters. Using gene knockdowns we have now extended these studies to slow
growing mycobacteria like Mycobacterium tuberculosis. Furthermore using BLAST-P alignments
and predictions of a 3D structure we identified unique domains in the mycolate processing
enzymes, and present functional studies on the same.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
145
124 Mycobacterial Biology & Physiology
20/09/2016
Mycobacterial HspX is both necessary and sufficient for the unipolar sorting of misfolded
protein aggregates
J.H. Zhu, Y.W. Zhang, X.H. Zheng, Y. Wu, B. Javid
Tsinghua University School of Medicine, Beijing, China
Protein misfolding is an inevitable consequence of protein synthesis and therefore all cells have
evolved mechanisms with which to deal with misfolded proteins, which have a tendency to form
aggregates known as inclusion bodies (IBs) in bacteria. In mycobacteria, in common with other
model organisms such as Escherichia coli, inclusion bodies tend to form at the poles of the cells.
Recent work has highlighted the importance of the heat shock proteins/ chaperones DnaK and ClpB
in dealing with misfolded and irreversibly oxidized proteins respectively at the poles of
mycobacteria, but how inclusion bodies get to the poles in mycobacterial cells has, until now, not
been known. We decided to investigate the role of the small heat shock protein, HspX in
mycobacterial inclusion body formation. HspX is significantly upregulated in tuberculous
granulomata and under conditions of hypoxia, and is an immunodominant antigen, but its precise
role in mycobacterial physiology is not known. Here, with the use of novel inclusion body fluorescent
protein reporters, we show that the formation of large inclusion body aggregates and their sorting to
predominantly to a single pole of the mycobacterial cell are both critically dependent on HspX.
HspX also localizes at the poles and IBs tend to form at the pole with greater HspX expression. In
an hspX null mutant of Mycobacterium smegmatis, small aggregates still form, but no longer
coalesce into inclusion bodies and are found throughout the cell. Importantly, the siting of IBs is
dependent on HspX: when HspX is relocalised to the septum, IBs now form, but are septally
located, verifying the causal role of HspX in IB localization. Our work identifies the cellular function
of the important mycobacterial antigen HspX and determines the mechanism for unipolar IB sorting
– one of the many mechanisms that mycobacteria employ for the generation of phenotypic diversity.
146
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
125 Mycobacterial Biology & Physiology
20/09/2016
Epigenetic control of Tuberculosis persistence: The role of the different sigma factors
L. Kaminski3, A. Benard1, N.R. Thomson1, G. Dougan2, W.R. Jacobs3
1
Microbial Pathogenesis 2Pathogen Genetics, The Wellcome Trust Sanger Institute, Hinxton,
Cambridge, United Kingdom 3Microbiology and Immunology, Albert Einstein College of Medicine,
Bronx, New York, United States
Mycobacterium tuberculosis (Mtb) infects 30% of the human population with the typical outcome of
causing a latent infection. The persistence phenotype in Mtb is a phenomenon of cells where a
fraction is resistant to a killing assault as a consequence of a transient gene expression or protein
modifications. This phenomenon of persisters is being unmasked at the presence of bactericidal
drugs or host adaptive immunity and it is an important impediment to tuberculosis control. While
several genes were previously proposed as contributing to the emergence of persistence, the
complete regulatory network is still unknown and it is clearly not attributed to one 'persister gene'.
To identify the specific genetic network causing the emergence of persistent cells we made a null
deletion mutant library of the twelve non-essential sigma factor transcription initiation factors in Mtb
and tested the effect of bactericidal drugs using these mutants as a screen for persister cells.
We have identified one of these sigma factor mutants to be sterilized in the presence of the first line
drug Isoniazid (INH). The RNA sequencing analysis showed differential expression of several
additional genes involved in transcription regulation and some genes in the oxidative regulation.
Other ongoing efforts are aimed to confirm the sterilization of this mutant in vivo in the presence of
the first line INH as a monotherapy.
Screening of the transcription factor mutants yields a future way in understanding their role in vivo in
the face of other drugs and host immunity.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
147
126 Mycobacterial Biology & Physiology
20/09/2016
Proteolytic control of the mycobacterial cell cycle
J. Kester2, O. Kandror1, T. Akopian1, A. Goldberg1, S. Fortune2
1
Cell Biology, Harvard Medical School 2Immunology & Infectious Diseases, Harvard University,
Boston, United States
Tuberculosis (TB) is the primary infectious killer worldwide, in part owing to increased resistance of
Mycobacterium tuberculosis—the etiologic agent of TB—to the current suite of antibiotics.
Bactericidal drugs typically target growth and division events, yet regulators of the mycobacterial cell
cycle remain undiscovered. In this work, we seek to identify key players in mycobacterial cell cycle
control, potentially leading to new antibacterial targets.
Mycobacteria share a unique pattern of growth and division with the model bacterium Caulobacter
crescentus. Caulobacter initiates its cell cycle through the highly controlled proteolysis of key cell
cycle regulators using the housekeeping protease complex, ClpXP. Therefore, we postulated that a
ClpX-dependent mechanism might underlie cell cycle control in mycobacteria as well.
To test this hypothesis, we depleted the essential AAA+ ATPase ClpX in the nonpathogenic model
Mycobacterium smegmatis (Msm) and used dynamic live cell imaging to assay for growth and
division effects. Loss of ClpX results in cell cycle arrest, supporting a role for ClpX and one or more
of its substrates in cell cycle regulation. To identify the set of ClpX substrates, we used a small
molecule inhibitor in whole cell lysates to trap potential substrates within the barrel of ClpX
hexamers. We used tandem MS/MS, biochemical examination, and molecular techniques to
validate cell cycle factors as substrates of ClpX.
In addition to providing a framework for understanding the unique essentiality for ClpX in
mycobacteria, this study also suggests potential cell cycle targets for new antibiotics. Additionally,
this work provides a methodology for biochemical identification of high-confidence substrates of
essential ATPases/chaperones.
JCK supported by NIH F31 AI120616-01 and NSF GRFP DGE-1144152
148
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
127 Mycobacterial Biology & Physiology
20/09/2016
The role of glycosyltransferase Rv1459c in mycobacteria
M. Šarkan2, M. Gilleron1, J. Nigou1, K. Mikušová2, M. Jackson3, J. Korduláková2
1
Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, Toulouse,
France 2Department of Biochemistry, Faculty of Natural Sciences, Comenius University in
Bratislava, Bratislava, Slovakia 3Department of Microbiology, Mycobacteria Research Laboratories,
Immunology, and Pathology, Colorado State University, Fort Collins, United States
The cell envelope of mycobacteria is a complex structure, which is important not only for the
integrity of mycobacterial cell, but also for pathogenicity and virulence of mycobacteria. It is
composed of plasma membrane, mycolyl-arabinogalactan-peptidoglycan core intercalated by
variety of lipids and glycolipids and polysaccharide rich capsule. Despite intense research
investigating the machinery of its biogenesis during last decades, several steps in the individual
pathways still need to be clarified. Much knowledge, especially regarding the processes essential
for mycobacteria, arose from the study of its family member corynebacteria. However, functions of
some homologous mycobacterial enzymes studied in corynebacteria are still uncertain.
The corynebacterial protein MptB was characterized as alpha(1→6) mannosyltransferase involved
in the initiation of core mannan synthesis of corynebacterial phosphatidylinositol-based lipomannan
and of multi-mannosylated lipopolysaccharide based on glucosyluronic acid-diacylglycerol, which
was not detected in mycobacteria (1). While its homologues Rv1459c from M. tuberculosis and
MSMEG_3120 from M. smegmatis demonstrated similar activity in vitro, deletion of msmeg_3120
gene in M. smegmatis did not affect the production of mycobacterial lipopolysaccharides
lipomannan and lipoarabinomannan (1).
To address the function of mycobacterial proteins we generated conditional knock-down mutant of
msmeg_3120 gene using TET-PIP OFF system (2) and examined the cell envelope composition of
this strain grown in different conditions.
1. Mishra, A. K., et al., (2008), Mol. Microbiol., 68, 1595-1613
2. Kolly, G. S., et al., (2014), Mol. Microbiol., 92, 194-211
Acknowledgements: This work was supported by the Slovak Research and Development Agency
under the contract No. APVV-15-0515 and NIH-NIAID grant AI064798.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
149
128 Mycobacterial Biology & Physiology
20/09/2016
Mycofactocin and cellular metabolism in Mycobacteria
G. Krishnamoorthy3, P. Kaiser3, R. Hurwitz2, H.J. Mollenkopf1, S.H. Kaufmann3
1
Core Facility Microarray/Genomics 2Core Facility Protein Purification 3Immunology, Max Planck
Institute for Infection Biology, Berlin, Germany
“Mycofactocin” (Rv0691A, Rv0692) is a short peptide clustered together with a radical S-adenosyl
methionine enzyme (Rv0693) and a lactate dehydrogenase (Rv0694) in Mycobacterium
tuberculosis (Mtb). This uncharacterized gene cluster is found in many actinobacteria and it is
predicted that mycofactocin requires post-translational modification by Rv0693 for its functioning
that is still unknown. In this study, we found that the transcript levels of mycofactocin components
(Rv0692, Rv0693, Rv0694) increased up to 30 folds under restricted oxygen condition, although
these genes were not essential for survival under hypoxic/anoxic conditions. Intriguingly, both under
normoxic and hypoxic conditions, deletion of Rv0693 prevented Mtb growth in the presence of
fermentable carbon source, whereas deletions of Rv0692 and Rv0694 accelerated the growth rate
under similar conditions. Furthermore, targeted metabolic measurements revealed that at least
under low oxygen growth conditions, ∆Rv0692 and ∆Rv0694 produced increased abundance of
lactate, while ∆Rv0693 exhibited increased extracellular pyruvate. These findings imply that
mycofactocin could partner with Rv0694 which mediates the interconversion of pyruvate or lactate
and thus regulates glycolytic flux. In addition, the growth of mycofactocin mutants in cholesterol was
poor with concomitant decrease in intracellular ATP levels and increased ratio of NADH/NAD+. We
found that the observed cholesterol growth defect in the mycofactocin mutants was due to added
ethanol, a solvent that is used to solubilize cholesterol. Transcriptome analysis revealed the
presence of significant induction (>3 folds) of genes associated with oxidative stress as well as
those regulated by DosS/T/R in ∆Rv0693 treated with ethanol, suggesting their inability to resist
redox imbalance. Taken together, these results demonstrated that mycofactocin can act as an
alternative electron carrier to possibly enable the function/s of dehydrogenases and/or associated
redox enzymes, and could potentially contribute to Mtb pathogenesis and persistence in human
host.
150
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
129 Mycobacterial Biology & Physiology
20/09/2016
A new class of anti-tuberculosis drug : a Griselimycin targeting DnaN
P. Laboudie2, E. Fontaine2, S. Klieber3, I. Blanc2, C. Lair2, X. Boulenc2, C. Cantalloube3,
M. Magnon3, S. Sans2, M. Doubovesky2, M. Geslin2, L. Collière1, A. Bauer1, G. Zech1,
S. Lagrange2,
E. Bacqué2, L. Fraisse2, A. Kling5, R. Müller5, A. Upton6
1
LGCR, Chilly-Mazarin, Frankfurt 2TSU ID, Marcy L'etoile 3DSAR, Montpellier, Vitry Sur Seine,
France 4LGCR, Sanofi R&D, Frankfurt 5Helmholtz Institute for Pharmaceutical Research Saarland,
Saarbrücken, Germany 6TB Alliance, New York, United States
Novel anti TB (tuberculosis) agents with mechanisms of action distinct from current TB drugs are
urgently needed due to the increasing prevalence of Drug Resistant (DR) TB.
As part of our TB drug discovery strategy, we revisited forgotten antibiotics with high anti-TB
potential and re-discovered griselimycin (GM). GM is a natural cyclic depsipeptide, identified in the
1960’s, structurally unrelated to any known TB drug and active against sensitive and DRMycobacterium tuberculosis. We developed a total synthesis to access to GM analogues and to be
able to prosecute a lead optimization program. Following an optimization program, we discovered
an outstanding TB compound, Cyclohexylgriselimycin (CGM) which was selected for in depth
profiling. This compound displayed excellent anti-TB properties comprising in-vivo activity alone and
in combinations.
We discovered that resistance to griselimycins, occurring at very low frequency, was associated
with amplification of a chromosomal segment containing DnaN. Our results demonstrated that
griselimycins had high translational potential for tuberculosis treatment, validated DnaN as an
antimicrobial target.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
151
130 Mycobacterial Biology & Physiology
20/09/2016
Anti-tuberculosis in vitro and in vivo activity of a new Sequanamycin-like Macrolide
C. Lair2, Y. Benedetti3, I. Blanc2, X. Boulenc1, C. Briot1, L. Fraisse2, S. Lagrange2, M. Magnon1,
E.L. Nuermberger4, S. Sans2, S. Silve2, A. Upton5, T. Kaneko5, T.J. Yang5, J. Zhang3
1
Disposition Safety and Animal Res, Chilly, Montpellier 2TSU ID, Marcy L'etoile 3LGCR/ Chemistry,
Sanofi R&D, Vitry, France 4Johns Hopkins University, Baltimore 5TB Alliance, New York, United
States
Medicinal chemistry optimization of Sequanamycin A, a 14-membered ring macrolide related to
Erythromycin, has lead us to a new class of macrolides with potential for the treatment of
Tuberculosis (TB). Here, we present the in vitro and in vivo activity profile of SEQ-9, an optimized
Sequanamycin that has demonstrated a promising contribution to anti-TB drug combinations in the
infected TB mouse models.
SEQ-9 is active on the replicative and non-replicative Mycobacterium tuberculosis (M.tb)
phenotypes. It is also potent against M.tb within mouse macrophages. Its even activities across
different M.tb lineages and strains mono-resistant to anti-TB drugs are in line with a new mode of
action in the TB area.
Like classical macrolides, SEQ-9 targets the bacterial ribosome thereby inhibiting protein synthesis
but unlike macrolides, it displays a different mechanism of resistance. In the case of classical
macrolides, the mechanism of resistance is well known and is linked to the methylation of the
ribosome following expression of the Erm37 gene. Contrary to Clarithromycin and Erythromycin, the
over expression of this gene in M.bovis or M.tb strains, doesn’t confer resistance to SEQ
compounds. On the other hand, mutants resistant to SEQ display an unprecedented deletion in
rRNA 23S in M.bovis BCG Pasteur strain.
Mouse PK parameters for SEQ-9 have been shown to be suitable for once-daily oral dosing. After
single administration of 300 mg/kg, Cmax of 3.1 µg/mL, t1/2 of 5.6 hours and AUC of 21 µg.h/mL
were obtained along with a lung/plasma ratio around 14 fold and oral bioavailability at 23%.
The minimum effective dose (MED) was reached at 300 mg/kg in the acute TB mouse model. In the
chronic TB model, SEQ-9 displayed a dose-dependent cidal efficacy from 75 mg/kg onward with a
1.7 log CFU reduction at 300mg/kg. In combination studies, SEQ-9 increased the bactericidal
activity of Pyrazinamide, Linezolid, Pa824 and Bedaquiline.
152
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
131 Mycobacterial Biology & Physiology
20/09/2016
Bioenergetic rewiring: turning the respiratory flexibility of Mycobacterium tuberculosis
against itself
D. Lamprecht1, P. Finin1, M. Rahman1, B. Cumming1, S. Russell1, S. Jonnala3, J. Adamson1,
A. Steyn1-4-5
1
KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), Durban, South
Africa 2University of Michigan Medical School, Ann Arbor 3NIAID, Tuberculosis Research Section,
NIH, Bethesda 4Centers for AIDS Research and Free Radical Biology 5Department of Microbiology,
University of Alabama at Birmingham, Birmingham, United States
The Mycobacterium tuberculosis (Mtb) electron transport chain (ETC) has received significant
attention as drug target, however its vulnerability may be affected by its flexibility in response to
chemical disruption. To determine the effect of the ETC inhibitors bedaquiline and Q203 on the Mtb
ETC and the value of the ETC as a drug target, we measured Mtb’s respiration upon inhibitor
addition using existing eukaryotic extracellular flux technology. We found that Mtb’s ETC rapidly
reroutes election flux around the sites of inhibition by these inhibitors and increases its total
respiration to maintain ATP levels, showing to greater metabolic flexibility than has previously been
appreciated.
Our data shows that, unlike in eukaryotes, back pressure does not substantially impede ETC activity
and allows for the increase respiration in the presence of these ETC inhibitors. The lack of back
pressure suggests that an energy spilling pathway modulates PMF in Mtb to prevent excessive
accumulation, which would ultimately stop ETC flux. We propose a model whereby both BDQ and
Q203 increase total ETC flux and respiration due to loss of feedback inhibition of carbon catabolism
and the TCA cycle by ATP. Rather than suggesting that the ETC is an undesirable drug target,
however, our results have demonstrated a manner to turn this plasticity against Mtb. In combination
targeting of the Mtb ETC by using respiratory inhibitors to rewire energy metabolism, we potentiate
CFZ’s production of bactericidal reactive oxygen species (ROS) which lead to enhanced killing in
vitro. The efficacy of ETC targeting drug combinations in vitro was validated in a macrophage
infection model, which showed no toxicity towards host cell lines.
Overall, our findings revealed novel insights into the Mtb ETC as a drug target that can be
effectively exploited for new therapeutic intervention strategies. We discovered an example of
combination-targeting that denies Mtb of the ATP generation benefits of the ETC, while also
exploiting Mtb’s metabolic flexibility to potentiate ETC-mediated ROS toxicity leading to enhanced
killing both in vitro and in a macrophage infection model.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
153
132 Mycobacterial Biology & Physiology
20/09/2016
Sodium chloride alters mycobacterial physiology and antibiotics tolerance
G. Larrouy-Maumus2, L. Marino1, A. Madduri4, T. Ragan3, D. Hunt3, L. Bassano2, M. Gutierrez3,
B. Moody4, F. Pavan1, L. De Carvalho3
1
School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil 2Life
Sciences, Imperial College London 3The Francis Crick Institute, Mill Hill Laboratory, London, United
Kingdom 4Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital and
Harvard Medical School, Boston, United States
Deciphering the mechanisms and host clues utilized by the bacterium to adapt and resist to
pharmacological sterilization is essential for improved development of novel therapeutics. Among
the factors that affect Mtb within host, sodium chloride appeared to be one of the key molecules
identified as a novel environmental cue. Hypotonic cultures of Mycobacterium tuberculosis are
widely used in basic research and to determine the effects of antibiotics. However, natural infection
and persistence within intra- and extracellular host compartments involve exposure to varying salt
concentrations. Using kinetic, organism-wide multi-omic measurements we show that exposure of
M. tuberculosis to high salinity markedly restricts growth and rapidly generates an adaptive
response, known as halotolerance. Our results demonstrate a broad remodelling of metabolism and
the cell envelope that occurs in two steps: a fast metabolic response independent of a late response
involving gene regulation and lipid remodelling. Together, these remodelling responses render M.
tuberculosis less sensitive to saline stress and, importantly, also to several clinically used
antibiotics. These findings broadly define the dynamic process of halotolerance in M. tuberculosis,
providing a new perception of envelope remodelling and drug action, as well as improved strategies
for antimicrobial sensitivity testing.
154
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
133 Mycobacterial Biology & Physiology
20/09/2016
Elucidating a new mechanism of drug activation
E. Meiler1, D. Blanco-Ruano1, K. Abrahams2, L. Guijarro-Lopez1, J. Rullas-Trincado1, A. MendozaLosana1, G. Besra2, C. Alemparte1, E. Perez-Herran1
1
Diseases of the Developing World, TB DPU, GlaxoSmithKline, Tres Cantos, Madrid,
Spain 2Institute of Microbiology and Infection, School of Biosciences, University of Birmingham,
Birmingham, United Kingdom
Drug-resistant tuberculosis (TB) is a major concern from a global health perspective. Thus new
treatments are needed to combat multidrug and extensively drug resistant TB strains that mainly
affect patients in developing countries with limited treatment success. The current treatment against
TB consists of combinations of drugs and can take longer than two years for drug-resistant strains.
Over the last couple of years, a big effort in phenotypic screening was carried out to identify hits
against Mycobacterium tuberculosis (Mtb), the causative bacterium of TB. One series of compounds
was discovered to efficiently kill Mtb in such a screen and its biological and pharmacological
properties were examined. In spite of the promising in vitro potency and favorable pharmacological
properties, one disadvantage of this series appeared to be a relatively high frequency of resistance.
Its mechanism of action studies established that it is a prodrug and needs to be activated by a nonessential bacterial enzyme AmiB2. Since the enzyme is not essential, it can be mutated in Mtb
causing a rather high level of resistance. This poster describes the profile of the hit and our targetidentification effort.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
155
134 Mycobacterial Biology & Physiology
20/09/2016
Disruption of carbonic anhydrase increases the intracellular virulence of Mycobacterium
tuberculosis and alters glycolysis
T. Mendum, R. Balhana, J. Mcfadden
University of Surrey, Guildford, United Kingdom
Carbon dioxide fulfils a number of universally important metabolic and physiological roles in many
bacteria, including M. tuberculosis. These include, amongst others, the biosynthesis of lipids, the
production of a number of small molecules, parts of important connections within central carbon
metabolism, as well as roles in pH homeostasis. In cells, CO2 exists in equilibrium with carbonic
acid (H+ + HCO3-), a distinction that is important for many of these enzymatic and physiological
functions. The flux between CO2 and HCO3- needs to be sufficient to supply these processes,
however, as the rate constant for the hydration of CO2 is relatively low many organisms produce a
carbonic anhydrase. This requirement for CO2/HCO3 flux means that carbonic anhydrases have
been investigated as potential drug target. The M. tuberculosis genome encodes three such
carbonic anhydrases.
In this study one of these carbonic anhydrases, Rv1284c, was knocked out, to give a mutant that
exhibited a series of unexpected phenotypes:
1. The ΔRv1284 mutant was hyper-virulent in the THP-1 macrophage model of infection.
2. The ΔRv1284 mutant had an enhanced ability to grow on six carbon sugars.
3. The amount of CO2 incorporated into the cell, as measured by C13 analysis of proteogenic
amino acids, rose in ΔRv1284 when compared to wild type.
All these phenotypes could be complemented, and genomic analysis did not identify any SNP likely
to result in such behaviour. These seemingly unrelated phenotypes of macrophage virulence,
glycolysis and CO2 incorporation suggest a hitherto unrecognised connection in M. tuberculosis
physiology.
156
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
135 Mycobacterial Biology & Physiology
20/09/2016
Molecular Basis of Heme Acquisition by Mycobacterium tuberculosis
A. Mitra2, A. Speer1, M. Niederweis2
1
Vrije University Amsterdam, Amsterdam, The Netherlands 2Microbiology, University of Alabama at
Birmingham, Birmingham, United States
Mycobacterium tuberculosis (Mtb) is primarily transmitted through aerosols which are taken up in
the lung by alveolar macrophages. However, Mtb efficiently inhibits macrophage maturation and
phagolysosome formation. Acquisition of iron within the macrophage is essential for Mtb survival
and virulence. Although iron is abundant it is efficiently sequestered in the human host, e.g. 90-95%
of iron is bound in heme and hemoproteins making heme the major iron source in humans. Mtb can
acquire iron through production of siderophores called mycobactins, which can solubilize iron from
host transferrin or other iron proteins but not from heme. While siderophore-mediated iron
acquisition is well studied in Mtb, knowledge about utilization of heme by Mtb is limited. In order to
identify molecular components involved in heme acquisition by Mtb we exploited the toxicity of
Gallium (III). Ga-PIX was synthesized and a high-density Mtb transposon library was screened for
resistance to Ga-PIX. Deletion of two canonical Mtb ppe genes resulted in complete resistance to
Ga-PIX relative to wild-type Mtb. Deletion of one ppe gene partially decreased heme utilization,
while deletion of the other ppe gene completely abrogated growth of Mtb on heme. Surface plasmon
resonance (SPR) spectroscopy indicated that both ppe genes encode heme-binding proteins.
Subcellular localization and surface detection experiments indicated that these PPE proteins are
cell surface-accessible membrane proteins. These experiments provide the first evidence for cell
surface heme-receptors of Mtb and establish a novel role of PPE proteins in Mtb physiology.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
157
136 Mycobacterial Biology & Physiology
20/09/2016
Towards understanding the molecular basis of PknB essentiality in mycobacteria
O. Turapov3, F. Forti1, D. Ghisotti1, P. Ajuh4, S. Waddell2, W. Vollmer5, G. Mukamolova3
1
Università degli Studi di Milano, Milano, Italy 2University of Sussex, Brighton 3University of
Leicester, Leicester 4Gemini Biosciences Ltd, Liverpool 5Newcastle University, Newcastle, United
Kingdom
Tuberculosis is a global infectious disease affecting 8 million people and resulting in 1.5 million
deaths annually. Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, has
evolved mechanisms for evasion of host immune system and can persist in vivo without causing an
active disease. Eleven serine/threonine protein kinases control Mtb growth, persistence and cell
wall biosynthesis. PknB is an essential serine/threonine protein kinase required for mycobacterial
growth, however the molecular basis of its essentiality remains unknown. We have designed a
special medium for supporting growth of PknB-depleted Mtb and conducted omics analyses of
PknB-depleted and PknB-producing Mtb bacilli. Our phosphoproteomics data indicate that 47
proteins were up-phosphorylated in the PknB producing strain compared to the PknB-depleted
strain and 9 out of them have been previously annotated as essential proteins. Our transcriptome
analysis showed that the depletion of pknB altered expression of 48 genes. Based on our results we
propose that PknB is dispensable for growth and division itself but it plays a critical role in
coordinating cell wall biosynthesis and cell growth. We are currently validating the biological
significance of phosphorylation of PknB substrates identified in our study. Elucidation of the
molecular mechanism of PknB essentiality will offer novel opportunities for treatment and prevention
of tuberculosis.
158
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
137 Mycobacterial Biology & Physiology
20/09/2016
Novel method to improve recovery of mycobacteria from difficult samples
T. Munshi, P.M. Lopez-Perez, J. Garcia, S. Hofmann, K. Hilpert, T. Bull
Institute for infection & immunity, St. George's, University Of London, London, United Kingdom
Isolation of mycobacteria is the gold standard diagnostic technique. Slow division rates combined
with stringent decontamination methods drastically slow down mycobacterial growth and require
long incubation times. In children, respiratory sampling can be difficult and faecal/ urine samples are
prone to high contamination rates. New more efficient methods are called for to decontaminate and
stimulate mycobacterial growth from samples. Here, we describe an innovative approach to
decontaminate and culture mycobacteria from difficult samples using sequential combinations of
antimicrobial compounds that kill contaminating flora while promoting mycobacterial growth.
We screened for a complex combination of antimicrobial compounds (TiKa-Kic) against bacteria
(Gram positive and negative) and fungi whilst having no deleterious effect on mycobacterial
species. We evaluated the overnight TiKa-Kic treatment of faeces spiked with Mycobacterium
tuberculosis (MTB), followed by culture in MGIT 320 detection system supplemented with a different
growth stimulating compound (TiKa-B08). This sample preparation method was highly effective at
decontaminating samples and gave rapid MTB growth with time to positivity in ~10 days for 1x106
organisms, ~16 days for 1x104 organisms, ~25 days for 1x102 organisms with no background
contamination. As a pilot study, we also used this system to culture mycobacteria from human and
animal (tissue, sputum, faeces and urine) samples with known mycobacterial disease. We were
able to isolate M. tuberculosis, M. bovis and M. avium subspecies paratuberculosis from these
samples in MGIT system as well as on solid media plates without any contamination.
The method provides an innovative and convenient technique that offers significant progress in
isolation of mycobacterial species from difficult samples and improving timely confirmatory
diagnoses. The technique is especially relevant for testing faecal samples from young children with
suspected MTB infection who tend to swallow rather than expectorate their sputum.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
159
138 Mycobacterial Biology & Physiology
20/09/2016
Supercoiling sensitive transcription and topology –transcription coupling in
Mycobacterium tuberculosis
W. Ahmed1, C. Sala2, S. Hegde1, S. Cole2, V. Nagaraja1
1
Department Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India 2Global
Health Institute, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
Transcription relies on DNA topology at different steps of the process. During initiation, the promoter
melting is facilitated by negative supercoiling of the template. Next, during transcription elongation,
positive and negative supercoils generated ahead and behind the moving elongation complex have
to be removed by the action of two different topoisomerases, namely DNA topoisomerase I and
DNA gyrase respectively. Thus, to maintain the topological homeostasis of the genome and to
maintain genome architecture, DNA topoisomerases catalyse DNA cleavage, strand passage and
rejoining of the ends. The transcription of the genes encoding topoisomerases are in turn regulated
in response to alterations in genome supercoiling. Gyrase expression is up-regulated by relaxation
stimulated transcription (RST) by distinct mechanisms in E.coli, Mycobacterium tuberculosis and
Mycobacterium smegmatis. More recently, we have uncovered additional regulatory mechanism of
gyr operon regulation in M.tuberculosis involving a process termed as reiterative transcription.
However, the molecular mechanism of transcriptional regulation of Topoisomerase I is not studied
so far. We describe supercoiling sensitive transcription (SST) of the topoI gene. The expression is
regulated by high transcription of the upstream gene and the promoter architecture of the topoI.
Perturbation of topology induced in the conditional knock down strains of these essential enzymes
affect transcription initiation and elongation leading to several pleiotropic downstream
consequences. To understand further the in vivo roles of DNA gyrase and Topoisomerase I in
chromosome dynamics and gene expression, we have carried out a genome-wide occupancy
studies of these enzymes along with RNA polymerase. These data provide the first glimpse of
topoisomerase traffic on the transcription units in vivo. The pattern of the distribution profile of RNA
polymerase and the topoisomerases suggests the operation of twin - supercoiled domain during
transcription validating the long standing model.
160
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
139 Mycobacterial Biology & Physiology
20/09/2016
Iron sensing via a catalytically divergent rhomboid protease
S. Nambi, C. Sassetti
Umass medical school, Worcester, United States
Environmental sensing in prokaryotes can be accomplished by the release of regulatory proteins
from the membrane following two sequential proteolytic events. In Mycobacterium tuberculosis
(Mtb), Rip1, a zinc metalloprotease similar to other described site-2 proteases mediates the final
release of anti-sigma factors as part of this process. However, site-1 protease(s) that sense
environmental signals and initiate the cascade have not been identified in Mtb. We report that a
functionally non-annotated and highly divergent member of the rhomboid family of intramembrane
proteases performs this activity. Despite lacking a canonical rhomboid active site, Rv3193c (RomA)
uses distinct catalytic residues to perform the site 1 cleavage of the anti-sigma factor (RsmA),
priming it for processing by Rip1. A physical interaction between the PDZ domain of Rip1 and a
proline-rich region in extracytoplasmic domain of RomA facilitate the signaling cascade. Structural
analysis of the extracytoplasmic domain revealed a conserved iron coordination site, indicating a
likely environmental trigger for this system. Indeed, we found that RsmA processing was altered by
extracellular iron concentration, and RomA-deficient bacteria were unable to replicate in low iron
conditions due to altered levels of iron homeostatic genes. The unique topology, active site, and
sensory domains of catalytically divergent rhomboid family (cdRhom) impart an environmental
sensing function that is unprecedented for this family of proteins.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
161
140 Mycobacterial Biology & Physiology
20/09/2016
Fishing for bortezomib mechanisms of resistance in mycobacteria
G.J.Y. Ngan, W. Moreira, T. Dick
Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National
University of Singapore, Singapore, Singapore
We previously identified bortezomib, a human 26S proteasome drug, as a potent inhibitor of
ClpP1P2 activity and bacterial growth in mycobacteria. Here we aimed to elucidate mechanism of
resistance of bortezomib in mycobacteria. We selected spontaneous resistance mutant of
bortezomib in Mycobacterium smegmatis and characterized them. We ruled out the genetic
modification and changes in expression level of the target ClpP1P2, and the adaptor proteins and
the regulator of Clp (ClpC1, ClpX, clgR). Phenotypic characterization of bortezomib mutants
suggests correlations between resistance level, growth, ATP level, membrane potential and
resistance to a panel of other antibiotics (Rifampicin, Vancomycin, Isoniazid, Bedaquiline and
Ethionamide). Based on these observations, we speculate that alteration of cell envelope
permeability may contribute to resistance. Currently, we employ whole genome sequencing to
determine the mechanism of resistance of mutants.
Acknowledgements: This work was supported by the Singapore Ministry of Health’s National
Medical Research Council grant (NMRC/ CBRG/0022/2012) and its Centre grant MINE / Research
core no. 4 (NMRC/CG/013/2013) to T.D., and is part of the Singapore Programme of Research
Investigating New Approaches to Treatment of Tuberculosis (SPRINT-TB; www.sprinttb.org) led by
Nick Paton. G.N. receives a SMA3 fellowship from Singapore MIT Alliance for Research and
Technology (SMART).
Reference: Moreira W, Ngan GJY, Low J. L, Poulsen A, Chia BCS, Ang MJY, Yap A, Fulwood J,
Lakshmanan U, Lim J, Khoo AYT, Flotow H, Hill J, Raju RM, Rubin EJ, Dick T. 2015.Target
mechanism-based whole-cell screening identifies bortezomib as an inhibitor of caseinolytic protease
in mycobacteria. mBio 6(3):e00253-15. doi:10.1128/mBio.00253-15.
162
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
141 Mycobacterial Biology & Physiology
20/09/2016
Significant under expression of the DosR regulon in M. tuberculosis complex Lineage 6 in
sputum
B. Ofori-Anyinam1-2, G. Dolganov6, T. Van6, L. Davis5, N. Walter4, B. Garcia4, M. Voskuil4,
K. Fissette1, M. Diels1, M. Driessen1, C. Meehan1, M. Coscolla3, S. Gagneux3, M. Antonio2,
G. Schoolnik6, F. Gehre1-2, B. De Jong1
1
Institute of Tropical Medicine, Antwerpen, Belgium 2Medical Research Council Unit, The Gambia,
Fajara, Gambia 3Swiss Tropical and Public Health Institute, Basel, Switzerland 4University of
Colorado Denver, Aurora, Colorado 5Yale University, New Haven, Connecticut 6Stanford University,
Stanford, California, United States
Background: Mycobacterium africanum L6 is associated with HIV-infection, extrapulmonary
disease, and with slower progression to active tuberculosis disease as compared with
Mycobacterium tuberculosis sensu stricto. The biology underlying these clinical differences remains
poorly understood. Within the host, the DosR regulon of M. tuberculosis complex allows for quick
adaptation to oxygen limitation and genetic mutations within the regulon leads to growth and
virulence attenuation. Early literature indicated a preference of M. africanum for microaerobic
growth. Therefore, we suspected that the dormancy regulon would be differentially induced between
M. tuberculosis and M. africanum L6.
Methods: We compared the ex vivo expression of 2179 genes of M. tuberculosis and M. africanum
L6 in sputa from 9 HIV-negative TB patients from The Gambia, six with M. tuberculosis and 3 with
M. africanum L6. Further, we compared polymorphisms in the differentially expressed genes, as
well as related genes, in 44 M. africanum L6 genomes from TB patients from The Gambia and
Ghana. Lebek’s test was used to confirm differences in oxygen requirements for growth.
Results: All genes within the DosR regulon had lower expression in M. africanum L6 compared to
M. tuberculosis with half (n=26) being significantly under expressed (Modified Fisher’s exact p-value
=9.59E-13, Bonferroni Adjusted p-value = 1.06E-11). Lineage 6 defining nonsynonymous mutations
were detected in PhoR as well as the intergenic region of Rv0080 and the foremost gene regulator
in M. tuberculosis during hypoxia, Rv0081. In Lebek’s test, M. tuberculosis grew only at the aerobic
surface while M. africanum L6 grew throughout the medium.
Conclusions: Based on the relative under expression of DosR regulon genes, M. africanum L6
appears to have adapted to growth under hypoxic conditions potentially through DosR-independent
mechanisms or to different biological niches. These findings fit with genomic changes in related
genes and a preference for microaerobic growth as well as the association with extrapulmonary
disease.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
163
142 Mycobacterial Biology & Physiology
20/09/2016
Evaluation of Genotype MTBDRsl version 2 for rapid detection of second-line antituberculosis drug resistance in Ghana identifies the first pre-XDR TB case
S. Osei-Wusu2-3, A. Asante-Poku2, I. Otchere Darko2, S. Omari1, A. Forson1, D. Yeboah-Manu2
1
Department of Chest Diseases, Korle-Bu Teaching Hospital 2Noguchi Memorial Institute for
Medical Research, University of Ghana, Legon, Ghana 3West Africa Centre for Cell Biology of
Infectious Pathogens, University of Ghana, Legon, Ghana, Accra, Ghana
Background: The emergence of drug resistance TB strains requires rapid but sensitive and specific
tools for control. We evaluated the genotype MTBDRsl v2 for detecting second-line anti-TB drug
resistance in Ghana.
Method: We screened 117 resistant TB isolates against moxifloxacin (MOX), streptomycin (STR)
and amikacin (AMK) using the Epsilometer test (Etest) and with the MTBDRsl rapid kit. Drug
resistant conferring genes (gyrA, gyrB, eis, rrs, tap, whiB7 and tlyA) of strains resistant to MOX or
AMK were PCR amplified and sequenced for mutation analysis.
Results: Isolates phenotypically resistant to STR, AMK and MOX were 38 (32.5%), 13 (11.1%) and
1 (0.9%) respectively. Among the isolates, only 1 multidrug resistant (MDR) isolate was diagnosed
resistant to both MOX (MIC=3µg/mL) and AMK (MIC =16µg/mL) by both the phenotypic assay and
MTBDRsl LPA. Gene sequencing analysis led to the identification of gyrA (G87C) and rrs (A514C
and A1401G) in this isolate. Whereas none of the remaining isolates were resistant to MOX
(MIC<0.125µg/mL), 9/76 (11.8%) MDR, 1/14 (7.1%) rifampicin resistant and 3/27 (11.1%) isoniazid
resistant isolates were resistant to AMK with MICs ranging from 1 to 8µg/mL. However, none of
these were identified as resistant to AMK by the MTBDRsl LPA. Except one MDR isolate harbouring
a tlyA mutation (N236K), no other mutation was detected. The agreement between the phenotypic
and the LPA was 87.1% and 100% whereas the sensitivity/specificity was 7.7%/100% and
100%/100% for AMK and MOX respectively.
Conclusion: We report the first instance of a pre-XDR-TB case in Ghana. Our findings support the
use of the MTBDRsl v2 for detecting second anti-TB drug resistance in Ghana.
164
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
143 Mycobacterial Biology & Physiology
20/09/2016
Single-cell growth and antibiotic tolerance in mycobacteria
M. Priestman1, B. D. Robertson1, V. Shahrezaei2
1
Centre for Molecular Bacteriology and Infection 2Department of Mathematics, Imperial College
London, London, United Kingdom
Drug tolerance is the phenomenon by which bacteria that are otherwise genetically susceptible to
an antibiotic are able to survive extended treatment. This is usually associated with growth arrest
and shutdown of protein synthesis, to form 'persister' cells. Persister cells form a small subset of
genetically homogenous populations, with a phenotypic switch triggering the entry and exit into
persistence.
Persistence in mycobacteria remains relatively poorly characterised, and given the lengthy
treatment regime required to completely eradicate M. tuberculosis infections, understanding how
mycobacteria enter into this drug-tolerant state may be of great value for drug development and
strategies.
Microfluidics and timelapse microscopy allows the tracking of single cells and their subsequent fate
upon exposure to antibiotics. Growth conditions can be rapidly changed, and antibiotics added at
specific timepoints, allowing the precise control of the environment, revealing how growth affects
survival to antibiotics at the single cell level.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
165
144 Mycobacterial Biology & Physiology
20/09/2016
Quiescence survival in Mycobacterium tuberculosis requires a cAMP mediated metabolic
switch
E. Rittershaus, S. Baek, S. Nambi, C. Sassetti
Microbiology & Physiological Systems, Sassetti Lab, University of Massachusetts Medical School,
Worcester, Massachusetts, United States
Upon exposure to respiratory stress imparted by the host, e.g. nitric oxide or hypoxia, M.
tuberculosis slows its growth and is relatively unresponsive to antibiotic treatment. In this study, we
utilized a combination of metabolite profiling in tandem with comprehensive mutant fitness analysis
to identify critical regulatory or metabolic pathways that could be targeted to kill quiescent cells.
Metabolomics revealed alterations in abundance of TCA intermediates that correlated with
increased cAMP. The parallel genetic study identified the responsible adenylate cyclase (Rv1359)
and a cAMP-binding protein acetyl-transferase (Pat/Rv0998) that were conditionally essential for
survival in hypoxia. 13C-labeled carbon tracing in hypoxia revealed that this cAMP pathway
mediates a switch in the TCA cycle from oxidative to reductive reactions. Inhibition of Rv1359 and
Pat to make this metabolic shift resulted in over-reduction and reduced viability. Genetic inhibition of
TCA enzymes verified the importance of reductive reactions in hypoxic cultures and murine lung.
Under both conditions inhibition of reductive TCA resulted in rapid cell death, suggesting a new
strategy for eradication of quiescent bacteria.
166
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
145 Mycobacterial Biology & Physiology
20/09/2016
Identifying the critical oxygen response regulators using a novel regulator overexpression
mutant pool screen
T. Rustad1, J. Farrow-Johnson1, K.J. Minch1, D.R. Sherman1-2
1
Center for Infectious Disease Research 2Department of Global Health, University of Washington,
Seattle, United States
Hypoxia is one of the stresses encountered by M. tuberculosis (MTB) during the course of infection
and is associated with latent disease. In response to lowered oxygen tension, MTB alters the
expression of hundreds of genes, including dozens of transcription factors (TFs), any of which could
be participating in generating the hypoxic response. The initial transcriptional adaptation to hypoxia
is mediated by the TF DosR, but little is known about the regulators of the much larger subsequent
transcriptional signature.
To identify which TFs are critical for the adaptation to hypoxic stress, we developed a regulator
overexpression mutant pool (ROMP) assay to identify TFs with stress-specific phenotypes. This
method involves a pool of 207 MTB mutants that each contain a TF under control of a tetracycline
inducible promoter. This approach allowed us to screen the regulons associated with all known TFs
for growth phenotypes in response to reduced oxygen tension, using high throughput sequencing of
mutant-specific tags to follow all 207 mutants in a single experiment. This ROMP assay identified
five critical oxygen responsive regulators (CORRs) with inhibited growth when recovering from
hypoxic stress.
Using our previously generated transcriptional regulatory map we found that these CORR TFs coregulate a significant number of genes, suggesting that these TFs operate as a tightly knit
regulatory subnetwork. Finally, we show that the majority of the genes that change expression in
response to hypoxia interact with one or more of the CORRs.
In summary, this work presents a novel approach that can be used to screen for stress-specific TF
phenotypes as well as a previously undescribed hypoxic response regulatory network.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
167
146 Mycobacterial Biology & Physiology
20/09/2016
A two-stress model for the generation of viable M. tuberculosis cells that do not form
colonies
K. Saito2, T. Warrier1, S. Somersan-Karakaya2, J. Mi1, S. Park1, X. Jiang1, B. Gold1, J. Roberts1,
C. Nathan1
1
Department of Microbiology and Immunology, Weill Cornell Medicine 2Department of Medicine,
Division of Infectious Diseases, Weill Cornell Medicine/NY Presbyterian Hospital, New York, United
States
Despite the need for more rapidly effective therapy for tuberculosis, limited understanding of
phenotypically drug-tolerant states of Mycobacterium tuberculosis (Mtb) hampers the development
of new drugs. Non-replicating Mtb becomes relatively or absolutely tolerant to most TB drugs.
Among phenotypically tolerant and temporarily non-replicating Mtb populations are cells that have
lost the ability to grow as CFU on agar and yet are proven viable in other ways, marking them as
“differentially detectable” (DD). Some DD Mtb can be detected and quantified by culturing replicate
limiting dilution (LD) series in liquid medium, with or without fresh Mtb culture filtrate (CF). The
highest dilution that produces growth is used to back-calculate the original quantity. A statistical
formula can adjust for differences in the dilution limit seen in each replicate series to identify the
most probable number (MPN). LD will exaggerate the MPN if clumps of cells break up during
dilution. This artifact handicaps study of DD Mtb despite its potential clinical importance, as does the
inconsistent impact of CF, the divergent identifications of effector molecules in CF, and the lack of
confirmation of reported methods for generating DD Mtb in vitro. In an effort to minimize such
obstacles, we studied numerous technical variables—first with manual handling and then with
robotics—until quantification of Mtb by CFU and by LD produced indistinguishable numbers, except
after certain treatments. We then tested nutrient starvation, hypoxia, acidification, human plasma,
neutrophils or their lysates, reactive nitrogen species, TB drugs and prolonged stationary phase for
their ability to induce DD Mtb. We could only reproducibly generate populations of >=90% DD Mtb
with two stresses in sequence: starvation followed by exposure to rifampin. The proportion of DD
Mtb increased with the duration of starvation. Longer drug exposure and longer starvation allowed
DD Mtb generation with less rifampin. CF had no effect. This model supports two lines of inquiry.
First, we are trying to identify genes required for entering or sustaining the DD Mtb state. Second,
we are testing compounds that can kill Mtb in other states and classifying whether they generate, kill
or have no effect on DD Mtb.
168
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
147 Mycobacterial Biology & Physiology
20/09/2016
Non-replicative in vitro tests for Tuberculosis drug research
S. Sans1, B. Gold3, H. Boshoff2, L. Gouilleux1, C. Couturier1, K. Kissane1, I. Blanc1, K. BurnsHuang3, L. Lopez Quezada3, E. Ballinger3, J. Roberts3, Y. Ling3, D. Zhang3, G. Mondesert1,
E. Bacqué1, L. Fraisse1, C. Nathan3, S. Lagrange1, C. Roubert1
1
TSU Infectious Disease, Sanofi R&D, Marcy L'etoile, France 2Tuberculosis Research Section,
NIAID, Bethesda 3Departments of Microbiology & Immunology, Weill Cornell Medical College, New
York, United States
Tuberculosis (TB) is the first leading infectious cause of death worldwide and WHO estimates that
one third of the world population is silently carrying M. Tuberculosis (Mtb), the etiological agent of
tuberculosis.
This pathogen is highly adapted to humans, its exclusive host. Humans and Mtb have co-evolved
for more than 70000 years. In most cases of primary infections, the human immune system is able
to clear the bacteria or at least “trap” the bacteria, thereby controlling the disease. However, Mtb
has developed a “persister” state in which its metabolism is minimized in order to escape the host
immune system, to tolerate drug treatment and to adapt to different micro-environments. These socalled Mtb persisters are believed to be the potential source of reactivation of active TB, years after
primary infection. As a consequence, a complete cure of tuberculosis requires a combination of
drugs that not only inhibit growth of replicating bacteria but also kill the persister bacteria.
For characterizing new compounds with potential against the persister sub-population, multiple nonreplicative (NR) assays using different stress factors as well as multiple readouts have been
reported. In this poster, we will describe, compare and highlight pros and cons of these in vitro
models of non-replicative bacteria, using standard anti-tuberculosis drugs.
Furthermore, in order to develop new anti-tuberculosis drugs active on NR Mtb, we have screened a
80 K compound library against a Mtb strain under different conditions designed to mimic four
physiological stresses encountered by Mtb in one or more of its microenvironments in the host:
acidity, reactive nitrogen intermediates, hypoxia, and fatty acid as a carbon source (Gold et al.;
2015). Following active hit triaging, we have been able to identify several series that display specific
patterns under the different NR conditions.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
169
148 Mycobacterial Biology & Physiology
20/09/2016
EspR-dependent ESAT-6 secretion of Mycobacterium tuberculosis requires the presence of
virulence-associated response regulator PhoP
V. Anil Kumar, R. Goyal, R. Bansal, N. Singh, A. Kumar, D. Sarkar
Microbiology and biochemistry laboratory, CSIR - Institute of Microbial Technology, Chandigarh,
India
M. tuberculosis (Mtb) uses the ESX-1 secretion system to transport virulence factors in host cell.
ESX-1 secretion system is encoded by the genes of the esx-1 locus, which is highly conserved in
members of the Mtb complex and in other pathogenic mycobacteria. Attenuation of M. bovis BCG
strain is related to the loss of RD1-encoded ESX-1 secretion system. M. tuberculosis ESX-1 system
secretes virulence factor ESAT-6 which plays a critical role in modulation of host immune system for
establishment of productive infection. Previous studies suggest that among the reasons for
attenuation of M. tuberculosis H37Ra is a mutation in the phoP gene that interferes with the ESX-1
secretion system and inhibits secretion of ESAT-6. Here, we identify a totally different and distinct
regulatory mechanism involving PhoP and transcription regulator EspR on transcriptional control of
the espACD operon, which is required for ESX-1-dependent ESAT-6 secretion. While both these
regulators are capable of influencing espACD expression, we show that activation of espACD
requires direct recruitment of both PhoP and EspR within the promoter. The most fundamental
insights derive from the inhibition of EspR binding at the espACD regulatory region of phoP mutant
strain owing to PhoP-EspR protein-protein interactions. Based on these results a model is proposed
suggesting how PhoP and EspR protein-protein interactions contribute to activation of espACD
expression, and in turn, controls ESAT-6 secretion, an essential pathogenic determinant of M.
tuberculosis. Together, these results have significant implications on the mechanism of virulence
regulation of M. tuberculosis.
170
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
149 Mycobacterial Biology & Physiology
20/09/2016
Investigating the cost of rifampicin-resistance in mycobacteria
B. Sewgoolam, A. Koch, V. Mizrahi, D. Warner
Dept. Pathology, Molecular Mycobacteriology Research Unit, University of Cape Town, Cape
Town, South Africa
Rifampicin (RIF), the front-line anti-tubercular drug, targets the rpoB-encoded β-subunit of RNA
polymerase (RNAP), blocking transcription. In Mycobacterium tuberculosis (Mtb), mutations in rpoB
are primarily associated with RIF resistance (RIFR), which poses a major threat to tuberculosis (TB)
control. Some rpoB mutations have been associated with fitness costs, however the observed
phenotypes are often dependent on strain background and the assay system used to measure
them. Therefore, determining the implications of RIFR-associated rpoB mutations for mycobacterial
physiology is essential for understanding the development and maintenance of RIFR, as well as the
spread and pathogenesis of drug resistant strains of Mtb. In other bacterial systems, mutations in
rpoB have been associated with alterations to core bacterial functions and physiology. Similarly, in
Mtb, recent reports have identified changes to components of the cell wall, the proteome, and fatty
acid metabolism; increased expression of dnaE2, encoding a DNA damage-inducible mutagenic
repair polymerase; and increased ofloxacin minimum inhibitory concentrations (MICs). In all studies
to date, RIF exposure was used to select the resulting rpoB mutant strains; therefore, it is unknown
whether secondary mutations, or potentially even epigenetic effects, might have influenced the
assay outcome. To address this potential limitation, we have applied site-directed mutagenesis and
two-step allelic exchange to insert a RIFR SNP into the native Mtb rpoB gene in the absence of RIF
selection. Whole-genome sequencing will be used to determine whether the defined rpoBS531L
(TCG->TTG) mutant contained any secondary mutations. Thereafter, this RIF naïve strain will be
applied in standard microbiological and immunological assays to investigate the impact of a
clinically relevant rpoB mutation on functions unrelated to drug resistance. Priority assays include
characterization of the transcriptome via RNA-seq, and macrophage survival assays. In addition, a
flow cytometry assay is being optimized in parallel for corresponding mutants of M. smegmatis that
will allow culture free determination of cell numbers in growth-based assays. This will allow for
accurate determination of the fitness of the rpoB mutant strain in competition with the wild-type
parental strain, under a range of growth conditions that are considered relevant to host infection.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
171
150 Mycobacterial Biology & Physiology
20/09/2016
Serine/threonine protein phosphatase of Mycobacterium tuberculosis is necessary for
precise cell division
A.K. Sharma2, D. Arora3, L.K. Singh2, A. Sajid2, V. Molle1, Y. Singh2, V.K. Naandicoori3
1
CNRS-Universite de Montpellier II, Montpellier, France 2CSIR-Institute of Genomics and
Integrative Biology 3National Institute of Immunology, Delhi, India
Signal sensing and transduction leads to a wide range of cellular responses, and thus must be
tightly regulated in pathogenic bacteria to allow optimal survival under variable conditions. A
common mode of regulation of the cell’s response to external cues is via
phosphorylation/dephosphorylation of specific target proteins that lead to cellular responses like
altered subcellular localization of proteins, protein turnover rates, and protein-protein interactions.
The analysis of the M.tuberculosis whole genome sequence identified the presence of 11
eukaryotic-like STPKs, one Ser/Thr phosphatase (PstP), one tyrosine kinase (PtkA), and two
tyrosine phosphatases (PtpA and PtpB). All these kinases and phosphatases have now been
characterized and found to be catalytically active. The lone serine/threonine phosphatase of M.
tuberculosis, PstP belongs to Metal- dependent family of phosphatase whose members strictly
require Mn2+ ion for their activity. While two metal centers are found in the catalytic core in most
PP2C phosphatases, crystal structural of PstP showed presence of three metal-binding centers.
The conserved active site residues in D38 and D229 are involved in metal binding and R20 involved
in binding with the phosphate moiety in the target proteins, bridging together to stabilize the active
structure. Previous data from our laboratory have shown that PknA and PknB phosphorylate PstP
on specific residues in its cytosolic domain, and this phosphorylation is influenced by the presence
of Zn2+ ions and inorganic phosphate (Pi). The phosphatase-dead PstP mutant PstPcD38G is more
efficiently phosphorylated by PknA and PknB. Importantly, we found that the phosphorylated PstP is
more active compared with its unphosphorylated counterpart, suggesting a possible reverse
regulation mediated through phosphorylation cascades by STPKs. Investigations carried out thus
far are in-vitro studies using purified PstP. The present study aims to investigate the impact of PstP
overexpression or depletion scenario on the growth of the pathogen and its survival within the host.
Using these mutants we investigated the role of various domains of PstP in modulating cellular
events, cell division and host adaptation. Also we identified mechanistic pathway important for cell
division of M.tuberculosis.
172
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
151 Mycobacterial Biology & Physiology
20/09/2016
Survival strategies of Mycobacterium tuberculosis: Insights from the vitamin C dormancy
model
K. Sikri, S. Batra, M. Nandi, J. Tyagi
TB Molecular Biology Lab, Department of Biotechnology, All India Institute of Medical Sciences,
New Delhi, India
Mycobacterium tuberculosis (Mtb) is an intracellular pathogen that successfully resides in the hostile
phagosomal environment by resisting multiple stresses including hypoxia, low pH, nutritional
deficiency, and oxidative and nitrosative stresses. The success of tubercle bacilli comes from their
ability to employ strategies that enable their adaptation to a state of ‘dormancy’/ persistence. A
serendipitous discovery was made in our laboratory that vitamin C (vit C) triggered the induction of
the DevR ‘dormancy’ regulon in axenic cultures of Mtb, owing to its O2 scavenging properties. Vit C
also induced bacteriostasis and an isoniazid-tolerant phenotype, an accepted indicator of bacterial
adaptation to hypoxia. Further, vitC-treated cultures developed a ‘viable but non-culturable’
antibiotic-tolerant state, which was reversed on withdrawal of vit C. Vit C is reported to have a
significant effect on macrophage physiology and on the outcome of tuberculosis infection.
Therefore, a global gene expression analysis was undertaken to decipher the molecular events
underlying the adaptive response of Mtb to vit C. A massive transcriptional response was noted in
axenic cultures that included the DevR ‘dormancy’ regulon, triacylglycerol synthases, efflux pumps
and oxidative stress response genes. These responses were consistent with development of a state
of ‘dormancy’/ persistence in Mtb. Notably, the transcriptional response to vit C mimicked bacterial
responses to (1) multiple intracellular stresses, thus generating a multiple stress environment for the
bacterial cell, and more importantly (2), an activated macrophage-like condition. These findings
signify that vitamin C has wide-ranging regulatory effects on Mtb and offers a new perspective on
the possible contribution of vit C to Mtb adaptation. In the current scenario of TB disease, vit C
could either modulate the efficacy of anti-tubercular drugs or contribute to TB control among
immunocompromised individuals, as in HIV subjects.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
173
152 Mycobacterial Biology & Physiology
20/09/2016
Investigating the role of toxin-antitoxin systems in physiology and persistence of
Mycobacterium tuberculosis
P. Tiwari, S. Agarwal, S. Kidwai, R. Singh
Vaccine and Infectious Disease research centre, Translational Health Science and Technology
Institute, Haryana, India
Toxin-antitoxin (TA) systems are highly conserved in members of the Mycobacterium tuberculosis
(Mtb) complex and have been postulated to play an important role in physiology and virulence. By
overexpressing each of the putative toxins in M. bovis BCG using inducible systems, we have
identified functional toxins. We report that overexpression of these TA systems is bacteriostatic and
few of these also disrupt the membrane integrity. We have constructed various TA deletion mutants
of Mtb to understand their role in physiology and persistence. The construction of various single-,
double- and triple-mutant strains revealed that these ribonucleases contribute synergistically to the
ability of Mtb to adapt to conditions such as oxidative stress, nutrient depletion and drug exposure.
The genes induced in strains overexpressing these ribonucleases were identical to those induced in
persister, hypoxic and nutritionally starved bacteria. Moreover, guinea pigs infected with TA systems
deficient strains exhibits significantly reduced bacterial loads and pathological damage in infected
tissues in comparison to parental strain-infected guinea pigs. Currently, experiments are in progress
to identify cellular targets for these TA systems and to understand the cellular mechanisms by which
these TA systems are activated. The present study highlights the importance of TA systems in Mtb
stress adaptation, drug tolerance and virulence.
174
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
153 Mycobacterial Biology & Physiology
20/09/2016
Genetic geography of mycobacterium tuberculosis beijing genotype
V. Slizen1, L. Surkova2, A. Zalutskaya2
1
Microbiology, Virology, Immunology, Belarusian State Medical University 2Republican ScientificPractical Center of Pulmonology and Tuberculosis, Minsk, Belarus
High disease rate of extended and multi-drug resistant tuberculosis (XDR, MDR) in some countries
may be associated with the spread of highly virulent genetic variants, such as Beijing, the proportion
of which in the TB structure can vary from depending on the geographical region. The aim was to
identify genotypes of pre-XDR strains of MTB, isolated from patients in Belarus.
Methods: Genotypes of sixty five pre-XDR strains of MTB were detected by MIRU-VNTR typing
method based on the evaluation of differences in 15 VNTR-loci. Mutations in the gene gyrA of MTB
were detected by sequencing as well as by GenoType MTBDRsl (HAIN), qPCR with hydrolysis
probes.
Results: It was shown the genotype Beijing to be dominant, comprising 58% of all pre-XDR MTB.
MTB, belonging to genotype Beijing, carried nucleotides ACC but not AGC in codon 95 of gene
gyrA. Haarlem and TUR genotypes were detected in 23 and 16% of isolates, respectively. Pre-XDR
strains of MTB belonging to genotype Beijing demonstrated sufficient genetic homogeneity and had
identical MIRU-VNTR profile 2-3-5-3-3-5-4-4-4-2-3-4-6- 7-5 (sequence loci MIRU04- MIRU40MIRU26- MIRU10- MIRU16- MIRU31- ETRA- ETRC- Mtub04- QUB4156- MTUB39- MTUB30QUB11b- QUB26- MTUB21). MTB, belonging to genotype Haarlem and TUR were mostly with 2-35-5-3-3-3-3-2-3-3-4-3-3-3 and 1-2-1- 5-1-3-3-4-4-3-3-4-2-6-2 MIRU-VNTR profiles, respectively. Out
of 65 pre-XDR isolates, 83% were found to harbour mutations within gyrA. The most common
mutations were observed at codon 94 (n =35), where the wild-type aspartic acid was replaced with
glycine (Asp94Gly; n =19), alanine (Asp94Ala; n = 7), or tyrosine (Asp94Tyr; n = 7) or histidine
(Asp94His; n=1).
Conclusion: In Belarus, as in other post-Soviet countries, the epidemic process of TB is mainly
sustained by genotype Beijing. Continuous circulation of genotype Beijing results in the
accumulation of mutations and increases the proportion of MDR and XDR.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
175
154 Mycobacterial Biology & Physiology
20/09/2016
Investigation of the spatio-temporal dynamics of peptidoglycan synthesis in
Mycobacterium tuberculosis
J. Vaubourgeix2, H. Botella2, G. Yang1, O. Ouerfelli1, C. Nathan2, S. Ehrt2
1
Organic Synthesis Core Facility, Memorial Sloan-Kettering Cancer Center 2Microbiology and
Immunology, Weill Cornell Medicine, New York, United States
Peptidoglycan (PG), a complex polymer of glycan strands cross-linkeded by short D-amino acidcontaining peptides, is an essential component of the bacterial cell wall. PG biosynthesis is the
target of beta-lactam antimicrobials, some of which have recently been found to be cidal for
Mycobacterium tuberculosis (Mtb), including for non-replicating populations (e.g., Gold et al., J.
Med. Chem. in press). We used supravital fluorescent D-alanine analogs (FDAAs) that incorporate
into peptidoglycan to investigate the spatio-temporal dynamics of PG synthesis in Mtb in axenic
culture and in macrophages. FDAA incorporation into Mtb is predominantly polar, but with striking
variations in polar dominance as a function of cell cycle. Immediately after cytokinesis, FDAAs are
incorporated chiefly at the old pole. In contrast, just before cytokinesis, FDAAs are incorporated
comparably at both poles. These observations suggest that specific enzymes involved in PG
synthesis are localized in functional compartments at the poles and further, that Mtb possesses a
mechanism for maturation of the new pole. Deeper knowledge of the cell and molecular biology of
mycobacterial PG synthesis may help in discovering drugs that disable previously unappreciated
steps in the process.
176
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
155 Mycobacterial Biology & Physiology
20/09/2016
Plasticity of M. tuberculosis NADH dehydrogenases and their role in virulence
C. Vilcheze2, L. Leung3, G. Cook1, W.R. Jacobs Jr2
1
Dept of Microbiology and Immunology, University of Otago, Dunedin, New Zealand 2Microbiology
and Immunology, Howard Hughes Medical Institute at AECOM, Bronx, New York 3Grayboxbiology,
New York, United States
The NADH dehydrogenases, which oxidize the electron donor NADH into NAD+, are the first
enzymes in the electron transport chain. Mycobacterium tuberculosis possesses two types of NADH
dehydrogenases: the proton-pumping NdhI, a 15704 bp operon composed of 14 subunits (NuoA-N,
Rv3145 to Rv3158), and the non-proton pumping, single polypeptide NdhII, which is represented by
two enzymes, Ndh (Rv1854c) and NdhA (Rv0392c). To determine the role of each NADH
dehydrogenase in M. tuberculosis, we deleted them and studied their in vitro and in vivo
phenotypes. The nuoA-N gene cluster and ndhA have been shown to be non-essential genes while
ndh is considered essential. Surprisingly, we were able to obtain a ndh deletion mutant in both M.
tuberculosis H37Rv and CDC1551, along with the nuoA-N and ndhA deletion mutants. We also
constructed the double ndh nuoA-N knockout and ndhAnuoA-N knockout but were unable to obtain
the ∆ndhndhA double knockout suggesting that M. tuberculosis requires at least one NADH
dehydrogenase type II enzyme. In these knockouts, we determined the following in vitro
phenotypes: level of NADH dehydrogenase activity, NADH/NAD+ ratios, expression level of nuoA-N,
ndh, and ndhA, growth under aerobic and hypoxic conditions, and susceptibility to tuberculosis
drugs and NADH dehydrogenase inhibitors. We also measured their growth in cultured
macrophages as well as their growth and survival in immunocompromised and immunocompetent
mice. The following observations were made. The nuoA-N, ndhA and ndhAnuoA-N deletion mutants
showed no growth defect either in vitro or in vivo. In contrast, a longer lag phase was observed in
the ndh and the ndhnuoA-N knockouts in vitro cultures. Furthermore, mice infected with the ndh
knockout survived longer despite granulomatous inflammation and high number of lymphocytes
observed in the lungs. Interestingly, although, the nuoA-N knockout had no phenotype in vivo, the
ndhnuoA-N double knockout was the most severely attenuated strain in mice. These experiments
allow us to conclude that Ndh is the main NADH dehydrogenase of M. tuberculosis and that
compounds that could target both type I and type II NADH dehydrogenases could be good
candidates for tuberculosis drug development.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
177
156 Mycobacterial Biology & Physiology
20/09/2016
Recruitment and regulation of the mycobacterial mutasome
M. Reiche2, Z. Martin2, D. Lang1, N. Dhar3, J. Mckinney3, V. Mizrahi2, D. Warner2
1
Confocal and Light Microscope Imaging Facility, Dept. of Human Biology 2Dept. of Pathology and
IDM, University of Cape Town, Cape Town, South Africa 3Global Health Institute, Swiss Federal
Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
Previous work in our laboratory identified a DNA damage-inducible pathway that is responsible for
adaptive mutagenesis in Mycobacterium tuberculosis (Mtb), including the development of drug
resistance during in vivo infection. In subsequent genetic studies, we have subsequently
established that this mycobacterial “mutasome” comprises a minimum of three essential
components: a dnaE2-encoded DNA Polymerase IIIα subunit, which potentially serves as an errorprone translesion polymerase, and imuB- and imuA’-encoded accessory factors, whose roles within
the mutasome remain cryptic. In order to gain molecular insight into mutasome function, we
investigated the recruitment dynamics and sub-cellular localization of ImuA’ and ImuB following
exposure of bacilli to genotoxic stress. To this end, we constructed a panel of M. smegmatis (Msm)
reporter mutants encoding fluorescently tagged mutasome proteins, and confirmed the functionality
of the recombinants in DNA damage survival and induced mutagenesis assays. Fluorescence
profiles were visualized and quantified utilizing both standard confocal and microfluidic imaging
techniques, and population-wide expression characteristics assessed by flow cytometry. Our results
reveal differential recruitment and localization of ImuA’ and ImuB in bacilli exposed to genotoxic
stress: ImuA’ diffuses rapidly throughout the mycobacterial cell immediately post exposure to DNA
damage, whereas ImuB forms distinct foci that co-localize with the dnaN-encoded β processivity
factor. In addition, multiple ImuB-β foci are observed at discrete intracellular locations throughout
the Msm filaments which form following arrest of cell-division in DNA-damaged cells. These
observations reinforce a mutasome model in which ImuB functions as adaptor protein through its
interaction with the β clamp and, simultaneously, provide novel insight into the potential for
multiploid filaments to accelerate emergence of drug resistance during chronic exposure to sublethal genotoxic stress. Ongoing work is directed towards establishing the temporal order of
recruitment of ImuB and β to stalled replication forks, as well as elucidating the function of ImuA’
within the mutasome. Notably, the approach adopted here confirms the utility of combining
fluorescence imaging with functional genetics to elucidate the mechanisms regulating expression
and activity of a major error-prone damage tolerance pathway in a human pathogen that is
increasingly associated with drug resistance.
178
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
157 Mycobacterial Biology & Physiology
20/09/2016
Extremely Drug Resistant Small-Cell Survival Morphotype in Mycobacteria
M.L. Wu4, M. Gengenbacher6, C.L. Chan4, J.C.S. Chung3, S.L. Chen3-5, H.J. Mollenkopf1,
S.H. Kaufmann2, T. Dick4
1
Core Facility Microarray/Genomics 2Department of Immunology, Max Planck Institute for Infection
Biology, Berlin, Germany 3Infectious Diseases, Genome Institute of Singapore 4Antibacterial Drug
Discovery Laboratoy, Department of Microbiology and Immunology 5Department of Medicine,
Division of Infectious Diseases 6Tuberculosis Research Laboratory, Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,
Singapore
Mycobacteria, generally believed to be non-sporulating, are well known to survive shock starvation
in saline for extended periods of time in a non-replicating state without any apparent morphological
changes. Here we uncover that mycobacteria can undergo cellular differentiation by exposing
Mycobacterium smegmatis to mild starvation conditions. Traces of various carbon sources in saline
triggered the development of a novel small resting cell (SMRC) morphotype. Development of
SMRCs could also be observed for other mycobacteria, suggesting evolutionary conservation of this
differentiation pathway. Fluorescence microscopic analyses showed that development of SMRCs
progresses via septated multi-nucleoided cell intermediates, which divide to generate mononucleoided SMRCs. Intriguingly, saline shock-starved large resting cells (LARCs), which did not
show cell size or surface changes when observed by scanning electron microscopy, remodeled
their internal structure to septated multi-nucleoided cells, similar to the intermediates seen during
SMRC development. Comparative transcriptome analyses of SMRC and LARC development
showed large consistency with our cellular observations. These results suggest that mycobacteria
harbor a starvation-induced differentiation program that differs from the canonical cell division cycle,
in which at first septated multi-nucleoided cells are generated. Under zero-nutrient conditions
bacteria terminate development at this stage as LARCs. In the presence of traces of a carbon
source, these multi-nucleoided cells continue differentiation into mono-nucleoided SMRCs. Both
SMRCs and LARCs exhibited extreme antibiotic tolerance. SMRCs showed increased long-term
starvation survival, which was associated with the presence of lipid inclusion bodies. Interestingly,
the stringent response regulator relA appeared to play the role of cellular differentiation other than a
starvation survival factor in this process.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
179
158 Mycobacterial Biology & Physiology
20/09/2016
Mycobacterium komossense: A surrogate organism for screening new antibiotic against
tuberculosis and for studying the emergence of antibiotic resistance mutation
H. Xiao, K. Oravcova, M. Scott, H. Stacey, R. Hammond, S. Gillespie, A. Gupta
School of Medicine, University of St.Andrews, St.andrews, United Kingdom
Antibiotic resistance of tuberculosis is a significant cause of morbidity and mortality worldwide.
However, the long generation time and pathogenicity of Mycobacterium tuberculosis have been
obstacles for researches investigating mechanisms of resistance in M. tuberculosis. This study
aimed at exploring whether, Mycobacterium komossense, a non-pathogenic fast growing
mycobacterium, can serve as a surrogate to study antibiotic resistance in M. tuberculosis and have
the potential to be a drug screening tool.
MIC test against some first and second-line anti-tuberculosis drugs found that M.Komossense share
similar level of susceptibility as susceptible M. tuberculosis. The antibiotics include Rifampicin,
ciprofloxacin, isoniazid, ethambutol and bedaquiline. Mutants that are resistant to these antibiotics
were subsequently generated by fluctuation assay and validated by exposing the microorganism to
4×MIC of the antibiotics. Fitness costs associated with the development of rifampicin resistance
were investigated by competition assay. Through DNA sequencing and MIC measurement,
characterization and degree of resistance are mapped to specific mutations.
We found that M. komossense and M. tuberculosis resistant strains shared similar mutations in the
RRDR of rpoB. Two commonly reported RIFr mutants in M. tuberculosis, S531L and H5265Y, were
observed in M. komossense. Results of relative fitness of three isolates with S531L mutation and
one isolate with H526Y mutation suggest that resistance mutations are most commonly associated
with a fitness cost; the degree of which is mutation specific. The fitness deficit was similar with to
that previously reported in M. tuberculosis. These data suggest that M. komossense can be a
suitable model to study the evolution of resistance in M. tuberculosis.
Due to the lack of published information of M.komossense’s genome sequence, whole genome
sequencing of M.komossense and annotation of the sequence is going to be carried out to assist us
to characterize mutants against other antibiotics more accurately and effectively.
180
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
159 Mycobacterial Biology & Physiology
20/09/2016
Caseinolytic protease gene regulator (clgR) is a substrate for the mycobacterial Clp
protease
Y. Yamada1, T. Dick2
1
Department of Medicine 2Department of Microbiology and Immunology, National University of
Singapore, Singapore, Singapore
Recent discoveries of anti-mycobacterial compounds which target the caseinolytic protease (Clp)
demonstrated that Clp inhibition is one of the promising approaches to battle drug-resistant
tuberculosis. To further explore potential drug targets associated with mycobacterial Clp protease,
we introduced a promoter-reporter library into M. bovis BCG Pasteur (BCG) and measured the
effect of Clp inhibition. From a sub-set for the Clp-related gene promoters we found that exposure to
the Clp inhibitor Bortezomib (BZ) activated the genes under control of transcriptional activator ClgR
(such as ClpP1, Acr2 and ClgR) in a dose-dependent manner. Via ClgR-mCherry fusion proteins,
we showed that ClgR is a substrate of Clp and that the C-terminal APVVSLAVA is required for
degradation. Taken together, our findings revealed that Clp inhibition activated the ClgR signaling
pathway by inhibiting degradation of ClgR and caused ClgR dysregulation.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
181
160 Mycobacterial Biology & Physiology
20/09/2016
Phenotypic and genotypic analyses of pyrazinamide resistance in Mycobacterium
tuberculosis clinical isolates
M.M.K. Yee1-2, P. Gopal1, R.T.H. Ong2, Y.Y. Teo2, M. Gengenbacher1, T. Dick1
1
Department of Microbiology and Immunology, Yong Loo Lin School of Medicine 2Saw Swee Hock
School of Public Health, National University of Singapore, Singapore, Singapour
Pyrazinamide (PZA) was discovered in the 1950s and has since been playing an essential role as a
key treatment-shortening drug in tuberculosis (TB) chemotherapy. Yet, the mechanism of
action/resistance of PZA (a pro-drug) and its active form, pyrazinoic acid (POA) remains poorly
understood. This translates into challenges in accurately diagnosing PZA resistance in the clinical
setting. Phenotypic methods, the gold standard in diagnosing TB drug resistance, are limited for
PZA because of the low potency of the drug in vitro and the uncertain predictive value of the current
growth assays. Molecular methods (detecting mutations in pncA gene as PncA is involved in
conversion of PZA into POA) lack sensitivity as some clinical isolates are shown to be PZA resistant
with wildtype pncA, suggesting alternative mechanism(s) of PZA/POA resistance. In order to
address these shortcomings in current diagnostic methods, we study the correlation between
various growth / growth inhibition assays and genetic polymorphisms of PZA resistant and
susceptible Mycobacterium tuberculosis clinical isolates. The various growth / growth inhibition
assays include classical broth dilution method vs. BACTEC MGIT 960. The broth dilution assays
include media with different pH. Furthermore, MICs are determined for PZA and POA. First results
of our ongoing analyses will be discussed.
182
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
161 Mycobacterial Biology & Physiology
20/09/2016
Mycobacterial rifampicin exposure leads to a semi-heritable accumulation of RNA
polymerase that is correlated with a survival programme
J.H. Zhu1-2, M.M. Pan2, E.H. Rego3, B. Javid2
1
PKU-Tsinghua-NIBS Graduate Program, School of Life Sciences, Peking University 2Collaborative
Innovation Centre for the Diagnosis and Treatment of Infectious Diseases, Tsinghua University,
Beijing, China 3Department of Immunology and Infectious Disease, Harvard T.H. Chan School of
Public Health, Boston, United States
Rifampicin (RIF) is a first-line anti-tuberculous antibiotic and still the most important antimicrobial in
the fight against tuberculosis. Unlike most anti-tuberculous agents, which kill in a dose-independent
manner, rifampicin-mediated killing is strongly dose-dependent, both in vitro an in vivo. This
suggests that there is a broad susceptibility landscape to RIF in the mycobacterial population at the
single-cell level. Here, we show that upon RIF exposure a significant subpopulation of
mycobacterial cells continue to grow. The proportion of ‘growers’ (phenotypic resistors) varies
inversely with rifampicin concentration, but is still easily detectable at 20XMIC. Using time-lapse
microscopy, we find that there is a strong correlation between survivors and their daughters,
suggesting a degree of semi-heritability. Rifampicin exposure leads to a binary decision tree for
mycobacterial individuals: death or survival. Survivors in each case strongly upregulate expression
of the RNA polymerase beta subunit (RpoB) through a specific transcriptional programme shortly
after encountering RIF, which allows further survival and growth in the context of ongoing drug
exposure. Importantly, survivors and their daughters exhibit a greater degree of rifampicin-specific
phenotypic resistance over time. This was mirrored in (drug-sensitive) M. tuberculosis isolated from
sputum from patients started on standard therapy: over time, the proportion of mycobacteria
surviving drug-treatment was progressively more phenotypically resistant to rifampicin. Our findings
have important implications for understanding tolerance and phenotypic resistance to rifampicin,
and the reason why prolonged antibiotic therapy is required for tuberculosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
183
162 Transport systems & Secretion in Mycobacteria
20/09/2016
Understanding the role of Rv3921c, a preprotein translocase, in Mycobacterium
tuberculosis
P. Thakur1-2, M.Z. Abdin2, N. Agarwal1
1
Translational Health Science and Technology Institute, Faridabad 2Jamia Hamdard, New Delhi,
India
Transport of proteins to cell surface or extracellular environment is accomplished by specialized
protein translocases, which regulate multiple physiological activities along with secretion of
virulence factors or effector molecules in the pathogenic organisms. In human pathogen
Mycobacterium tuberculosis (Mtb) three major protein export pathways are known: the general
secretion (Sec), the twin-arginine transport (TAT) and the ESX pathway. Recent studies have
shown that in several bacteria the Sec and TAT pathways involve a preprotein translocase known
as YidC for membrane insertion of the transported protein, whereas in mitochondria and
chloroplasts insertion of membrane proteins is solely accomplished by YidC orthologues, Oxa and
Alb proteins, respectively.
Analysis of mycobacterial genome sequences reveals the conserved occurrence of yidC
homologues, which is encoded by a gene Rv3921c in Mtb. Importantly, not only the yidC but the
entire locus, surrounded by cluster of genes involved in protein synthesis and membrane biogenesis
is conserved in different mycobacterial species, thus suggesting an essential role of YidC in
mycobacteria. In an earlier study we observed that yidC is essential for in vitro growth of Mtb,
however, an exact function of this gene in the pathophysiology of Mtb remains to characterize.
Here, we show that YidC is expressed in both fast-growing M. smegmatis and slow-growing Mtb at
all growth stages, and a controlled expression of this protein is absolutely required for maintaining
the optimal in vitro growth of Mtb. Interestingly, expression of YidC is modulated by cell envelope
stress agents such as detergent and heat shock. Further, we observed that transient
overexpression of YidC in Mtb alters membrane integrity and causes upregulation of many genes
including SDS-regulons such as sigE and sigB that encode extracytoplasmic function sigma factors.
Taken together these results suggest that YidC plays a pivotal role in biogenesis of mycobacterial
cell envelope, which can be explored as novel anti-TB drug target.
184
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
163 Transport systems & Secretion in Mycobacteria
20/09/2016
The ESX-5 System of Pathogenic Mycobacteria Is Involved In Capsule Integrity and
Membrane Rupture Through Its Substrate PPE10
L. Ates2-5, A. Van Der Woude4-5, J. Bestebroer5, G. Van Stempvoort4, R. Musters7, J. GarciaVallejo6, D. Picavet3, R. Van De Weerd5, M. Maletta1, C. Kuijl5, N. Van Der Wel3, W. Bitter5-4
1
Eyen SE, Prague, Czech Republic 2Pathogénomique Mycobactérienne Intégrée, Institut Pasteur,
Paris, France 3Department of Cell Biology and Histology, Academic Medical Center, University of
Amsterdam 4Department of Molecular Microbiology, VU University 5Department of Medical
Microbiology and Infection Prevention 6Department of Molecular Cell Biology and
Immunology 7Department of Physiology and Cardiology, VU University Medical Center,
Amsterdam, The Netherlands
Mycobacteria produce a capsule layer, which consists of glycan-like polysaccharides and a number
of specific proteins. In this study, we show that, in slow-growing mycobacteria, the type VII secretion
system ESX-5 plays a major role in the integrity and stability of the capsule. We have identified
PPE10 as the ESX-5 substrate responsible for this effect. Mutants in esx-5 and ppe10 both have
impaired capsule integrity as well as reduced surface hydrophobicity, as well as reduced amounts of
surface localized proteins and glycolipids. Since capsular proteins secreted by the ESX-1 system
are important virulence factors, we tested the effect of the capsular defect on virulence
mechanisms. Both esx-5 and ppe10 mutants of Mycobacterium marinum were shown to be
impaired in ESX-1-dependent hemolysis and rupture of the phagosomal membrane. The observed
defects in capsular integrity resulted in a reduced recruitment of ubiquitin in cellular infections and
intermediate attenuation in zebrafish embryos, confirming the importance of capsular integrity in the
early stages of infection. These results provide a pivotal role for PPE10, a substrate of ESX-5, in the
capsular integrity of pathogenic mycobacteria. These findings open up new roads for research on
the mycobacterial capsule and its role in virulence and immune modulation.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
185
164 Transport systems & Secretion in Mycobacteria
20/09/2016
Functional interaction between Mycobacterium tuberculosis RipA, an invasion associated
secretory protein, and MoxR1
M. Bhuwan2, N. Arora2, A. Sharma1, M. Khubaib2, S. Pandey2, T.K. Chaudhari1, S.E. Hasnain1,
N.Z. Ehtesham2
1
Kusuma school of Biological Sciences, IIT Delhi, Delhi 2Inflammation Biology and Cell signaling,
National Institute of Pathology, New Delhi, India
Mycobacterium tuberculosis (M.tb) is a leading cause of death worldwide. The Tat (twin-arginine
translocation) protein secretion system is present at the cytoplasmic membrane of mycobacterium
and is known to transport folded proteins and is reported to be essential for many of the important
bacterial processes that include cell wall biosynthesis. RipA, a secretion and invasion protein of
M.tb has endopeptidase activity. The MoxR1 protein belongs to ATPases family and is associated
with various cellular activities. RipA and MoxR1 were expressed and purified to homogeneity from
E.coli. Recombinant MoxR1 protein was characterized in terms of its biophysical feature. Protein
aggregation assay of recombinant MoxR1 at higher temperature showed resistance to aggregation.
Furthermore, recombinant MoxR1 was able to prevent the aggregation of Maltodextrin glucosidase
(MalZ) protein at higher temperature which is enhanced in the presence of ATP suggesting its role
as a chaperone. Role of MoxR1 was also demonstrated in in vivo folding of RipA in E. coli cells. Insilico analyses pointed to a protein-protein interaction between M.tb RipA and MoxR1 as well as
RipA and Mce2B (mammalian cell entry protein). Furthermore, using bimolecular fluorescence
complementation (BiFC) assay the interaction of these proteins was confirmed in HEK293T using
confocal microscopy. It has been shown earlier that recruitment of the autophagy protein to sites of
bacterial entry leads to lysosomal degradation of the invading bacteria. Our bioinformatics analysis
of RipA protein using eukaryotic linear motif resources (www.elm.eu.org) showed the presence of
canonical LIR motif that binds to Atg8/LC3 protein family members to mediate processes involved in
autophagy. The conserved sequence for this “SFAGV” was found at 390394 amino acid in RipA
protein. Overall our study demonstrates M.tb MoxR1 as novel molecular chaperone involved in
maturation or refolding of specific protein complexes with likely role in pathogenesis.
186
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
165 Transport systems & Secretion in Mycobacteria
20/09/2016
Exploiting the reconstituted ESX-5 system to understand the role of substrates in system
functioning
C. Bunduc5, K. Beckham4, L. Ciccarelli2-1-3, A. Parret4, M. Wilmanns4, T. Marlovits1-2-3, W. Bitter5-6,
E. Houben5
1
Institute of Molecular Biotechnology GmbH 2Research Institute of Molecular Pathology, Vienna,
Austria 3Centre of Structural Systems Biology, University Medical Center Eppendorf 4European
Molecular Biology Laboratory, Hamburg, Germany 5Molecular Microbiology, Vrije Universiteit,
Amsterdam 6VU University Medical Center, Amsterdam, The Netherlands
Pathogenic mycobacteria, including Mycobacterium tuberculosis, use type VII secretion (T7S)
systems to secrete important virulence factors across their distinctive cell envelope. These bacteria
have up to five of such systems, termed ESX-1 to ESX-5. T7S does not resemble known
specialised secretion pathways in other bacteria, reflecting the unique properties of the
mycobacterial cell envelope. Although some of these secretion systems are crucial for
mycobacterial growth or virulence, the actual mechanism of membrane transport is still largely
unknown. As a first step in this analysis, we have previously isolated the ESX-5 membrane complex
from Mycobacterium marinum and determined that this macromolecular machinery has a size of 1.5
MDa and consists of four membrane components, i.e. EccBCDE (Houben et al., Mol Microbiol
2012). The aim of this study is to elucidate and further characterize the T7S membrane channel.
To more easily analyse the functionality of this membrane complex in secretion, we reconstituted
the ESX-5 system in the avirulent and fast-growing mycobacterial species Mycobacterium
smegmatis that lacks ESX-5. The reconstituted system proved to be active and was efficiently
secreting EsxN and specific PE/PPE substrates. Importantly, we noticed that the different
components were relatively overexpressed and the complex was properly assembled in the
membrane. This reconstitution therefore not only opened up the possibility to more easily analyse
the role of specific Esx and PE/PPE proteins in the secretion process, but also to check possible
interactions between substrates and components of the translocation channel. In addition, we
purified this ESX-5 membrane complex, determined the molecular composition and examined the
structure through negative stain electron microscopy (see also abstract of Beckham et al.). In
conclusion, we functionally reconstituted and overproduced the ESX-5 system in M. smegmatis,
which opened up the possibility to efficiently and more thoroughly analyse the functioning and
structure of this secretion system.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
187
166 Transport systems & Secretion in Mycobacteria
20/09/2016
Outer membrane localization of CpnT and toxin translocation is dependent on type VII
secretion in Mycobacterium tuberculosis
K. Doornbos1, O. Danilchanka2, J. Sun1, M. Niederweis1
1
Microbiology, University of Alabama at Birmingham, Birmingham 2Epiva Biosciences, Cambridge,
United States
Mycobacterium tuberculosis (Mtb) secretes a NAD-glycohydrolase which causes necrotic death in
host cells. This tuberculosis necrotizing toxin (TNT) constitutes the C-terminal domain of the outer
membrane protein (OMP) CpnT and is a major cytotoxicity factor of Mtb. The absence of classical
export signals within CpnT raised the question of how CpnT is integrated into mycobacterial outer
membranes and how TNT is secreted. In this study we demonstrate that localization of CpnT
depends on an N-terminal Esx motif which target proteins to the type VII secretion systems of Mtb,
the Esx systems. A separate, C-terminal Esx motif is required for TNT translocation across the outer
membrane, but not for the channel activity of CpnT in the outer membrane. Thus, CpnT is the first
OMP whose assembly is controlled by type VII secretion. Esx motifs similar to those in CpnT are
found in 29 other Mtb proteins including the virulence factor LipY, indicating that this novel OMP
assembly mechanism is widespread. Taken together, this study not only advances our mechanistic
understanding of toxin secretion by Mtb, but also reveals a previously unknown function of Esx
systems and has important implications for OMP assembly and cell permeability of this major
human pathogen.
188
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
167 Transport systems & Secretion in Mycobacteria
20/09/2016
Biochemical characterization of the mycobacterial MFS transporter Rv1410 and
investigation of its role in the correct assembly of the cell wall
Michael. Hohl, F. Arnold, A. Krügel, Markus. Seeger
University of Zurich, Institute of Medical Microbiology, Zürich, Switzerland
Mycobacterium tuberculosis is surrounded by a complex cell wall that naturally protects the bacilli
against noxious substances. The mycobacterial operon rv1410-rv1411 encoding a major facilitator
superfamily (MFS) transporter (Rv1410) and a lipoprotein (Rv1411, LprG) is strongly conserved
among many mycobacterial species and is critical for virulence in M. tuberculosis. While initial
studies on Rv1410 proposed a role in drug efflux, more recent studies showed a connection of the
MSF transporter with LprG to play an important role in cell wall assembly. Recent work focused on
the lipoprotein alone, showing that cell wall components such as lipoarabinomannans (LAMs) bind
to LprG, which transports them to the cell surface. Metabolomic studies using M. tuberculosis
rv1410-lprG knockout cells showed the accumulation of triacylglycerides (TAGs) in the cytoplasm,
demonstrating a role of this locus in TAG transport. Crystal structures of LprG bound to a LAM
precursor and a TAG molecule have been solved. In contrast, biochemical data on the MFS
transporter Rv1410 are scarce. We constructed gene deletions of the corresponding operon as well
as of the MFS gene alone in M. smegmatis leading to attenuated growth in the presence of
ethidium. Introduction of the operons from M. tuberculosis, M. marinum and M. abscessus fully
recovered the growth deficit. Further, we were able to complement the MFS gene deletion with
rv1410 of M. tuberculosis and identified an aspartate to asparagine (D-to-N) mutation, which fully
inhibits the transport activity of Rv1410. To study the molecular basis of the interplay between
Rv1410 and LprG we purified these two proteins upon heterologous expression in E. coli as wellfolded, monomeric proteins. Size exclusion chromatography (SEC) and surface plasmon resonance
(SPR) analyses, however, did not reveal a physical interaction between these proteins, suggesting
a functional interaction between these two transport proteins for the export of LAMs and TAGs.
Through a combination of X-ray crystallography, biochemical in vitro assays and functional in vivo
experiments we aim to reveal the molecular function of the MFS transporter Rv1410 and thereby
may help to design novel TB-drugs.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
189
168 Transport systems & Secretion in Mycobacteria
20/09/2016
Role of specific chaperones in determining system specificity in type VII secretion systems
T. Phan1, R. Ummels2, N. Korotkova3, K. Korotkov3, W. Bitter1-2, E. Houben1
1
Vrije Universiteit Amsterdam 2VU University Medical Center, Amsterdam, The
Netherlands 3University of Kentucky, Kentucky, United States
Type VII secretion (T7S) is used by mycobacteria to export protein effectors across their highly
unusual cell envelope. There are five paralogous T7S systems in Mycobacterium tuberculosis,
named ESX-1 to ESX-5, each having their own role in viability and/or virulence. The mycobacteriaspecific PE/PPE proteins are one group of proteins that are secreted via these systems, of which at
least some are exported as heterodimers. We have previously shown that the cytosolic chaperone
EspG specifically interacts with cognate PE/PPE dimers and is required for their successful export1.
To further investigate the substrate specific binding of the EspG chaperone we have obtained the
crystal structure of the ESX-5 chaperone EspG5 with the ESX-5 substrate pair PE25/PPE412. The
structure reveals that the chaperone exclusively interacts with the elongated end of PPE41, a region
containing several hydrophobic residues that are conserved in PPE substrates of different secretion
systems. Introducing point mutations in these conserved residues of multiple ESX-5 PPE substrates
abolished the interaction with the cognate EspG chaperone. Disrupting the interaction with EspG
also blocked their secretion by Mycobacterium marinum, indicating that chaperone binding is an
important prerequisite for protein secretion. In this study, we show that also for an ESX-1 dependent
PPE protein these conserved residues are important for secretion. Subsequently, we exchanged the
complete EspG-binding domain between an ESX-1 and an ESX-5 PPE protein and tested their
route of export using different M. marinum ESX-1 and ESX-5 mutant strains. These experiments
revealed that the ESX-1 substrate containing an EspG5 binding domain is secreted independently
on its original ESX-1 system, but is now entirely dependent on the ESX-5 system for secretion.
These results suggest that the EspG chaperone is the primary factor for determining system
specificity in M. marinum. Rerouting of specific ESX-1 substrates via ESX-5 could be used to
understand the role of individual ESX-1 substrates in virulence and to improve the current live
vaccine strain BCG, that lacks the ESX-1 system, through modulation of its antigen repertoire.
1. Daleke, M.H., et al. (2012) J Biol Chem 287, 31939-3194710.
2. Korotkova, N., et al. (2014) Mol Microbiol 94, 367-382.
190
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
169 Transport systems & Secretion in Mycobacteria
20/09/2016
Type VII Secretion Pathway Involved in Mycobacterium abscessus Intracellular Survival
L. Laencina1, V. Le Moigne1, V. Dubois1, C. Dupont2, N. Van Wyk2, E. Rubin3, L. Kremer2,
J.L. Herrmann1, F. Girard-Misguich1
1
U1173 INSERM Infection, Inflammation, Université de Versailles Saint-Quentin-en-Yvelines,
Montigny-Le-Bretonneux 2Centre d’études d’agents Pathogènes et Biotechnologies pour la Santé,
FRE3689 CNRS / UMR Mycobacterial Pathogenesis and Novel Therapeutic Targets, Montpellier,
France 3Department of Microbiology and Immunology, Harvard Medical School, Boston, United
States
Mycobacterium abscessus represents, with Mycobacterium avium, the major mycobacterial
pathogen responsible for pulmonary infections in patients with cystic fibrosis. The M. abscessus
complex comprises three subspecies: abscessus, massiliense and bolletii. M. abscessus behaves
as an intracellular pathogen, able to grow and survive in macrophages, survival usually not
observed for rapid growing mycobacteria (RGM). Genome analysis of M. abscessus showed that
this mycobacterium is endowed with several virulence genes, not found in saprophytic RGM,
allowing its interaction with free-living amoeba in the environment, and potentially explaining its
survival and growth in human macrophages. However, very little is known about the genes helping
M. abscessus to grow and survive in antigen presenting cells such as macrophages or in
environmental protozoa such as amoeba.
A transposon mutant library of a clinical strain of M. abscessus subsp. massiliense was screened
based on its ability to persist in amoeba and macrophages. One hundred and thirty-six out of six
thousand individually screened mutants exhibited a reduced ability to survive in macrophages
and/or amoeba. Among them, sixteen were mutated in genes belonging to M. abscessus esx-4
gene locus, belonging to the type VII secretion system. One gene, eccB, was chosen to create a
deletion mutant by replacing eccB by a zeocin resistance cassette in both the smooth and rough
variants of the CIP104536T M. abscessus. These mutants are used to confirm the attenuated
phenotype linked to a defective ESX-4 secretion system, and to complement this defect by
introducing a functional copy of the gene.
The ESX-4 locus is conserved in all mycobacteria and is considered as the ancestral gene cluster of
the five ESX subtypes. Overall, our results indicate, for the first time, that ESX-4 participates in
growth and survival of M. abscessus in macrophages and amoeba. Experiments are currently on
going to address whether ESX-4 in M. abscessus shares a biological role similar to the one
described for ESX-1 in M. tuberculosis to allow the bacteria to communicate with the cytosol in
infected macrophages.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
191
170 Transport systems & Secretion in Mycobacteria
20/09/2016
Colorimetric assay for detecting ESX-3 secretion system disruption in mycobacteria
M. Maerk1, T. Strand1, S. Bandyopadhyay2, T. Flo1, M. Steigedal1
1
Dept. of Cancer Research and Molecular Medicine, Centre of Molecular Inflammation
Research 2Dept. of Chemical Engineering, Norwegian University of Science and Technology,
Trondheim, Norway
Tuberculosis (TB) remains one of the major infectious diseases causing morbidity and death in large
parts of the world. Various classes of drug-resistant TB strains have emerged over the recent years,
which have prompted several initiatives to find new anti-TB drugs. M. tuberculosis, the causative
agent of TB, is a persistent bacterium with an unusual and highly impermeable cell envelope, which
is part of the reason it is difficult to develop new therapeutic strategies for treating TB infections.
Biogenesis and functioning of this unusual cell envelope is however also the target of many new
and existing TB drugs. In our current project we aim to identify compounds that target the
mycobacterial type VII secretion systems (T7SS), which are located in the cell envelope and secrete
effector proteins. Mycobacteria can reside and multiply within infected host cells, and the T7SS
secrete proteins across the cell envelope to the bacterial cell surface or into the host environment.
Five mycobacterial T7SS have been described; ESX-1 to -5. Three of these (ESX-1, -3 and -5) are
essential for virulence or viability. We are developing and evaluating a gold nanoparticle-based
colorimetric assay that can detect ESX-3 substrate secretion, and will subsequently use this to carry
out high-throughput screening (HTS) of chemical libraries in order to identify potential drug
candidates that target this system.
192
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
171 Transport systems & Secretion in Mycobacteria
20/09/2016
Plasmids expressing tagged ESAT-6 and CFP-10 as a tool for immunostaining of ESAT-6
and CFP-10 inside human macrophages
J. Raffetseder, V. Loitto, M. Lerm
Linköping University, Linköping, Sweden
With the global increase of multidrug-resistant strains of Mycobacterium tuberculosis (Mtb), new
treatments for tuberculosis are urgently needed, with one of the drug development strategies
evolving around Mtb virulence blockers. ESAT-6 is a major virulence factor of Mtb and has been
shown to be involved in many processes in the interaction between Mtb and host cells, such as
induction of necrotic host cell death, dissemination to other cells and granuloma formation. It has
been reported to have membrane-lysing activity, and mycobacteria deficient in ESAT-6 or the
ESAT-6 secretion system ESX-1 are unable to translocate from phagosomal compartments into the
cytosol. We wanted to investigate the role and the secretion kinetics of ESAT-6 in an established
infection model of human monocyte-derived macrophages, both under conditions when Mtb actively
replicates inside the macrophage leading to host cell death as well as during Mtb growth restriction
with uncompromised macrophage viability. The aim was to better understand how macrophages
restrict Mtb growth and under which conditions the secretion of the ESAT-6/CFP-10 heterodimer
can be inhibited. This information could be useful for the development of ESX-1 blockers. To
facilitate immunostaining of secreted ESAT-6 and CFP-10 after macrophage infection, we
transformed Mtb H37Rv strains with plasmids carrying gene sequences for fusion proteins of ESAT6 or CFP-10 and antigenic peptide tags (c-myc or hemagglutinin). Using Western Blotting as well as
immunofluorescent staining followed by quantification of fluorescence intensity, we found that
tagged ESAT-6 and CFP-10 were expressed and actively secreted during cultivation of Mtb in broth.
In contrary, antibody staining of tagged ESAT-6 or CFP-10 inside infected human monocyte-derived
macrophages resulted in only weak signals just above background which however correlated with
intracellular bacterial growth.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
193
172 Transport systems & Secretion in Mycobacteria
20/09/2016
Cryo-EM Structural characterization of the M. tuberculosis ESX-1 secreted virulence factor
EspB
R. Ravelli, A. Siroy, G. Tria, D. Chen, P. Afanasyev, N. Iakobachvili, C. Berger, K. Notelaers,
R. Bosmans, A. Gisbers, H. Furusho, C. Lopez-Iglesias, P. Peters
M4i-Nanoscopy, Maastricht University, Maastricht, The Netherlands
Virulent mycobacteria infect their host cells through a Trojan Horse strategy: they get ingested by
the normal phagocytosis route but instead of being digested through acidification and maturation of
the phagosome to phagolysosome, these bacteria enter the host’s cell cytosol where they replicate
freely. Controlled death of the host will allow the spreading of the descendants in the infected
organism.
A key step in the infectious life cycle of the virulent mycobacteria is their escape from the
phagosome compartment. This phenomenon is mediated by the release of different proteins
(EsxAB) that will perforate the phagosome membrane. These factors are secreted by the
specialized machinery ESX-1 of the recently discovered Type 7 Secretion System family. The ESX1 machinery is a multi-protein complex thought to span the entire mycobacterial cell envelope. It is
constituted by a core of structural proteins (EccACabBDE1) and the assembly and/or functioning of
the machinery seems to require the recruitment of additional proteins.
Among the different ESX-1 substrates, the 48 kDa protein EspB seems to carry multiple roles: this
secreted protein carries cytolytic activity on its own; it is co-secreted with EsxA (the main virulence
factor) and is required for EsxA secretion; EspB may also play a structural role suggested by its
ability to oligomerize as well as its proteolytic maturation by MycP1 during the translocation process.
Analysis of EspB primary sequence predicts two main structural domains: similar to other ESX
substrates, the N-terminal domain is made of a bundle of α-helices while the C-terminal one seems
unfolded. The structure of the N-terminal domain of EspB was recently solved by X-ray
crystallography and, combined with negative-stain electron microscopy 2D classes, a heptameric
model was computationally generated.
Here, we report structural characterization of EspB using transmission electron microscopy. We
determined the structure of the N-terminal helix bundle, the mature form as well as the pre-protein
EspB at sub-nanometer resolution. Moreover, we were able to identify several distinct oligomeric
states of the proteins that were not previously observed, reflecting a possible role of EspB as a
structural component of ESX-1.
194
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
173 Transport systems & Secretion in Mycobacteria
20/09/2016
Isolation and structural characterization of native Mycobacterium marinum ESX-1 secretion
machinery by cryo-EM
A. Siroy, D. Chen, G. Tria, N. Iakobachvili, C. Berger, K. Notelaers, R. Bosmans, A. Gijsbers,
H. Furusho, C. Lopez-Iglesias, R. Ravelli, P. Peters
M4i-Nanoscopy, Maastricht University, Maastricht, France
Pathogenic mycobacteria utilize a common “Trojan Horse” strategy to infect their host cells, i.e. they
get phagocytosed and should be killed. Instead, these bacteria are able to rupture the phagosome
membrane before its maturation into phagolysosome by secreting different proteins (EsxAB). Set
free in the cytosol, those pathogens can replicate and control the cell death of their host. These
“Phagosomal Escape” depends on the presence of the protein complex ESX-1 of the Type Seven
Secretion System family (T7SS).
This ESX-1 machinery is an elaborate protein complex of which not all the components are known
to date. Five known structural components of the complex (EccABCDE1) are encoded within the
RD-1 genomic region, alongside the substrates EsxAB, EspB and other less characterized ones.
More than a decade after the discovery of ESX-1, successful expression of the recombinant
machinery has yet to be reported and only partial structural characterization of the core components
is available, showing the difficulty of the task at hand.
We present here the first results regarding the investigation by cryo-electron microscopy of the
structure of native ESX-1 complex produced by Mycobacterium marinum, the fish equivalent to the
human pathogen M. tuberculosis. In this regard, we developed a new fluorescent probe that allows
the tracking of the protein complex during its isolation and purification, as well as monitoring its
location in a correlative fluorescent and electron microscopy workflow in cryogenic conditions. The
conditions for the extraction from the cell envelope were screened and the fluorescently-tagged
ESX-1 complex was purified by liquid chromatography. The ESX-1 enriched samples were imaged
by cryo-transmission electron microscopy and the micrographs analyzed using Single Particle
Averaging (SPA) techniques.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
195
174 Transport systems & Secretion in Mycobacteria
20/09/2016
Regulation of ESX-1-mediated Secretion in Mycobacterium tuberculosis
P. Soler, M. Zhang, C. Sala, S. Cole
GHI, Prof. Cole Lab (UPCOL), EPFL, Lausanne, Switzerland
Mycobacterium tuberculosis is an intracellular pathogen that requires a functional ESX-1 system to
secrete virulence factors and successfully spread inside the host. The ESX-1 secretory apparatus is
predicted to be a multi-subunit system encoded by more than twenty genes. Nowadays, the function
of each ESX-1 component and their regulation remain partially unknown. To better understand the
virulence mechanisms employed by M. tuberculosis, we are studying the unknown ESX-1
components and trying to assign a functional role to them. To do so, we have constructed an M.
tuberculosis mycP1-eccE1 double mutant and an M. tuberculosis whiB6 mutant to analyze their
phenotypes in vitro by looking at gene expression, ESX-1 protein secretion and possible interactors
as well as their localization in the cell. We also analyzed ex vivo virulence and cytokine production
upon macrophage infection and will present the findings.
196
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
175 Transport systems & Secretion in Mycobacteria
20/09/2016
Overexpression of a Mycobacterium abscessus efflux pump of the MmpL family confers
resistance to clarithromycin
C. Cortesía3, G. López3, A. Pawlik1, R. Brosch1, H. Takiff3-2
1
Integrated Mycobacterial Pathogenomics 2Unité de Genetique Mycobacterienne, Institut Pasteur,
Paris, France 3CMBC, Lab. de Genética Molecular, Instituto Venezolano de Investigaciones
Científicas, IVIC, Caracas, Venezuela
M. abscessus causes soft tissue infections after surgical and cosmetic procedures and respiratory
infections in cystic fibrosis patients. We found that the presence of MAB_2275 on a plasmid confers
high-level clarithromycin resistance in strains of the M. abscessus family without an inducible
methyltransferase. MAB_2275 encodes an efflux pump belonging to the MmpL family. Similar to
other MmpL genes, there is an adjacent putative transcriptional repressor. Efflux pump inhibitors
decrease the level of resistance of strains both with and without the plasmid based efflux pump.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
197
176 Transport systems & Secretion in Mycobacteria
20/09/2016
MMPL3 is the inner membrane flippase of trehalose monomycolate in mycobacteria
Z. Xu1, S. Chng1-2
1
Chemistry Department 2Singapore Centre on Environmental Life Sciences Engineering, National
University of Singapore, Singapore, Singapore
The mycobacterial cell envelope consists of two membranes, an inner membrane (IM) and a
mycolic acid-rich outer membrane (OM). The OM is an exceptional permeability barrier and is
essential for virulence. As the major lipid component of the OM, mycolic acids are biosynthesized as
trehalose monomycolates (TMMs) in the IM and then transported across the IM, the aqueous
periplasm and finally to the OM, where they become functionalized onto the bacterial cell wall. Few
proteins involved in TMM transport and assembly into the OM have been identified. In this study, we
set out to characterize the function of MmpL3, an essential IM protein belonging to the RND
(resistance, nodulation, and cell division) protein family. MmpL3 has been implicated in TMM
transport in the IM; however, the exact role of this protein is not known. It has also been
hypothesized to be a target of several drug candidates with varied structures, although it is not clear
if each of these compounds directly inhibits MmpL3 function or kill mycobacteria through other
mechanisms. In this work, we used a cell-based assay to demonstrate that MmpL3 is involved in
flipping TMM across the IM in vivo. We first established that newly synthesized mycolic acids
accumulated as TMM in the IM of mycobacterial spheroplasts. In fact, almost all accumulated TMM
molecules were exposed on the surface of these spheroplasts, indicating they resided in the outer
leaflet of the IM. Surface exposure of TMM was partly prevented when MmpL3 was inhibited by
selective drug candidates, which did not also affect the pmf (proton motive force). These results
define MmpL3 as the flippase for TMM in mycobacteria, and establish it as a direct target for a
subset of promising anti-mycobacterial compounds.
198
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
POSTER SESSION 2
Wednesday, September 21
SUMMARY
Biomarkers & Vaccines................................................................ 201
Biochemistry & Chemical biology ................................................ 228
177 Biomarkers & Vaccines
21/09/2016
Exploration of urinary biomarkers for the detection of paediatric tuberculosis
N. Andreas2, L. Tientcheu1, R. Basu Roy2, T. Togun1, M. Gomez-Romero3, N. Harvey3,
B. Jiménez3, E. Holmes3, B. Kampmann2-1
1
Vaccines and Immunity Theme, MRC Unit-The Gambia, Fajara, Gambia 2Centre for International
Child Health 3Section of Computational and Systems Medicine, Imperial College London, London,
United Kingdom
Background and aims: The diagnosis of tuberculosis in children is difficult and less than one third
of suspected cases are microbiologically confirmed [1]. The aim of this project was to analyse
paediatric urine samples, employing a metabonomic strategy, in order to identify novel diagnostic
biomarkers for paediatric tuberculosis. The samples analysed were collected longitudinally from
children diagnosed with TB (cases) or other respiratory infections (controls) in The Gambia.
Methods: 166 urine samples collected at enrolment, two weeks, two months and six months postenrolment from 95 children underwent 1H NMR spectroscopic analysis, in general profiling mode [2].
Spectra collected were normalised [3], aligned [4] and scaled [5]. The subsequent data were
analysed using multivariate modelling techniques, including principal components analysis (PCA)
and Orthogonal partial least squares-discriminant analysis (OPLS) combining probable and
confirmed TB cases.
Results: At enrolment, 9 confirmed, 12 probable and 53 non-TB samples were available for
analysis. Data from these samples were analysed using OPLS, classifying samples using their
clinical diagnosis, Figure 1.
Figure 1: OPLS scores plot of non-TB (blue) versus probable (red) and confirmed TB samples
(green). R2X=0.19, R2Y=0.61, Q2=0.27, n=70.
Samples separated out based on their diagnosis. The identification of metabolites discriminating
between the different diagnoses was investigated. Increased concentrations of a mammalianmicrobial co-metabolite, a metabolite associated with muscle metabolism and an as yet unidentified
metabolite were observed in the non-TB urine samples, whist several further unidentified
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
201
metabolites were increased in probable TB and confirmed TB samples at baseline. TB samples
were also seen to separate out from one another based on time of collection post-enrolment.
Conclusions: This novel work suggests that several host derived metabolites are altered during TB
infection and that the concentrations of these metabolites alters overs the course of infection.
However, to employ this approach as a diagnostic tool substantial development would be required
prior to implementation. Nevertheless, the data provide insights into the alterations in host
metabolism in response to infection with tuberculosis as well as in regard to drug metabolism.
References
1. Togun, T.O., et al., Contribution of Xpert (R) MTB/RIF to the diagnosis of pulmonary
tuberculosis among TB-exposed children in The Gambia. International Journal of Tuberculosis
and Lung Disease, 2015. 19(9): p. 1091-1097.
2. Dona, A.C., et al., Precision high-throughput proton NMR spectroscopy of human urine,
serum, and plasma for large-scale metabolic phenotyping. Anal Chem, 2014. 86(19): p. 988794.
3. Dieterle, F., et al., Probabilistic quotient normalization as robust method to account for dilution
of complex biological mixtures. Application in H-1 NMR metabonomics. Analytical Chemistry,
2006. 78(13): p. 4281-4290.
4. Veselkov, K.A., et al., Recursive Segment-Wise Peak Alignment of Biological H-1 NMR
Spectra for Improved Metabolic Biomarker Recovery. Analytical Chemistry, 2009. 81(1): p. 5666.
5. Eriksson L, J.E., Kettaneh-Wold N, Wold S, Introduction to multi- and megavariate data
analysis using projection methods (PCA & PLS) 1999: Umetrics.
202
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
178 Biomarkers & Vaccines
21/09/2016
Recombinant Attenuated Mycobacterium tuberculosis for Heterologous Expression and
Secretion of DTP Antigens: A Rationale to Construct a Multivalent Vaccine
E. Broset3-2, A.I. Kanno1, L.C. Leite1, C. Martin3-2, J. Gonzalo-Asensio3-2
1
Centro de Biotecnologia, Instituto Butantan, São Paulo, Brazil 2CIBER enfermedades
respiratorias, Instituto de Salud Carlos III, Madrid 3Genetica de micobacterias, Universidad de
zaragoza, Zaragaoza, Spain
Live attenuated BCG vaccine is effective in reducing severe forms of tuberculosis (TB) in children
but confers variable protection against pulmonary TB in adults (1). To overcome this handicap of
BCG, we have developed the live vaccine MTBVAC, based on the attenuation of Mycobacterium
tuberculosis by deletion of the phoP and fadD26 virulence genes (2). MTBVAC is currently in phase
Ib clinical trials in South Africa.(NTC02729571)
Due to its adjuvant capacity, BCG has been traditionally proposed as vector to deliver viral, bacterial
or parasite antigens (3). In this context, we propose the study of MTBVAC as a multivalent
recombinant vaccine against different infectious diseases including diphtheria, tetanus and
whooping cough.
Prior to construct recombinant MTBVAC (rMTBVAC) strains; we studied the possible immune
interference between MTBVAC and the current vaccines against diphtheria, tetanus and whooping
cough (DTP). Actually two formulations of DTP are administered depending on the region: either the
pertussis inactivated toxins (DTaP) or the pertussis whole-cell vaccine (DTwP). In this context, we
tested both DTP formulations in mice previously vaccinated with MTBVAC. Results were compared
with mice independently vaccinated with MTBVAC or DTP. Mice conserved similar immune
response against MTBVAC or DTP in all cases, which proves absence of interference between both
vaccines.
To construct rMTBVAC, genetically inactivated and codon optimized genes for diphtheria
(CRM197), pertussis toxoids (S1), or Fragment C of tetanus toxin (FC) were synthesized. Each DTP
antigen was cloned under the control of a new mycobacteriophage derived promoter (4). To lead
the secretion of these antigens, the signal sequence of Ag85A (a known mycobacterial secreted
protein) was placed in frame before each DTP antigen. Protein expression and secretion of
CRM197, S1 and FC was confirmed by Western blot using antigen-specific antibodies.
Preliminary experiments in mice showed that vaccination with rMTBVAC expressing the S1 toxoid
protects against Bordetella pertussis colonization. On-going experiments aimed to detect anti-DTP
antibodies in mice vaccinated with these rMTBVAC. Taking together, these results provide proof-of
concept that MTBVAC is an efficient antigenic vector for heterologous antigen expression and put
forward rMTBVAC to protect against different infectious diseases.
References:
1. Fine, P.E. (1995). Lancet 346(8986):1339-45.
2. Arbués et al. (2013). Vaccine 31(42):4867-73
3. Bastos et al. (2009). Vaccine 27(47): 6495-503.
4. Kanno et al. (2016). Appl. Environ. Microbiol 82(8): 2240-2246
Funding: Esther Broset was recipient of BES-2012-052937 grant by Ministerio de Economía y
Competitividad de España. This work was funded by BIO2014-5258P and TBVAC2020 projects
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
203
179 Biomarkers & Vaccines
21/09/2016
Differential Diagnosis of Multiplex PCR for Major Mycobacterium species causing Lung
Disease
H. Chae, S.J. Han, S.J. Shin
Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, South Korea
Not only distinguishing between Mycobacterium tuberculosis complex (MTBC) and nontuberculous
mycobacteria (NTM) but also identifying major pathogens causing mycobacterial lung disease at the
species level are medically important. In this study, we developed a multiplex PCR assay using
novel molecular targets that can differentiate the MTB Beijing family, a major member of MTB in
east-Asian countries including Korea, from other MTB lineages (Beijing vs Non-Beijing lineage) and
simultaneously identify four clinically important NTM species consisting of M. avium subsp.
hominissuis (MAH), M. intracellulare (MI), M. abscessus subsp. abscessus (MAB) and M.
abscessus subsp. massilience (MAS). A total of 7 primer sets of multiplex PCR are designed
according to the following purposes; 1) Detection of the genus Mycobacterium by 16s rRNA gene
(Pan-Mycobacterium detection), 2) Differentiation between MTBC and NTM by rv0577, 3)
Differentiation between MTB and other species inside MTBC by the region of difference (RD) 9
region, 4) Specific identification of Beijing family of MTB by mtbk_20680, 5) Identification and
differentiation of among Mycobacterium avium complex by IS1311 and DT1, and 6) Identification
and differentiation between MAB and MAS by mab_3256c. For the accuracy of the multiplex PCR
assay, 37 reference strains (4 MTBC, 4 MTB and 29 NTM) and 83 clinical isolates (5 MTBC, 58
MTB and 20 NTM) were evaluated. Spoligotyping and whole genome sequencing as the standard
assays were further employed to validate the multiplex PCR assay in 58 clinical MTB isolates.
Assay results demonstrated 100% sensitivity and 97.2% specificity for MTBC, respectively. In
addition, multi-locus sequencing analysis of NTM strains revealed 100% sensitivity and 100%
specificity, respectively. Collectively, our multiplex PCR assay is a simple, convenient and reliable
assay for simultaneous identification of the MTB Beijing lineage and major NTM species causing
lung disease in Korea.
204
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
180 Biomarkers & Vaccines
21/09/2016
In depth characterization of a BCG SapM transposon mutant, a potential candidate vaccine
against tuberculosis
N. Festjens, E. Houthuys, K. Vandewalle, E. Plets, D. Vanderschaeghe, K. Borgers, N. Callewaert
MBC, Unit for Medical Biotechnology, VIB - Ghent University, Zwijnaarde, Belgium
M. bovis Bacille Calmette Guérin (BCG), the only licensed vaccine against TB, shows variable
efficacy in protection against adult tuberculosis (TB). Development of a better vaccine is hampered
by the lack of reliable correlates of protection against TB. Earlier, we have described a BCG
transposon mutant in the secreted acid phosphatase SapM, which leads to prolonged survival of
mice infected with M. tb. We have characterized the genome and transcriptome of this sapM::Tn
disruption mutant versus parental BCG Pasteur 1721. Our sequencing data indicate only a few
variations compared to the parental M. bovis BCG Pasteur str 1173P2 reference genome, most of
which are present in both strains. We also show that transcription of sapM is considerably
downregulated and that transcription of sapM’s neighboring genes is directly influenced by the
presence of the transposon. By ELISA and Western blot, we detected a substantial decrease in
SapM protein for the mutant culture supernatant samples versus the WT. Additionally, the enzyme’s
activity was measured by an in vitro phosphatase assay and shown to be significantly reduced.
Additionally, we further analyzed host immune response upon vaccination with BCG sapM::Tn
versus parental BCG. We show now a better control of BCG sapM::Tn growth compared to WT
BCG following vaccination. Previous observations indicated that more effective recruitment of
inflammatory dendritic cells (iDCs) towards the draining lymph nodes might account for superior
protection by the sapM::Tn mutant in Balb/c mice. Studies in C57BL/6J mice now show that the
kinetics of iDC recruitment differ upon BCG sapM::Tn and WT vaccination: iDC recruitment to
lymphoid organs starts earlier when mice are vaccinated with the BCG sapM::Tn strain. Intriguingly,
vaccination with BCG sapM::Tn induced reduced frequencies of IFNγ-producing CD4+ and CD8+ T
cells at early time points post-vaccination. Studies with a sapM::Tn complementation mutant and an
M. bovis BCG SapM knock out demonstrate that the observed phenotypes are caused by the
decrease in sapM expression, and are not due to the polar transcriptional effects of the transposon
on neighboring genes, strengthening the fact that absence of SapM might lead to an improved
priming vaccine based on live Mycobacterium.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
205
181 Biomarkers & Vaccines
21/09/2016
Parenteral Adenoviral Boost Enhances BCG Induced Protection, but not Long Term
Survival in a Murine Model of Bovine TB
C. Garcia-Pelayo3-2, A.K. Daryan3-2, P.R. Webb2, E.E. Wooff2, V.S. Bachy3-2-1, P.J. Hogarth3-2
1
Orbio Laboratoire, BRON, France 2Department of Bacteriology, Animal & Plant Health Agency
(APHA) 3Vaccine Immunology Team, Addlestone, United Kingdom
Boosting BCG using heterologous prime-boost represents a promising strategy for improved
tuberculosis (TB) vaccines, and adenovirus (Ad) delivery is established as an efficacious boosting
vehicle. Although studies demonstrate that intranasal administration of Ad boost to BCG offers
optimal protection, this is not currently possible in cattle. Using Ad vaccine expressing the
mycobacterial antigen TB10.4 (BCG/Ad-TB10.4), we demonstrate, parenteral boost of BCG
immunised mice to induce specific CD8+ IFN-γ producing T cells via synergistic priming of new
epitopes. This induces significant improvement in pulmonary protection against M. bovis over that
provided by BCG when assessed in a standard 4 week challenge model. However, in a stringent,
year-long survival study, BCG/Ad-TB10.4 did not improve outcome over BCG, which we suggest
may be due to the lack of additional memory cells (IL-2+) induced by boosting. These data indicate
BCG-prime/parenteral-Ad-TB10.4-boost to be a promising candidate, but also highlight the need for
further understanding of the mechanisms of T cell priming and associated memory using Ad delivery
systems. That we were able to generate significant improvement in pulmonary protection above
BCG with parenteral, rather than mucosal administration of boost vaccine is critical; suggesting that
the generation of effective mucosal immunity is possible, without the risks and challenges of
mucosal administration, but that further work to specifically enhance sustained protective immunity
is required.
206
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
182 Biomarkers & Vaccines
21/09/2016
A robust genetic marker to identify and quantify Mycobacterium tuberculosis Complex
based on evolutionary conservation criteria
G. Goig Serrano1, R. Borrás3, A. Gil-Brusola2, I. Comas1
1
Tuberculosis Genomics Unit, Instituto de Biomedicina de Valencia - CSIC 2Microbiology Service,
La Fe University Hospital 3Microbiology Service, University Clinic Hospital, INCLIVA Research
Institute, Valencia, Spain
Background: A number of nucleic acid amplification tests have been developed with the aim of
specifically detect Mycobacterium tuberculosis Complex DNA. However, many times these tests rely
on the amplification of genetic regions that are not unique to the MTBC, or does not take into
account the genetic diversity within the complex. This situation can led to sensitivity problems
across the globe. Now, we have the chance to develop diversity-aware diagnostic markers thanks to
the availability of large genomic databases representative, not only of the known MTBC diversity,
but also of the other non-tuberculosis mycobacteria. In this work, we identify by comparative
genome analyses, potential candidate loci specific to the MTBC. Furthermore, we validate our
approach by real time PCR with a panel of non-tuberculosis mycobacteria, representative MTBC
strains and sputum samples.
Material/methods: A list of fully conserved genes among Mycobacterium tuberculosis Complex
members was obtained by analyzing genomes of 219 strains representative of the global diversity of
the complex. An homology similarity search of these genes in the NCBI non-redundant nucleotide
database, was then performed in order to obtain potential markers that are unique to the MTBC.
DNA from 18 non-tuberculosis mycobacteria (NTM) species, different representative lineages of the
MTBC, and 10 clinical sputum samples, was extracted and then used to validate bioinformatic
results by real time PCR (RT-PCR). Sensitivity and specificity of one of these markers was finetuned to explore its potential as a diagnostic tool.
Results: We identified 49 genes to be totally conserved within the complex, from which 10 showed
to be unique to the MTBC. We tested five of them against non-tuberculosis Mycobacteria DNA
collection and representative strains of the MTBC. We further developed one of the markers to be
used for quantification and diagnosis of MTBC DNA. All markers tested by RT-PCR showed
amplification for MTBC DNA, and consistent amplification of DNA quantities up to 2E-5 ng (4CFU)
could be achieved when optimizing the reaction setup for one of them. Furthermore, none of the
reactions with non-tuberculosis mycobacteria DNA yielded a positive result, while robust
amplification was produced for every MTBC lineage. For clinical sputum samples, 9 out of 10
produced detectable amplification, being the remaining smear and culture negative.
Conclusions: We provide a reference set of 10 specific markers for the Mycobacterium
tuberculosis complex together with the empirical evaluation of 5 of them. When optimizing the
reaction setup for one of this markers, we achieve a 100% specificity for the MTBC, as well as a
high sensitivity up to 2E⁻5 ng of template DNA. In addition, since all markers are single copy genes,
these can be confidently used in real time PCR experiments to perform absolute quantification of
DNA and genome copies, so the bacterial load of a sample can be estimated.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
207
183 Biomarkers & Vaccines
21/09/2016
Analysis of differential expression of MDR and Susceptible Mycobacterium tuberculosis
proteins in profiles of INF-ɣ and its regulatory cytokines (IL-10) in MDR-TB patients DC cells
as a potential of therapeutic or TB vaccine strategy
A. Hadizadeh Tasbiti, S.H. Yari, M. Ghanei, M. Shokrgozar, A. Fateh, S. Siadat, F. Vaziri,
A. Bahrmand
TB, Pasteur institute of iran, Tehran, Iran
Tuberculosis is still a major public health challenge worldwide. One of the major challenges in
treating this disease is the widespread development of MDR-TB. The completion of the genomic
sequence provides a strong foundation for subsequent identification and characterization of proteins
of M.tuberculosis, leading to understanding of protein function and discovery of new drug targets
against tuberculosis. Membrane proteins are functionally important and structurally challenging.
This study has employed proteomic approach, which is a direct method, to identify proteins from
resistant M.tuberculosis isolates compared to sensitive isolates and consequently, we aimed to
investigate the cytokine response in MDR tuberculosis patients following stimulation with protein
candidate (Rv0379) of Mycobacterium tuberculosis. For this purpose, TB bacilli mechanically
disrupted and proteins extracted by Triton X-114 detergent phase separation. Proteins were
precipitated by adding refrigerated ethanol or saturated ammonium sulfate to the supernatant. Pellet
resuspended in PBS and then dialyzed for 24 h against PBS pH 7.4. IEF was carried out using the
Ettan IPGphor 3 isoelecteric focusing system. Proteins were separated in second dimension on
12% SDS-PAGE in a vertical electrophoretic dual gel. 2D gels were analyzed using Image Master,
Melanie 7.0 software. Mass spectrometry was performed using Autoflex II TOF/TOF. Protein
sequences were retrieved from Tuberculist server hosted by Pasteur Institute, Paris. PBMC were
isolated and suspended in RPMI-1640. Six days after starting DC culture, autologous T cells were
isolated from PBMC by negative selection. Cells were cultured subsequently in 24 well plates at 37,
5% CO2 and stimulated with 10 µg/ml protein candidate for 72 h. Cell culture supernatants were
assayed for cytokine production (IL-10 and INF-ɣ) by ELISA method. Results showed that majority
of commonly expressed/upregulated proteins belonged to the cellular metabolism and respiration
category (Rv 0946c, Rv 3331, Rv3586, Rv0866, Rv3057c, Rv3248c, Rv1133c, Rv0462, and
Rv1876). Rv 0946c: Involved in glycolysis and in gluconeogenesis. Rv 3331: Thought to be involved
in transport of sugar across the membrane. Responsible for the translocation of the substrate
across the membrane. Rv3586: Conserved hypothetical protein. Another hypothetical protein
(Rv2744c) and two membrane and cell wall fraction proteins (Rv0379, Rv1886c) were identified.
Analysis revealed increased percentage of INF-ɣ and decreased IL-10 levels in MDR-TB patients
compared to normal subjects. Fully matured monocyte derived DC are known to be strong induces
of Th1 dominated immune responses. This might be caused the cell wall fraction proteins, Rv0379,
induces a Th1 differentiation and committed towards a Th1 response. This present investigation
may allow the identification of some valuable vaccine and drug target candidates and this provide
basement for future designing of preventive, diagnostic and therapeutic strategies against TB.
208
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
184 Biomarkers & Vaccines
21/09/2016
Attenuated vaccinia virus expressing RpfE of Mycobacterium tuberculosis induces
effective immune responses in mice
E. Shin, H. Cha, S. Lee, J. Yoo, H. Jeong
Division of Vaccine Research, Korea Centers for Disease Control and Prevention, Osong, South
Korea
There is licensed BCG vaccine but the protective effect against infectious Mycobacterium
tuberculosis (M. tuberculosis) is limited in adults, so novel development of tuberculosis vaccines is
urgently required. Resuscitation promoting factor E (RpfE) is one of the five Rpf-like proteins in M.
tuberculosis. These Rpf-like proteins are secretory, which make them recognize by the host immune
system. Vaccinia virus as recombinant vectors and delivery models has several advantages, which
can carry many foreign genes and stimulate production of high level cell mediated immunity. In this
study, we constructed recombinant vaccinia virus expressing RpfE (VV/RpfE) of M. tuberculosis
using the attenuated vaccinia virus KVAC103. We immunized C57BL/6 subcutaneously with BCG or
VV/RpfE, then boosted with VV/RpfE. As results, VV/RpfE induced prodection of antigen specific
IgG (P<0.001) and CD4 polyfunctional T cell secreting cytokines (IFN-γ, IL-2 and TNF-α) in both
prime and boost vaccination. Also, the level of IFN-γ from lung after boost vaccination was
significantly increased. In further study, we will analyze protective efficacy through performing
challenge test and measuring immune response factors in mice.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
209
185 Biomarkers & Vaccines
21/09/2016
Mycobacterium tuberculosis Rv3112 elicits antigen-specific T cell immune response and
confers protective immunity against a predominant Beijing/K strain
H. Kim, W.S. Kim, J.S. Kim, S.B. Cha, S.J. Han, K.W. Kwon, S.J. Shin
Department of Microbiology, Brain Korea 21 PLUS Project for Medical Science, Yonsei University
College of Medicine, Seoul, South Korea
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), has affected human health for
thousands of years and remains a major cause of human death worldwide. Effective vaccination is
the most cost-effective measure to achieve long-term control of tuberculosis (TB). To achieve this
goal, the identification of vaccine target antigens recognized by the immune system during early
phase of infection with a powerful antigen-specific memory Th1 immune response is an essential
step in TB vaccine development. Here, we evaluated vaccine efficacy of Rv3112, a molybdenum
cofactor biosynthesis protein D (moaD1), against a predominant Beijing/K strain of South Korea.
First, we confirmed that host immune system is able to recognize Rv3112 during early phase of Mtb
K infection by generating Rv3112-specific T cell response. Second, Rv3112-immunized mice
remarkably maintained multifunctional CD4+ T cells co-expressing IFN-γ, TNF-α, and IL-2 in lungs
up to 10 weeks post-challenge. Finally, significant reductions of bacterial CFUs and lung
inflammation were observed in Rv3112-vaccinated mice. Collectively, our results demonstrate that
Rv3112 contributes to the generation of antigen-specific protective CD4+ T cell responses and may
be helpful in the design of improved TB vaccines.
210
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
186 Biomarkers & Vaccines
21/09/2016
Structure based epitope mapping of a 28kDa secretary antigen: a probable diagnostic
marker from Mycobacterium tuberculosis
P. Kundu, R. Biswas, A. Dutta, A.K. Das
Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
MTC28 is a proline-rich secreted antigen of Mycobacterium tuberculosis, conserved only in
pathogenic strains of mycobacterium. MTC28 have a diagnostic value in TB detection and it
increases the immunogenic response when used in combination with other major antigens such as
Ag85b, ESAT-6 and CFP-10. Therefore, we characterize this antigen by solving its crystal structure
and determine two immunogenic epitopes by both in silico and experimental epitope mapping. The
recombinant protein is purified and the structure is solved at 2.8Å (PDB ID: 4OL4) by MIRAS
method. Simulation studies are used for epitope prediction. In experimental mapping, trypsin
digested peptides of MTC28 are separated and purified by HPLC. The peptides are screened by the
ELISA with healthy control and patient sera. Deletion mutagenesis further confirms the epitope part.
A total of 30 pulmonary TB patients and 20 healthy individuals are included in the study. The
structure possesses a β sandwich like fold and belongs to space group P3121 with one molecule in
the asymmetric unit. Simulation studies predict five probable epitope sites in the structure. Two
peptides 1240.861 Da (128ALDITLPMPPR137) and 2141.102 Da (138WTQVPDPNVPDAFVVIADR156)
found to be reactive against tuberculosis patient sera are experimentally mapped and belong to the
predicted probable epitopes. ELISA studies show a significant reduction of reactivity of MTC28
A127_156Rdel against patient serum in comparison to MTC28. In conclusion, The crystal structure
of MTC28 is solved and two epitopes of this antigen are mapped experimentally. These epitopes
may serve as a diagnostic marker for TB detection in human.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
211
187 Biomarkers & Vaccines
21/09/2016
Engineering a pro-apoptotic BCG strain to improve the immunogenicity of the current TB
vaccine
A. Lau, V. Singh, H. Soualhine, Z. Hmama
Division of Infectious Diseases, Department of Medicine, University of British Columbia,
Vancouver, Canada
BCG vaccine, introduced almost 100 years ago, is the only option to prevent TB disease. It
efficiently protects newborns from meningeal TB but fails to prevent adult pulmonary TB. In fact, TB
kills 1.3 million people annually in areas where BCG vaccination is widely practiced. Thus, more
efficient TB vaccines are urgently needed.
Others and we have shown that BCG mimics features of virulent M. tuberculosis, in particular
attenuation of essential macrophage functions such as antigen presentation and apoptosis. One of
these studies revealed that defect in antigen presentation is largely due to down-regulation of the
cysteine protease cathepsin S (CatS), which prevents MHCII molecule maturation and proper
antigen peptide loading. Recent studies also suggested a potential role for cysteine proteases in the
regulation of apoptosis, a key cellular process used by the macrophage to i) contain and process
ingested bacteria and ii) facilitate cross-talk antigen presentation between the macrophage and
dendritic cells.
To reverse the phenotype of vaccine-mediated macrophage attenuation, we engineered a novel
BCG strain that expresses and secretes active CatS (BCG-CatS). Since caspase-3 plays a central
role in the execution of apoptosis, we also constructed a BCG strain secreting an active form of
caspase-3 (BCG-C3).
Macrophages infected with either recombinant strain elicited a pro-apoptotic phenotype as indicated
by increased annexin V staining, PARP degradation, and active caspase-3 production compared to
wildtype BCG. Furthermore, transcriptomic profiling of macrophages infected with BCG-CatS
revealed upregulation of key pro-apoptotic genes and downregulation of anti-apoptotic genes, which
were further confirmed by quantitative PCR analysis. Immunogenicity studies in mice vaccinated
with BCG-CatS or BCG-C3 showed increased antigen-specific CD4 and CD8 T-cell response, as
well as enhanced cytokine production and proliferation upon ex vivo restimulation. Of particular
note, immunogenicity responses from mice vaccinated with BCG-C3 exceeded the effects observed
with BCG-CatS, demonstrating that induction of apoptosis is key to achieving high immunogenicity
Collectively, this study shows that BCG modifications to promote host cell apoptosis alleviate
adverse traits of the wildtype BCG strain, resulting in increased antigen presentation, which leads to
highly immunogenic TB vaccines.
212
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
188 Biomarkers & Vaccines
21/09/2016
Improving T-cell assays for diagnosis of latent TB infection: potential role of a diagnostic
test based on Interleukin-2
S. Mahmoudi2, S. Mamishi1, B. Pourakbari1
1
Pediatric infectious disease research center 2Tehran University of Medical Sciences, Tehran, Iran
Background: Since gamma interferon release assays (IGRAs) cannot differentiate between active
tuberculosis and latent tuberculosis infection (LTBI), development of rapid and specific diagnosis
tools are essential for discriminating between active TB from LTBI. Both IGRAs are based on
Mycobacterium tuberculosis-specific antigens, namely, early secretory antigenic target 6 (ESAT-6)
and 10 KD culture filtrate (CFP-10). The aim of this study was to evaluate the potential value of IL-2
secretion by whole blood cells after stimulation with rESAT-6 and rCFP-10 for discriminating
between active and latent tuberculosis.
Methods: Interleukin-2 and IFN-γ were measured after blood stimulation of 90 cases (30 with active
TB, 30 with LTBI and 30 healthy controls) with recombinant ESAT-6 and CFP-10. Receiver
operating characteristic (ROC) curve analysis was conducted to determine the best IL-2 and IFN-γ
result thresholds in discriminating between cases with active or latent TB, and correspondent
sensitivity and specificity were recorded.
Results: The IFN-γ release assay demonstrated a good sensitivity and specificity (sensitivity 8384% and specificity 92%) for diagnosis of tuberculosis. The discrimination performance of IL-2
assay (assessed by the area under ROC curve) between LTBI and patients with active TB were
0.75 and 0.8 following stimulation with rESAT-6 and rCFP-10, respectively. Maximum discrimination
was reached at a cut-off of 11.6 pg/mL for IL-2 after stimulation with recombinant rESAT-6 with 72%
sensitivity and 79% specificity and 10.7 pg/ mL for IL-2 following stimulation with rCFP-10 with 75%
sensitivity 79% specificity, respectively.
Conclusion: This study demonstrates that rESAT-6 and rCFP-10 can provide a sensitive and
specific diagnosis of TB. In addition, it was shown that IL-2 may be serving as a marker for
discriminating of LTBI and active TB.
Key words: IL-2, discrimination, active TB, LTBI
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
213
189 Biomarkers & Vaccines
21/09/2016
Reactivation of a latent mycobacterial infection in adult zebrafish
M. Niskanen5, H. Myllymäki5, H. Luukinen5, M. Parikka5-4, M. Rämet5-3-2-1
1
Department of Pediatrics, Medical Research Centre 2Department of Children and Adolescents,
Oulu University Hospital, Oulu 3Department of Pediatrics 4Oral and Maxillofacial Unit, Tampere
University Hospital 5BioMediTech, University of Tampere, Tampere, Finland
An estimated one third of human population have a latentMycobacterium tuberculosis infection, and
have a 5 – 10% lifetime risk of reactivation to active tuberculosis. Often reactivation is caused by
immunosuppression, but otherwise the mechanisms of reactivation are poorly understood, hence
reactivation cannot be prevented. Lack of adequate animal models has hampered tuberculosis
research. Zebrafish (Danio rerio) has emerged as a promising model organism for research. Like
humans, adult zebrafish has both innate and adaptive immunity. In addition, a natural fish pathogen
and a close relative of M. tuberculosis, Mycobacterium marinum, causes a disease that resembles
human tuberculosis, having both a natural latency with granuloma structures, and reactivation
phases.
Our aim was to develop a method to reactivate latent mycobacterial infection in adult zebrafish. Fish
were infected with a low dose of mycobacteria, and the following latent infections were reactivated
by suppressing their immune system with cortisone feeding. Three different cortisones,
prednisolone, methylprednisolone and dexamethasone, were tested. Samples were collected in
different time points and bacterial counts of each fish were determined by qPCR. The granuloma
structures and dissemination of the bacteria were visualized from histological samples with ZiehlNeelsen and Trichrome staining protocols. Further effectivity of ethambutol was tested in both latent
and reactivated infections. To utilize the method in testing effectiveness of potential, new vaccine
candidates, latently infected fish were first vaccinated with groups of selected antigens and
cortisone feeding was started four weeks later to evaluate effectiveness of the vaccines.
Dexamethasone showed to be the most effective and safest cortisone to reactivate the latent
mycobacterial infection. Reactivation was seen as increased bacterial burdens: after six weeks, the
bacterial counts per fish had increased 50 – 450-fold. Histological staining confirmed increased
bacteria counts. Ethambutol was not effective against latent infection, but controlled progression of
reactivation. Of the DNA-based vaccine candidates, one showed protection against reactivation of
latent M. marinum infection. The developed reactivation method provides a novel way to screen
new drug and vaccine candidates and enables the study of reactivation of mycobacterial infection in
detail.
214
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
190 Biomarkers & Vaccines
21/09/2016
Construction and characterization of TB vaccines based on MTBVAC in modern lineages of
Mycobacterium tuberculosis
I. Pérez Sánchez2, C. Martín2-1, J. Gonzalo-Asensio2-1
1
CIBER enfermedades respiratorias. Instituto de Salud Carlos III, Madrid 2Genética de
micobacterias, University of Zaragoza, Zaragoza, Spain
Tuberculosis (TB) is caused by Mycobacterium tuberculosis (MTB) and is one of the infectious
diseases that cause more mortality (WHO, 2015). BCG is the only actual vaccine against TB, but its
efficacy against pulmonary diseases in adults is variable. To overcome this problem of BCG, it has
been constructed a new live attenuated vaccine candidate, MTBVAC, which is in clinical trials
(phase Ib, NCT02013245). MTBVAC is based on the deletion in phoP and fadD26 genes, both
related with virulence factors (Arbués, 2013). MTBVAC belongs to lineage 4 (Euro-American-Asian)
of Mycobacterium tuberculosis (Mtb).
The principal objective of this work is to construct the same deletions of MTBVAC in two strains of
linages 2 (Asian Beijing) and 3 (African-Indian). These are modern lineages, which together with
lineage 4, are highly distributed and responsible for the current transmission of MTB in humans.
We have selected clinical strains from lineages 2 and 3 using RFLP and spoligotyping techniques.
Vaccines will be constructed in these lineages using the same method that was used to construct
MTBVAC. Suicide plasmids which contains the gene (phoP or fadD26) disrupted by an antibiotic
resistance gene and a counter-selectable (negative) marker will be used (Arbués, 2013). Mutants
will be constructed through a two-step process involving a positive/negative double selection. The
antibiotic marker will be eliminated by using a γδ-resolvase which acts on res sites flanking the
resistance marker since these constructions will be used as potential vaccine candidates.
MTBVAC has been extensively characterized. It has been studied the transcriptome of the
transcription factor PhoP, its lipidomics profile by thin-layer chromatography in which it was
observed the devoid of cell-wall lipids phthiocerol dimycocerosates (DIM), diacyltrehaloses and
polyacyltrehaloses (DAT/PAT) as direct consequence of fadD26 and phoP deletions, respectively
(Arbués, 2013). It has also been studied the expression profile of the principal PhoP-regulated
genes (pks2, pks3, espA…) by qRT-PCR (Gonzalo-Asensio, 2008; Solans, 2014) and the inability
to secrete ESAT-6 (Gonzalo-Asensio, 2014) by Western blot. To study whether these newly
constructed vaccines show the same properties as MTBVAC, we plan to deeply characterize these
phenotypes prior to conduct to preclinical studies.
References:
•
•
•
•
•
•
Arbues, A., et al., Construction, characterization and preclinical evaluation of MTBVAC, the
first live-attenuated M. tuberculosis-based vaccine to enter clinical trials. Vaccine, 2013.
31(42): p. 4867-73.
Gonzalo-Asensio, J., et al., PhoP: a missing piece in the intricate puzzle of Mycobacterium
tuberculosis virulence. PLoS One, 2008. 3(10): p. e3496.
Gonzalo-Asensio, J., et al., Evolutionary history of tuberculosis shaped by conserved
mutations in the PhoPR virulence regulator. Proc Natl Acad Sci U S A, 2014. 111(31): p.
11491-6.
Solans, L., et al., The PhoP-dependent ncRNA Mcr7 modulates the TAT secretion system in
Mycobacterium tuberculosis. PLoS Pathog, 2014. 10(5): p. e1004183.
WHO (2015). Global Tuberculosis Report 2015.
Funded by: Diputación General de Aragón (DGA). European Project TBVAC2020.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
215
191 Biomarkers & Vaccines
21/09/2016
The mycobacterial capsule has an impact on the protective efficacy of BCG vaccination
against Mycobacterium tuberculosis in mice
R. Prados-Rosales
CICbioGUNE, Derio, Spain
Bacille Calmette-Guérin (BCG) is widely used for the prevention of tuberculosis (TB) throughout the
world, despite limited efficacy. Although the production of BCG vaccine for human use involves the
growth of the bacilli as a pellicle, most preclinical studies of BCG vaccine or Mycobacterium
tuberculosis (Mtb) strains grow bacteria in the presence of detergent to prevent clumping, which
also strips the mycobacterial capsule. The impact of the capsule on vaccine efficacy has not been
explored by the field. Here, we show that vaccination of mice with encapsulated BCG promotes a
more diverse and potent immune response relative to vaccination with unencapsulated BCG,
including higher polysaccharide-specific capsule antibody titers, higher IFN-g and IL-17 splenic
responses, and more multifunctional CD4+ T cells. When the same group of mice was challenged
with unencapsulated Mtb, we observed lower bacterial burden in encapsulated BCG-immunized
mice. We also infected mice with encapsulated Mtb. The combination of vaccination and challenge
with encapsulated strains resulted in the greatest protection efficacy. The transcriptome of Mtb
grown with and without detergent showed a response similar to that of starvation, hypoxia,
stationary phase or non-replicating persistence. In summary, the presence of capsule on BCG
affects the outcome of vaccination.
216
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
192 Biomarkers & Vaccines
21/09/2016
Control of Mycobacterium tuberculosis extrapulmonary dissemination in mice by an
arabinomannan-protein conjugate vaccine
R. Prados-Rosales
CICbioGUNE, Derio, Spain
Currently there are a dozen or so of new vaccine candidates in clinical trials for prevention of
tuberculosis (TB) and each formulation attempts to elicit protection by enhancement of cellmediated immunity (CMI). In contrast, most approved vaccines against bacterial pathogens are
believed to mediate protection by eliciting antibody responses. However, it has been difficult to
apply this formula to TB because of the difficulty in reliably eliciting protective antibodies. Here, we
developed capsular polysaccharide conjugates by linking mycobacterial capsular arabinomannan
(AM) to either Mtb Ag85b or B. anthracis protective antigen (PA). Further, we studied their
immunogenicity by ELISA and AM glycan microarrays and protection efficacy in mice. Immunization
with mycobacterial capsular AM conjugates to Mtb Ag85b and B. anthracis PA, elicited an AMspecific antibody response in mice. AM binding antibodies stimulated transcriptional changes in
Mtb. Sera from AM conjugate immunized mice reacted against a broad spectrum of AM structural
variants and specifically recognized arabinnan fragments. Conjugate vaccine immunized mice
infected with Mtb had lower bacterial numbers in lungs and spleen, and lived longer than control
mice. These findings provide strong evidence that humoral immunity can contribute to protection
against Mtb and suggest the inclusion of AM in future anti-TB vaccine formulations.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
217
193 Biomarkers & Vaccines
21/09/2016
Early biomarkers associated with progression of Latent Tuberculosis Infection to clinically
active disease - a longitudinal cohort study
N. Rakotosamimanana3, V. Richard4, T.M. Doherty1, B. Gicquel2, A. Zumla5, V. Rasolofo3
1
GSK, Wavre, Belgium 2Institut Pasteur Paris, Paris, France 3Mycobacteria Unit, Institut Pasteur de
Madagascar, Antananarivo, Madagascar 4Institut Pasteur Dakar, Dakar, Senegal 5UCL, Londres,
United Kingdom
Identifying those Mycobacterium tuberculosis (Mtb) latently infected individuals most at risk of
developing active tuberculosis (TB) using routine clinical and laboratory tests remains a huge
challenge in TB control efforts. We conducted a prospective longitudinal study to assess the risk of
developing active TB in latent Mtb infection.
HIV-negative household contacts (n=296) of pulmonary TB patients (n=85) and community controls
(n=186) monitoring clinical features, full blood cell counts, TST, and chest X-rays were recruited and
followed up regularly during 18 months. Paired statistical tests, the Kaplan -Meier method and the
Cox proportional hazard modeling were performed on variables between those contacts progressing
or not progressing to active TB.
The appearance of TB disease symptoms within the contacts was significantly associated with an
elevated peripheral percentage of blood monocytes (p = 0.01), a TST response ≥ 14 mm (p = 0.04)
and an increased monocyte: lymphocyte ratio (p = 0.03). A strong association was found with both
elevated blood monocyte percentage and TST ≥ 14 mm with risk of progression to TB in the
contacts (aHR= 11.31 (95%CI 3.29 to 38.85), p<0.001).
Elevated percentage of peripheral blood monocytes plus an elevated TST response are potential
biomarkers for identifying contacts of TB patients at risk of developing active TB.
218
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
194 Biomarkers & Vaccines
21/09/2016
Novel anti-tuberculosis vaccine carrier development and assessment by in vivo imaging
N. Redinger2, A. Zelmer4, D. Christensen1, P. Carroll3, K. Hagens2, P. L. Andersen1, T. Parish3,
U. E. Schaible2
1
Adjuvant Research, Department of Infectious Disease Immunology, Statens Serum Institute,
Copenhagen, Denmark 2Cellular Microbiology, Research Center Borstel, Borstel, Germany 3Barts
and The London School of Medicine & Dentistry, Garrot Building 4Department of Immunology and
Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
The main focus of this project is to combine novel imaging approaches with M. tuberculosis reporter
strains and/or QDot labeling of vaccine carrier nanoparticles to assess and monitor efficacy and
biodistribution of novel vaccine formulations against tuberculosis. The nanoparticle-based vaccine
formulations analysed were based on recombinant fusion protein H56 comprising Ag85B, ESAT-6
and Rv2660c linked to cationic liposomes (CAF01) containing TDB as adjuvant. Apart from
accelerating testing vaccines for protective efficacy, this approach will optimize vaccine formulations
and candidates. Fluorescent strains of M. tuberculosis were used in the murine tuberculosis model
and imaging individual animals and excised lungs over time by IVIS. The detection limit of
fluorescent M. tuberculosis in living mice was below detection limit, however, we were able to image
excised lungs to assess mycobacterial load.
We found that H56-CAF01 used for both, priming and boosting, induced protection against M.
tuberculosis but only when given subcutaneously (s.c.) but not via the intranasal (i.n.) route.
Fluorescent signals and CFU counts correlated well indicating the usability of the reporter system
for vaccine testing. Using H56-CAF01 as a boost for BCG primed immunity, we observed that BCG
alone already induced protection leading to reduced mycobacterial loads at the detection limit of the
imaging approach, which is less sensitive than the classical CFU assay.
In order to reveal the differential efficacy of the s.c. vs. the i.n. vaccination route, we analysed the
biodistribution of the vaccine carriers and delivery to draining lymphoid organs, which is a
prerequisite for T cell priming. We used QDots as tags to monitor vaccine carrier fate upon i.n. vs.
s.c. vaccination. Upon i.n. vaccination, IVIS imaging revealed that fluorescent signals quickly (<4 d)
faded from the lungs whereas some fluorescent were retained in the tracheal lymph nodes (LN) up
to day 10. Upon s.c. application, the dermal signal faded at around d15 but draining inguinal LN
retained the strong vaccine carrier signals at least till day 30 after vaccination. Immunohistology and
FACS analysis of draining LN showed a strong influx of CD11b+ macrophages and identified
CD11b+/CD11c+ myeloid cells as the predominant vaccine-carrying population. Here we show
superiority of the s.c. over the i.n. route for efficient vaccine delivery due to delayed retention in
draining LN.
In conclusion IVIS imaging can be used to monitor and evaluate the efficacy of new vaccine
candidates as well as the course of M. tuberculosis infection in excised lungs and to characterize
vaccine delivery and biodistribution in the mouse in order to assess immune defense against TB.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
219
195 Biomarkers & Vaccines
21/09/2016
Activation profile of Mycobacterium tuberculosis-specific CD4+ T cells reflects disease
activity, irrespective of HIV status
C. Riou3-2, T. Oni3-1-4, H.P. Gideon3-1, R. Goliath3-1, R. Wilkinson3-1-6-5, K.A. Wilkinson3-1-6
1
Clinical Infectious Diseases Research Initiative 2Department of Pathology, Division of Medical
Virology 3Institute of Infectious Disease and Molecular Medicine 4School of Public Health and
Family Medicine, Division of Public Health Medicine, University of Cape Town, Cape Town, South
Africa 5Department of Medicine, Imperial College London 6Mill Hill Laboratory, The Francis Crick
Institute, London, United Kingdom
The diagnosis of pulmonary tuberculosis in HIV-infected individuals is particularly challenging, as
HIV-induced alterations of the immune system lead to reduced cavitations, limiting the sensitivity of
sputum-based assays. Thus, alternate markers are needed to distinguish between latent (LTBI) and
active TB (aTB) in this high-risk group.
We therefore compared the potential of the activation and polyfunctional profiles of Mtb-specific
CD4+T cells to distinguish between LTBI and aTB in HIV-uninfected and HIV-infected individuals.
We analyzed 76 participants divided in four groups according to their TB and HIV status: LTBI/HIV-,
aTB/HIV-, LTBI/HIV+ and aTB/HIV+. Cryopreserved PBMCs were stimulated for 16 hours with
ESAT-6/CFP-10 peptide pool and stained using a live/dead marker and antibodies towards CD3,
CD4, CD8, HLA-DR, Ki67, CD38, IFN-γ, TNF-α and IL-2.
As previously shown, in HIV-uninfected persons, the levels of expression of HLA-DR, Ki67 and
CD38 on IFNγ+ Mtb-specific CD4+T cells were significantly higher in aTB participants when
compared to LTBI. Interestingly, while HLA-DR expression on Mtb-specific CD4+T cells in the
LTBI/HIV+ group (median 41.7%) was significantly higher when compared to the LTBI/HIV- group
(13.7%), HLA-DR expression on these cells was significantly further increased in HIV-infected
individuals with aTB (84%). HLA-DR expression on IFNγ+ Mtb-specific CD4+T cells distinguishes
LTBI and aTB in both the HIV- and HIV+ groups (AUC=0.98, p<0.0001; AUC=0.9, p<0.0001,
respectively). However, the optimum cutoff values discriminating LTBI from aTB were distinct for
HIV-uninfected (40%) and HIV-infected individuals (70%). When assessing the polyfunctional profile
of Mtb-specific CD4+ T cells our data show that the proportion of IFNγ+IL2+TNFα+ or IFNγ+IL2TNFα+ Mtb-specific CD4+T cells allowed the distinction between LTBI and aTB in HIV-uninfected
individuals (AUC=0.97, p<0.0001 and AUC=0.92, p<0.0001, respectively) but not in HIV-infected
persons.
Overall, these data show that HLA-DR expression level on Mtb-specific IFNγ+CD4+T cells
represents a robust marker to distinguish between LTBI and aTB in both HIV-uninfected and ART
naïve HIV-infected individuals. This suggests that despite HIV-induced systemic immune activation,
active bacterial replication promotes further up-regulation of HLA-DR on Mtb-specific CD4+T cells.
On the contrary, the polyfunctional profile of Mtb-specific CD4+T cells associated with TB status
solely in HIV-uninfected individuals. HLA-DR expression could represent an important alternate tool
to assess TB status irrespective of HIV status.
220
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
196 Biomarkers & Vaccines
21/09/2016
Blood transcriptomic diagnosis of pulmonary and extrapulmonary tuberculosis
J. Roe2, N. Thomas2, E. Gil2, K. Best2, E. Tsaliki2, S. Morris-Jones3, S. Stafford2, N. Simpson2,
K. Witt1, B. Chain2, R. Miller2, A. Martineau1, M. Noursadeghi2
1
Queen Mary University of London 2University College London 3University College London
Hospitals NHS Trust, London, United Kingdom
Background: Novel rapid diagnostics for active tuberculosis (TB) are required to overcome the time
delays and inadequate sensitivity of current microbiological tests that are critically dependent on
sampling the site of disease. Multiparametric blood transcriptomic signatures of TB have been
described, but the number of genes included remains a barrier to their development as a diagnostic
tool. We sought to identify the fewest possible blood transcripts that discriminate patients with active
TB from healthy individuals, and from those with other infectious diseases.
Methods: Support vector machine learning, combined with feature selection, was applied to new
and previously published blood transcriptional profiles in order to identify the minimum TB-specific
transcriptional signature shared by multiple patient cohorts including pulmonary and extrapulmonary
TB, and individuals with and without HIV-1 co-infection.
Results: We identified and validated elevated blood BATF2 transcript levels as a single sensitive
biomarker which discriminated active TB from healthy individuals, with receiver operating
characteristic (ROC) area under the curve (AUC) scores of 0.93-0.99 in multiple cohorts of HIV-1
negative individuals, and 0.85 in HIV-1 infected individuals. In addition, we identified and validated a
novel four-gene blood signature comprising CD177, haptoglobin, immunoglobin J chain and galectin
10, that discriminated active TB from other febrile infections giving ROC AUC of 0.94-1.
Conclusions: Elevated blood BATF2 transcript levels provide a sensitive biomarker that
discriminates active TB from healthy individuals, and a novel four-gene transcriptional signature
differentiates active TB and other infectious diseases in individuals presenting with fever.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
221
197 Biomarkers & Vaccines
21/09/2016
Evaluation and Assessment of Tuberculosis Diagnostic Testing Methods in Immigrant
Children (Ages 2-14)
A. Sharma, A. Lardizabal
Global Tuberculosis Institute, New Jersey Medical School, Somerset, United States
Objective: Assessment of current CDC recommended tuberculosis screening methods between the
United States and foreign countries, both the Mantoux Tuberculin Skin Test and Interferon-Gamma
Release Assay, used in immigrant children ages 2-14 years living in countries endemic to
tuberculosis.
Background: As per the CDC (Center for Disease Control) guidelines, children 2-14 years of age
immigrating to the United States from countries endemic to tuberculosis are required to undergo
one of two screening tests – tuberculin skin test (TST) or Interferon-gamma release assay (IGRA).
CDC defines countries endemic to TB as World Health Organization (WHO) estimated TB incidence
rate of greater than or equal to 20 cases per 100,000 people. TST is considered positive with
greater than or equal to 10 mm induration. The presence of IGRA in serum is considered positive. If
either test is positive, radiological chest tests are performed and appropriate management is
undertaken. This study will assess validity of screening tests performed on immigrant children ages
2-14 years by comparing the tests performed in the patient’s respective country of origin and the
tests upon immigrating to the United States.
Method: Cohort retrospective study of patients seen in affiliation with the New Jersey Medical
School Global Tuberculosis Institute will be reviewed. Charts of recent immigrant children, ages 214, seen at the Lattimer Clinic from 2013-2014 will be reviewed. Initial screening methods will be
reviewed in association with final diagnosis of these respective patients seen at the Lattimer Clinic.
Outcome: Based on cohort retrospective study, there was 38% concordance between the two tests
and 62% discordance. This can be due to multiple factors including non-skilled application and
interpretation of TSTs and recent BCG vaccination. Additionally, the study results put in question
the clause found in the CDC’s “Technical Instructions For Tuberculosis Screening and Treatment” of
not requiring immune response testing in patients with documented diagnostic test results from their
respective country of origin.
222
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
198 Biomarkers & Vaccines
21/09/2016
Immunodominant protein MIP_05962 from Mycobacterium indicus pranii displays
chaperone activity and induces Th1 mediated T cell response
A. Sharma2, M.J. Equbal2, M. Saqib3, J.A. Sheikh4, S. Pandey1, N.Z. Ehtesham4, S. Bhaskar3,
T.K. Chaudhuri2, S.E. Hasnain2-1
1
Dr Reddy’s Institute of Life Sciences, University of Hyderabad, Hyderabad 2Kusuma School of
Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas 3National Institute of
Immunology 4National Institute of Pathology, Safdarjung, New Delhi, India
Tuberculosis still remains a major threat to human beings. At present BCG is the only vaccine for
TB, however protective efficacy varies from 80% to none to all and that too is only effective in
preventing childhood TB and also fails to impart protection during reactivation of latent TB.
Therefore, vigorous efforts are on for new TB vaccine development. Mycobacterium indicus pranii
(MIP), a saprophytic, non-pathogen organism, is emerging as a promising intervention against a
number of diseases by virtue of its strong immunomodulatory functions and continues to be
extensively used as an intervention against leprosy. Comparative genome sequences analyses of
MIP, BCG and M.leprae, revealed that MIP and M.leprae contain 29 novel genes having significant
antigencity index revealed by in-silico analysis and absent in BCG (Saini V, et al. Nucleic Acids
Research, 2012). Most of these gene products are potentially highly immunogenic proteins. One of
these putatively immunogenic proteins, MIP_05962 belongs to Hsp20 protein family due to the
presence of α-crystallin domain and also has a very high protein identity with antigen of
M.tuberculosis and Hsp18 of M.leprae. Hsp18 of M.leprae, a homolog of MIP_05962, is a highly
antigenic protein, carries a number of various common T cell epitopes with M.tuberculosis and
elicits good CD4+T-cell response. With this background, the chaperone and immunological functions
of MIP_05962 gene were investigated. Purified recombinant MIP_05962, expressed in E.coli,
showed thermal aggregation prevention, thermal inactivation prevention and in-vitro refolding of
substrate proteins, exhibited interaction with non-native protein, provided in-vivo viability of E.coli
and also showed in-vivo assisted refolding of substrate protein. These properties of MIP_05962
protein lead us to conclude that the protein resembles molecular chaperone. Immunological
properties of this protein were investigated in mice model. MIP_05962 showed significant
proliferation of lymphocytes which confirmed that MIP_05962 is highly antigenic and induced
significant Th1 type of T cell mediated response both in-vitro and intracellularly. It also evoked
significant effector memory response in CD4+T cells. Th1-type cell mediated response is critical for
controlling TB infection; Therefore MIP_05962 has a potential for use as a subunit vaccine or
booster with BCG against tuberculosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
223
199 Biomarkers & Vaccines
21/09/2016
Th1-type T cell immunity driven by TLR2-mediated activation of dendritic cells and vaccine
potential against tuberculosis: An example of Mycobacterium tuberculosis Rv3628
W.S. Kim, J.S. Kim, S.B. Cha, H. Kim, K.W. Kwon, S.J. Shin
Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
Identification of vaccine target antigens (Ags) that induce Ag-specific Th1 immunity is the first step
toward the development of a tuberculosis vaccine. Here, we evaluated the Mycobacterium
tuberculosis (Mtb) protein Rv3628, a soluble inorganic pyrophosphatase, as a vaccine target and
characterized the molecular details of its interaction with dendritic cells (DCs). Rv3628 activated
DCs, increasing their expression of cell surface molecules and augmenting their production of TNFα, IL-1β, IL-6, and IL-12p70. Rv3628 mediated these effects by binding to TLR2 and activating
downstream MyD88-, MAPK- and NF-κB-dependent signaling pathways. Rv3628-stimulated DCs
induced the expansion of OVA-specific CD4+ and CD8+ T cells, which secreted IFN-γ and IL-2.
Rv3628-specific effector/memory T cells expanded to a similar extent as those stimulated with
ESAT-6 Ag in samples of lung and spleen cells collected from Mtb-infected mice. Finally, an Rv3628
subunit vaccine adjuvanted with dimethyldioctadecylammonium liposomes containing
monophosphoryl lipid-A caused significant reductions in bacterial counts and lung inflammation after
challenge with the hyper-virulent Mtb K strain. Importantly, protective efficacy was correlated with
the generation of Rv3628-specific CD4+ T cells co-producing IFN-γ, TNF-α and IL-2 and exhibiting
an elevated IFN-γ recall response. Thus, Rv3628 polarizes DCs toward a Th1 phenotype and
promotes protective immunity against Mtb infection.
224
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
200 Biomarkers & Vaccines
21/09/2016
Pulmonary immunity and durable protection induced by the ID93/GLA-SE vaccine
candidate against the hyper-virulent Korean Beijing Mycobacterium tuberculosis strain K
S.B. Cha1, W.S. Kim1, H. Kim1, K.W. Kwon1, S.N. Cho1, R.N. Coler2, S.G. Reed2, S.J. Shin1
1
Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
2
Infectious Disease Research Institute, Seattle, United States
The majority of tuberculosis (TB) vaccine candidates advanced to clinical trials have been evaluated
preclinically using laboratory-adapted strains. However, it has been proposed that challenge with
clinical isolates in preclinical vaccine testing could provide further and more practical validation.
Here, we tested the ID93/GLA-SE TB vaccine candidate against the clinical Mycobacterium
tuberculosis (Mtb) strain K (Mtb K) belonging to the Beijing family, the most prevalent Mtb strain in
South Korea. Mice immunized with ID93/GLA-SE exhibited a significant reduction in bacteria and
reduced lung inflammation against Mtb K when compared to non-immunized controls. In addition,
we analyzed the immune responses in the lungs of ID93/GLA-SE-immunized mice, and showed that
ID93/GLA-SE was able to elicit sustained Th1-biased immune responses including antigen-specific
multifunctional CD4+ T cell co-producing IFN-γ, TNF-α, and IL-2 as well as a high magnitude of IFNγ response for up to 10 weeks post-challenge. Notably, further investigation of T cell subsets in the
lung following challenge showed remarkable generation of CD8+ central memory T cells by
ID93/GLA-SE-immunization. Our findings showed that ID93/GLA-SE vaccine confers a high level of
robust protection against the hypervirulent Mtb Beijing infection which was characterized by
pulmonary Th1-polarized T-cell immune responses. These findings may also provide relevant
information for potential utility of this vaccine candidate in East-Asian countries where the Beijing
genotype is highly prevalent.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
225
201 Biomarkers & Vaccines
21/09/2016
Impared peripheral blood CD8 T cell differentiation in HIV patients with pulmonary
tuberculosis
E. Vasileva1-4, G. Maximov2, I. Kudryavtsev3, M. Serebryakova3, V. Verbov4, A. Totolian4
1
N.F. Gamaleya FRCEM, Moscow 2City Tuberculosis Dispensary 3Institute of Experimental
Medicine 4Saint-Petersburg Pasteur Institute, Saint-Petersburg, Russia
Background: Tuberculosis (TB) is the leading cause of death among HIV-positive patients.
Previously we have shown that there is no difference in the diagnostic value of QuantiFERON®-TB
Gold In-Tube test (QFT) among patients with HIV infection, in comparison with patients without HIV.
But it is known, that immune deficiency caused by HIV due primarily to a decrease in the number
and the functions of CD4 T cells, that result in reduction of production and biological action of
effector cytokines, especially IFNγ, that is measured in QFT.
Methodology/Principal Findings: To answer the question which cells have produced IFNγ, we
recruited 30 patients with HIV infection and pulmonary tuberculosis (TB+HIV+, n = 30) and patients
without HIV infection with active tuberculosis (TB+HIV-, n = 29). In addition to the QFT we assess
the degree of maturity of peripheral blood T cells using flow cytometry. Peripheral blood samples
were stained with antibodies to CD3, CD4, CD8, CD27, CD28, CD45, CD45RA and CD62L. There
was no significant difference (p=0,724) between the percentage of positive QFT results in TB+HIV+
(70%, median 1,36 IU/ml) and TB+HIV– (62 % median, 0,77 IU/ml).
If to speak about CD4+ T cells, the percentages of naïve (N, CD27+CD28+CD45RA+CD62L+),
central (CM, CD27+CD28+CD45RA+CD62L–), transitional (TM, CD27+CD28+CD45RA–CD62L–)
and effector (EM, CD27–CD28+) memory were significantly higher in TB patients without HIV
(p<0.001, p<0.001, p<0.001 and p=0.004, respectively). Whereas the percentages of TM, EM and
terminally-differentiated CD45RA-positive effector (CD27–CD28–) CD8 T cells were significantly
lower in TB patients without HIV (p=0.002, p<0.001 and p<0.001, respectively).
Conclusions: These findings indicate that there is a significant difference in relative contents of
CD8 T cell subsets in TB patients without HIV in comparison with TB+HIV+. Our data suggest that
HIV-TB coinfection impairs CD8+ T-cell differentiation. Thus, it is possible, that especially CD8 cells
produce IFNγ in patients with HIV in QFT. But in this case IFNγ has no protection against
Mycobacterium tuberculosis. So, it is important to examine this trend in a group of HIV patients
without TB.
226
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
202 Biomarkers & Vaccines
21/09/2016
Death and Detection: Development of a human challenge strain of Mycobacterium
tuberculosis
J. Wagner1, J. Fohtung1, A. Lotterman1, T. Mellors2, J. Hill2, B. Walker3, N. Cadieux3, S. Fortune1,
E. Rubin1
1
Harvard T.H. Chan School of Public Health, Boston, Ma 2Thayer School of Engineering,
Dartmouth College, Hanover 3Aeras, Rockville, United States
Human challenge studies utilizing model strains are a powerful tool for the development of effective
treatments and vaccinations for infectious disease. Due to the lengthy required treatment time, high
rate of relapse, and difficulty in quantitative assessment of bacterial load, no human challenge
model strain currently exists for the pathogen Mycobacterium tuberculosis. An ideal human
challenge strain will require both a “kill switch” to completely and efficiently eliminate the infection in
a host after a specified period of time, and a detection system designed to give real time
quantitative measurement of bacterial load. Here we report the development of systems to
accomplish these goals.
Utilizing an orthogonal system for the incorporation of the non-canonical amino acid p- iodo-Lphenylalanine (pIF) into essential proteins, we have developed a strain of M. smegmatis that is
dependent on the supplementation of pIF for survival with an escape rate of ~10-7-10-8
escapes/CFU. By placing strains grown in the presence of pIF into media lacking the supplement,
we observe several generations of growth until the “protein reservoir” is depleted resulting in growth
arrest of the bacterial population.
In order to quantify the bacterial load in the lungs, we have genetically engineered a strain of M.
tuberculosis to produce the volatile methyl salicylate, also known as the wintergreen scent. Volatile
production through GC/MS can be quantified using this molecular marker to monitor bacterial loads.
These two systems represent the first steps towards development of a human challenge strain of M.
tuberculosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
227
203 Biochemistry & Chemical biology
21/09/2016
Using a novel hybrid method to target DnaG primase for the design of a new class of
Mycobacterium tuberculosis antibiotics
B. Akabayov
Chemistry, Ben-Gurion University of the Negev, Be'er Sheva, Israel
The DNA replication process in Mycobacterium tuberculosis (Mtb) is a promising but underexploited
target for the development of novel antibiotics.
We have developed an approach to identify inhibitors for Mtb DnaG primase, which is a key enzyme
in the DNA replication machinery of Mtb. For the development process we have used DNA primase
from bacteriophage T7. T7 primase has several structural features that are similar to bacterial
(including Mtb) primases, making it an ideal model to study bacterial primases.
Using NMR screening, fragment molecules that bind T7 primase were identified and then exploited
in virtual filtration to select larger molecules from a virtual library. The molecules were docked to the
primase active site using the available T7 primase crystal structure and ranked based on their
binding energies to identify the best candidates for functional and structural investigations.
Biochemical assays revealed that some of the molecules inhibit T7 primase-dependent DNA
replication. The binding mechanism was delineated via NMR spectroscopy. Importantly, some of the
molecules inhibit also the activity of DnaG primase of Mtb. Our studies yielded new class of
antituberculous agents and provide new tools for fragment-based lead discovery.
228
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
204 Biochemistry & Chemical biology
21/09/2016
Lcp1 is a phosphotransferase responsible for ligating arabinogalactan to peptidoglycan in
Mycobacterium tuberculosis
J. Harrison2, G. Lloyd2, M. Joe1, T.L. Lowary1, E. Reynolds2, H. Walters-Morgan2, A. Bhatt2,
A. Lovering2, G. Besra2, L.J. Alderwick2
1
Alberta Glycomics Centre and Department of Chemistry, University of Alberta, Gunning-Lemieux
Chemistry Centre, Edmonton, Canada 2Institute of Microbiology & Infection, School of Biosciences,
University of Birmingham, Birmingham, United Kingdom
Mycobacterium tuberculosis, the etiological agent of tuberculosis, has a unique cell envelope which
accounts for its unusual low permeability and contributes to resistance against common antibiotics.
The main structural elements of the cell wall consist of a cross-linked network of peptidoglycan (PG)
in which some of the muramic acid residues are covalently attached to a complex polysaccharide,
arabinogalactan (AG) via a unique a-L-rhamnopyranose–(1®3)-a-D-GlcNAc-(1®P) linker unit. Whilst
the molecular genetics associated with PG and AG biosynthetic pathways have been largely
delineated, the mechanism by which these two major pathways converge has remained elusive. In
Gram positive organisms, the LytR-CpsA-psr (LCP) family of proteins are responsible for ligating
cell wall teichoic acids to peptidoglycan, through a linker unit that bears a striking resemblance to
that found in mycobacterial arabinogalactan. In this study we have identified Rv3267 as a
mycobacterial LCP homolog that encodes for a phosphotransferase which we have named Lcp1.
We demonstrate that lcp1 is an essential gene required for cell viability and show that recombinant
Lcp1 is capable of ligating AG to PG in a cell free radiolabeled assay.
Importance
Tuberculosis is an infectious disease caused by the bacterial organism Mycobacterium tuberculosis.
Survival of M. tuberculosis rests critically on the integrity of its unique cell wall, therefore a better
understanding of how the genes and enzymes involved in cell wall assembly work is fundamental
for us to develop new drugs to treat this disease. In this study, we have identified Lcp1 as an
essential phosphotransferase that ligates together arabinogalactan and peptidoglycan, two crucial
cell wall macromolecules found within the mycobacterial cell wall. The discovery of Lcp1 sheds new
light on the final stages of mycobacterial cell wall assembly and represents a key biosynthetic step
that could be exploited for new anti-TB drug discovery.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
229
205 Biochemistry & Chemical biology
21/09/2016
Cycloserines inactivate the PLP-dependent branched-chain aminotransferase IlvE from
Mycobacterium tuberculosis
T. Amorim Franco, L. Favrot, S. Cameron, O. Vergnole, J. Blanchard
Biochemistry, John S. Blanchard Laboratory, Albert Einstein College of Medicine, Bronx, United
States
D-cycloserine (DCS; or Seromycin) is a cyclic amino acid analog used in the treatment of multidrugresistant tuberculosis (MDR-TB). This natural product displays irreversible inhibitory activity against
the pyridoxal 5’-phosphate (PLP)-dependent alanine racemase (AR) and, reversible inhibition of the
D-alanine-D-alanine ligase (DAL). DCS has been shown to inactivate PLP-dependent enzymes
from different organisms, raising the question of whether the effects of this drug in MDR-TB are only
due to its inhibition of AR and DAL.
In the present study, we have examined the ability of DCS- and L-cycloserine (LCS) to inhibit the
essential PLP-dependent branched-chain aminotransferase from Mycobacterium tuberculosis
(MtIlvE). MtIlvE, is inhibited by DCS and LCS, both in a time- and concentration-dependent manner.
In addition, we have crystallized the D-cycloserine-inhibited enzyme, and solved the structure to 1.7
Å. The bound, noncovalent PMP-DCS adduct reveals that the DCS ring is aromatic as evidenced by
our 13C-NMR studies, and as proposed and observed in other enzyme systems. In addition, we
have evidence of a disulfide bond formation that covalently links the two monomers, unlikely any
other orthologue. MIC studies revealed that LCS is a much more potent inhibitor of M. tuberculosis
than D-cycloserine. Together these results confirm that the effects of cycloserine are broader than
previously thought.
230
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
206 Biochemistry & Chemical biology
21/09/2016
The active site architecture in peroxiredoxins: a case scenario for Mycobacterium
tuberculosis AhpE
B. Pedre, L. Van Bergen, A. Palló, V. Dufe, K. Wahni, I. Van Molle, L. Astolfi Rosado, H. Erdogan,
M. Alonso, F. De Proft, J. Messens
Structural Biology Brussel, Vrije Universiteit Brussel, Brussels, Belgium
Peroxiredoxins catalyze the reduction of peroxides, a process of vital importance to survive
oxidative stress. A nucleophilic cysteine, also known as peroxidatic cysteine, is responsible for this
catalytic process. We used the alkyl hydroperoxide reductase E from Mycobacterium tuberculosis
(MtAhpE) as a model to investigate the role of the chemical environment on the specificity of the
reaction. By an integrative structural (R116A - PDB 4XIH; F37G – PDB 5C04), kinetic and
computational approach, we explain the mutational effects of key residues in its environment. This
study shows that the active site residues are specifically oriented to create an environment which
selectively favours a reaction with peroxides.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
231
207 Biochemistry & Chemical biology
21/09/2016
The pMy vector series: a versatile toolkit for the recombinant production of mycobacterial
proteins
K. Beckham1, S. Staack1, N. Hanna1, A. Geerlof2, M. Wilmanns1, A. Parret1
1
European Molecular Biology Laboratory, Hamburg 2Institute of Structural Biology, Helmholtz
Zentrum München, Munich, Germany
Structural characterisation of mycobacterial proteins is a key step in the development of novel
therapeutic agents. Several studies have reported that the use of Escherichia coli as an expression
host for mycobacterial targets can result in misfolded or insoluble proteins1. Switching to the related
Mycobacterium smegmatis expression system2 offers several advantages, including the presence of
mycobacterium-specific chaperones and post-translational modification pathways which can
promote the correct folding of mycobacterial proteins. Here, we report a new series of modular
vectors designed for mycobacterial protein production in M. smegmatis. The pMy vector series
includes a range of promoters, resistance cassettes and affinity tag positions which can be used in
combination for co-expression of protein complexes.
In this study, we systematically investigated the expression of several difficult proteins including
membrane proteins, toxic proteins and protein complexes using different inducible promoter
systems. By exchanging the antibiotic resistance cassette, we have created vectors suitable for the
co-expression of two genes which can be controlled by two independent promoters. This vector
series expands the current toolkit for the overexpression of mycobacterial proteins in M. smegmatis.
All the vectors generated in the study are available at Addgene.
References
1. Bashiri and Baker (2015) Protein Science 24:1
2. Noens et al. (2011) BMC Biotechnology 11:27
232
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
208 Biochemistry & Chemical biology
21/09/2016
Deleting a unique polypeptide γ-loop of mycobacterial F-ATP synthase affects its ATP
catalysis and H+-pumping
A. Hotra3-2-1, M. Suter4, G. Biukovic4, P. Ragunathan2, S. Kundu4, T. Dick4, G. Grüber2
1
Nanyang Institute of Technology in Health and Medicine, Interdisciplinary Graduate School,
Nanyang Technological University, Singapore 2School of Biological Sciences, Nanyang
Technological University Singapore 3School of Physical and Mathematical Sciences, Nanyang
Technological University, Singapore 4Department of Microbiology and Immunology, Yong Loo Lin
School of Medicine, National University of Singapore, Singapore, Singapore
The discovery of the drug bedaquiline, which specifically targets mycobacterial F-ATP synthase,
proved that the enzyme is essential for both aerobically growing and dormant mycobacteria (1,2). In
mycobacteria the enzyme catalyses ATP synthesis by the catalytic domain (α3/β3 subunits) at the
expense of the torque energy that is generated by the rotating transmembrane H+-pump (a/c9
subunits) and the connecting central stalk γ/ε subunits. The reversible reaction, a weak or so called
“latent” ATP hydrolysis was also observed in some of the fast growing mycobacteria (3,4).
Previously, we described the solution structure of mycobacterial subunit γ (γ1-204) and its unique
polypeptide loop γ166-179, the latter was assigned by a sequence analysis in the close proximity of
rotating c-ring (5). Here we determined whether the loop γ166-179 affects ATP catalysis and
subsequent H+-pumping (6). We performed functional studies of F-ATP synthase containing γ (wild
type) or γ without the loop (Δγ166-179), using inverted membrane vesicles (IMV) of strain M.
smegmatis mc2 155 and a set of different F-ATP synthase inhibitors. The overall reduction of ATP
hydrolysis in the presence of different inhibitors, confirmed that the wild type (WT) F-ATP synthase
has a low ATP hydrolytic activity. An increase of the ATP hydrolytic activity by 34% and the
reduction of the ATP synthesis activity by 54% were observed in IMV of the mutant lacking the loop,
as compared with WT F-ATP synthase. So far, ATP hydrolysis driven H+-pumping was not observed
in mycobacteria (4). However, deletion of the γ-loop resulted in coupling of ATP hydrolysis and H+pumping (6). In summary, these results suggest that the γ-loop plays a distinctive role in regulating
and coupling of ATP synthesis/hydrolysis with H+-pumping.
1. Andries, K. et al. (2005). Science, 307, (5707), 223-227.
2. Koul, A. et al. (2008). J. Biol. Chem., 283, (37), 25273-25280.
3. Higashi, T. et al. (1975). J Biol Chem, 250, (16), 6541-6548.
4. Haagsma, A.C. et al. (2010). FEMS Microbiol Lett, 313, (1), 68-74.
5. Priya, R. et al. (2013). J Bioenerg Biomembr, 45, (1-2), 121-129.
6. Hotra, A. et al. (2016). FEBS J, 1-16.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
233
209 Biochemistry & Chemical biology
21/09/2016
Functional, biochemical and structural characterization of MSMEG_6753, a novel FabG-like
protein from Mycobacterium smegmatis
M. Blaise2, N. Van Wyk2, F. Baneres Roquet2, M. Drancourt1, Y. Guerardel4, L. Kremer2-3
1
Université Aix-Marseille, URMITE, UMR63, CNRS 7278, IRD 198, INSERM 1095,
Marseille 2Centre d’étude d’agents Pathogènes et Biotechnologies pour la Santé (CPBS), CNRSFRE3689, 1919 route de Mende, 34293 Montpellier 3INSERM-CPBS, Montpellier 4Unité de
Glycobiologie Structurale et Fonctionnelle, CNRS UMR 8576, IFR 147, Université des Sciences et
Technologies de Lille, Villeneuve D'ascq, France
Ketoacyl-acyl carrier protein (ACP) reductase (FabG) catalyse the reduction of thioesters linked to
an ACP. FabG are ubiquitous in bacteria and found associated to the type II fatty acid synthase
(FAS-II). Mining the Mycobacterium smegmatis genome database unravelled the existence of
several putative FabG-like enzymes. Among them, MSMEG_6753 exhibits 30% primary sequence
identity with FabG1/MabA, the essential ketoacyl-ACP reductase of FAS-II in Mycobacterium
tuberculosis. Interestingly, MSMEG_6753 is adjacent to MSMEG_6754, encoding a recently
characterized enoyl-ACP dehydratase and to MSMEG_6755, encoding a putative enoyl reductase.
Thus, this genome organisation is evoking a fatty acid biosynthetic pathway functionally related to
FAS-II.
Recombinant MSMEG_6753 was purified and assayed for ketoacyl-reductase activity by monitoring
the reduction of acetoacetyl-CoA in the presence of NADPH, albeit MSMEG_6753 is catalytically
less efficient than MabA from M. smegmatis. In addition, MSMEG_6753 was able to functionally
complement a FabG thermosensitive E. coli mutant, further confirming its activity in relation to fatty
acid synthesis. A ΔMSMEG_6753 deletion mutant was constructed in M. smegmatis, indicating that
this gene is dispensable for mycobacterial growth. However, the mutant exhibited differences in
fatty acid composition as compared to the parental strain. Furthermore, we have solved the crystal
structures of MSMEG_6753 in its apo form and as well as in complex with NADP+ at 1.9Å and 2.2Å
resolution, respectively. The overall structures were highly similar to the one of M. tuberculosis
MabA and to other bacterial FabG structures that are part of FAS-II. Unexpectedly, the binding of
NADP+ to MSMEG_6753 induces an important structural rearrangement. Our structural analyses
support also the capacity of MSMEG_6753 to process smaller acyl-chains than MabA.
Overall, these data add new structural insights towards our understanding of the enzymatic
mechanism of FabG proteins and demonstrate that MSMEG_6753 encodes a new FabG-like
member acting on fatty acids.
234
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
210 Biochemistry & Chemical biology
21/09/2016
Investigating Tuberculosis Drug Metabolism Phenotype (DMP) using Metabolomics
Approach
M. Das2, R. Arya2, S. Debnath1, R. Debnath1, A. Lodh1, S. Bishwal2-3, A. Das1, R. Nanda2
1
Department of Tuberculosis and Respiratory Disease, Agartala Government Medical
College,Tripura, Agartala 2Transnational Health Group, International Centre for Genetic
Engineering and Biotechnology (ICGEB), New Delhi 3School of Life Sciences, Sambalpur
University, Sambalpur, India
Treatment of tuberculosis involve lengthy (at least 6 months) and intensive intervention with multiple
antibiotics primarily Rifampicin (RMP), Isoniazid (INH), Pyrazinamide (PZA) and Ethambutol (ETB).
Drug metabolism phenotype or how a patient metabolizes these drugs determines its efficacy,
adverse effect vis a vis adherence and resistance development. Unbiased metabolomics is useful to
identify endogenous and exogenous metabolites like drugs and its metabolites present in different
circulatory and excretory biofluids. Untargeted metabolome analysis was carried out of urine
collected from drug naïve active TB patients (n=20) at time points (2/6/12/24/36/48 hours) post
dose, using a gas chromatography-time of flight-mass spectrometry (GC-Tof-MS). Broad objective
of this activity was to carry out a rigorous analysis to extract important insights from the generated
metabolome metadata (324x124). From this, we could successfully identify three parent drugs viz.
INH, ETB, PZA and majority of their metabolites and their excretion pattern at different time points.
We demonstrated drug metabolism phenotype (AcINH/INH, Pyrazinoic acid-POA/PZA) of the study
population. Most importantly, using correlation analysis we identified a novel metabolite of ETB and
few acute response metabolites of endogenous origin influenced by TB drugs. This novel metabolite
may explain mechanism of neuropathic side effect of ETB administration in some patients. Our
observations highlight the usefulness of untargeted metabolomics to understand drug metabolism
phenotype of TB patients at population level.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
235
211 Biochemistry & Chemical biology
21/09/2016
Mur pathway enzymes as potential drug targets: Development of a one-pot assay for
screening novel inhibitors targeting Mur enzymes
K. Eniyan1, A. Kumar2, G. Rayasam2, A. Perdih3, U. Bajpai1
1
Ant-Mycobacterial Drug Discovery Lab, Department of Biomedical Science, Acharya Narendra
Dev College 2Council of Scientific and Industrial Research, Open Source Drug Discovery, New
Delhi, India 3National Institute of Chemistry, Ljubljana, Slovenia
Background: The alarming rise in multi-drug resistance in Mycobacterium tuberculosis urges the
need for discovering novel drug targets in the pathogen. Enzymes involved in the cytoplasmic steps
of peptidoglycan biosynthesis in M. tuberculosis appear promising candidates for finding novel antituberculosis drugs.. Mur pathway enzymes (MurA–MurF) catalyze the synthesis of UDP-Nacetylmuramyl pentapeptide, a key precursor molecule required for the formation of the
peptidoglycan monomeric building blocks. The enzymes are essential for bacterial viability, their
homologues are reported to be absent in eukaryotic cells and have not yet been targeted by
clinically approved antibacterial compounds
Methods: In this study, the six enzymes (MurA-MurF) from Mycobacterium tuberculosis were
cloned, expressed and purified by affinity chromatography. Through successive coupled enzyme
assays, a one-pot assay designed to reconstitute the Mur pathway in vitro has been developed. The
intermediates and the final product formed in the assays were identified by ESI-MS. To examine the
sensitivity of the one-pot assay for screening of inhibitors, D-Cycloserine and a few furan-based
benzene-derived compounds were tested.
Results: A one-pot assay for the in vitro synthesis of UDP-MurNAc pentapeptide is established and
validated as an effective screen for potential inhibitors. D-Cycloserine and two furan-based
benzene-derived compounds have been identified as multi-target inhibitors of Mur pathway
enzymes.
Significance: Mur enzymes have not been tested for their potential as drug targets for antitubercular drug development. The one-pot assay developed in this study is amenable to high
throughput screening (HTS) and provides a strategy to screen for novel inhibitors against multiple
enzymes in Mur pathway of Mycobacterium tuberculosis. Also, it is a micro-titer based assay that
requires a short period of incubation and does not mandate addition of expensive intermediates
(sugar nucleotides) of the pathway, which further adds to its potential as a cost-effective HTS of
drug candidates.
236
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
212 Biochemistry & Chemical biology
21/09/2016
Structural Characterization of Mycobacterium smegmatis RimJ, an N-acetyltransferase
Protein
L. Favrot, S. Hegde, J. Blanchard
Biochemistry Department, Albert Einstein College of Medicine, Ny, United States
The GCN5-related N-acetyltransferases (GNAT) family is a major family of enzymes found in both
eukaryotes and prokaryotes. The members of the GNAT family are responsible for the acetylation of
a wide array of substrates, from small molecules to macromolecules. RimJ is an Nacetyltransferase, thought to transfer an acetyl group from acetyl-CoA (AcCoA) to the N-terminus of
the ribosomal protein S5 (Nα-acetylation). Other proposed functions for RimJ include the acetylation
of thymosin α1, an immunostimulating peptide, and the Z-domain protein in E. coli, as well as a role
in the ribosome assembly. Here we report the three-dimensional structure of Mycobacterium
smegmatis RimJ (MsRimJ), an ortholog of MtbRimJ, which was solved using single wavelength
anomalous dispersion. The enzyme was co-crystallized with AcCoA or CoA and the structures of
both complexes were determined to 1.9 Å and 1.7 Å, respectively. Additionally, the structure of an
apo form was solved to 1.4 Å. The MsRimJ structure displays a common GNAT fold. Based on gel
filtration experiments, MsRimJ functions as a monomer in solution, which is unusual for Nacetyltransferases.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
237
213 Biochemistry & Chemical biology
21/09/2016
Characterisation of DprE1-mediated benzothiazinone resistance in Mycobacterium
tuberculosis
C. Foo, B. Lechartier, G. Kolly, J. Neres, S. Boy-Röttger, J. Rybniker, A. Lupien, C. Sala, J. Piton,
S. Cole
EPFL, Lausanne, France
Benzothiazinones (BTZs) are a class of compounds found to be extremely potent against both drugsusceptible and drug-resistant Mycobacterium tuberculosis. The potency of BTZ is explained by
their specificity to their target decaprenylphosphoryl-D-ribose oxidase (DprE1), in particular by
covalent binding of the activated form of the compound to the critical C387 residue of the enzyme.
To probe the role of C387, we used promiscuous site-directed mutagenesis to introduce other
codons at this position in dprE1 of M. tuberculosis. The resultant viable BTZ-resistant mutants were
characterised in vitro, ex vivo and biochemically to gain insight into the effects of these mutations on
DprE1 function and on M. tuberculosis. Five different mutations at C387 (C387G, C387A, C387S,
C387N, C387T) conferred various levels of resistance to BTZ and exhibited different phenotypes.
C387G and N resulted in a slower growth rate of the mycobacterium on solid medium, which could
be attributed to the significant decrease in catalytic efficiency of the DprE1 enzyme. All five
mutations rendered the mycobacterium less cytotoxic for macrophages. Finally, differences in the
potency of covalent and non-covalent DprE1 inhibitors in the presence of C387 mutations were
revealed by enzymatic assays. As expected from the mechanism of action, the covalent inhibitor
PBTZ169 only partially inhibited the mutant DprE1 enzymes as compared to near complete
inhibition with a non-covalent DprE1 inhibitor Ty38c. This study emphasises the importance of the
C387 residue for DprE1 activity and in the killing action of covalent inhibitors such as BTZs and
other recently-identified nitro-aromatic inhibitors.
238
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
214 Biochemistry & Chemical biology
21/09/2016
Effect of doses for b6 and isoniazid under treatment of destructive tuberculosis
L. Gaevaya, L. Hayova
Department of Biological and Bioorganic chemistry, Bogomolets National Medical University, Kiev,
Ukraine
Purpose: Identify the optimal combination of drugs, the treatment of which reached at least the
residual kidney damage, assessment is made based on the results of the morphological
characteristics of the tissues of the internal organs in terms of treatment of different ratios of doses
of isoniazid and vitamin B6.
Materials and metod: Work the outcomes of destructive pulmonary tuberculosis different doses of
isoniazid and B6.
Results: Research on determination of protective action of isoniazid and B vitamins B6 respect to
the internal organs of guinea pigs under the experimental tuberculosis are intended to help find a
correlation between the dose of isoniazid and B6, which is essential to optimize treatment regimens
TB and evaluating antitoxic action of vitamin B6 in the conditions of an overdose of isoniazid.
Conclusions: The slightest manifestation of side effects on morphological characteristics, found in
cases of (10 + 50) and (32 + 5) mg / kg, which is the best result of treatment compared with
treatment doses (10 + 5) mg / kg, where side effects is more significant.
Keywords: TB of the lungs, spleen, liver, kidneys, indices of infestation, isoniazid, vitamin B6, the
optimal mathematical model
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
239
215 Biochemistry & Chemical biology
21/09/2016
Proteomic analysis of Mycobacterium tuberculosis pknG knockout mutant: insights into its
role in biological processes relevant for pathogenicity
A. Lima3, M. Gil3, A. Cascioferro1, J. Rossello3, B. Rivera3, M. Portela3-4, F. Le Chevalier1,
W. Frigui1, N. Lisa5, M. Bellinzoni5, M. Alvarez2, C. Batthyány3-2, R. Brosch1, P. Alzari5, R. Durán3
1
Integrated Mycobacterial Pathogenomics Unit, Institut Pasteur, Paris, France 2Departamento de
Bioquímica, Facultad de Medicina, UdelaR 3Analytical Biochemistry and Proteomics Unit, Institut
Pasteur de Montevideo and IIBCE 4Facultad de Ciencias, Universidad de la República,
Montevideo 5Structural Microbiology, Institut Pasteur, Paris, Uruguay
The success of Mycobacterium tuberculosis as human pathogen resides mostly in its ability to
maintain a latent infection in the host. It has been demonstrated that the Ser/Thr-kinase PknG from
M. tuberculosis regulates critical processes in bacterial physiology and pathophysiology by still
poorly understood mechanisms.
To further characterize the role of PknG in mycobacteria, we carried out interactomic and
quantitative proteomics approaches. Using a novel strategy for sequential elution of PknG partners
we obtain a list of potential kinase substrates that participate mainly in the control of nitrogen
metabolism and cell division. To validate these substrates as physiologically relevant, we performed
quantitative comparative proteomic studies of M. tuberculosis pknG knockout mutant (KO) and
H37Rv strain (WT) using 2-D Fluorescence Difference Gel Electrophoresis (2D-DIGE) strategy. 2DDIGE approach showed differences in the relative abundance of spots of the same protein but with
different pI, pattern consistent with potential changes in the phosphorylation status. We corroborate
that some of the substrates identified by the interactomic approach were indeed physiological
substrates of PknG. In addition we analyzed the global changes in the proteome of KO and WT
strains of M. tuberculosis. We identified more than one-thousand proteins for each condition using a
shotgun approach, and more than one-hundred proteins were differentially expressed between
strains. Altogether, functional analyses of differential proteins identified using both proteomic
approaches allowed us to distinguish diverse biological processes altered in the PknG deletion
mutant, including nitrogen metabolism, cell wall synthesis and complex lipid biosynthesis, processes
that are relevant for M. tuberculosis pathogenicity.
240
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
216 Biochemistry & Chemical biology
21/09/2016
Pyrazinamide resistance is caused by two distinct mechanisms: prevention of Coenzyme A
depletion and loss of virulence factor synthesis
P. Gopal1, M. Yee1, J. Sarathy1, J.L. Low1, J.P. Sarathy2, F. Kaya2, V. Dartois2, M. Gengenbacher1,
T. Dick1
1
MIcrobiology, National University of Singapore, Singapore, Singapore 2Public Health Research
Institute, New Jersey, United States
Pyrazinamide (PZA) is a critical component of first and second line treatment of tuberculosis (TB),
yet its mechanism of action largely remains an enigma. We carried out a genetic screen to isolate
Mycobacterium bovis BCG mutants resistant to pyrazinoic acid (POA), the bioactive derivative of
PZA, followed by whole genome sequencing of 26 POA resistant strains. Rather than finding
mutations in fatty acid synthase FAS I and ribosomal protein S1 / RpsA, we found resistance
conferring mutations in two pathways: missense mutations in aspartate decarboxylase panD,
involved in synthesis of the essential acyl carrier coenzyme A (CoA), and frameshift mutations in the
vitro non-essential polyketide synthase genes mas and ppsA-E, involved in the synthesis of the
virulence factor phthiocerol dimycocerosate (PDIM). The POA-resistant strains fell into different
phenotypic classes based on their resistance levels to POA, which corresponded with their
respective genotypic classes. Probing for cross resistance to two structural analogs of POA,
nicotinic acid and benzoic acid, showed that the analogs share the PDIM but not the CoA related
mechanism of action with POA. Sequencing ten POA resistant Mycobacterium tuberculosis H37Rv
isolates confirmed the presence of at least two distinct mechanisms of resistance to the drug.
Emergence of resistance through the loss of a virulence factor in vitro may explain the lack of clear
molecular patterns in PZA resistant clinical isolates, other than mutations in the prodrug converting
enzyme. The apparent interference of POA with virulence pathways may contribute to the drug’s
excellent in vivo efficacy compared to its modest in vitro potency.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
241
217 Biochemistry & Chemical biology
21/09/2016
Dare to be different - the ESX-1 protein secretion system in Staphylococcus aureus
F. Jäger, M. Zoltner, H. Kneuper, W. Hunter, T. Palmer
University of Dundee, Dundee, United Kingdom
The cell membrane represents a permeability barrier between the cell and its environment and
forms a permanently closed system. Therefore specialised transporters are necessary to allow
direct exchange of substrates and proteins between the cell cytoplasm and the external medium.
Transported proteins play essential roles in nutrient acquisition as well as in virulence. The Type
VII/ESX-1 protein secretion system was first described in Mycobacterium tuberculosis where it was
shown to secrete ESAT6 (early secreted antigen, 6kDa) and CFP10 (culture filtrate protein, 10 kDa).
Both of these proteins are important T-cell targets and essential for the virulence of M. tuberculosis.
A protein secretion system distantly related to the ESX-1 protein secretion system in M. tuberculosis
was later identified and characterized in the human and animal pathogen Staphylococcus aureus,
sharing ESAT6/CFP10-like and EssC-like components. EssC proteins are member of a family of the
FtsK/SpoIIIE family of ATPases [1], with three interlocking ATPase domains at the C-terminus. It
was recently shown that multimerisation of EccC (the EssC homolog in actinobacteria) depends
upon the binding of EsxB to a pocket on the most C-terminal ATPase domain [2].
I have been studying the four membrane proteins EssA, EssB, EssC and EsaA, of the Type VII/Ess
protein secretion system in S. aureus. While EssC is related to the actinobacterial EccC, EssA,
EssB and EsaA are essential components of the secretion machinery that are unique to firmicutes.
Crosslinking and blue native PAGE analysis have shown that the EssB, EssC and EsaA proteins
individually form homomeric complexes, but do not appear to interact with one another, or with
EssA. Surprisingly, I was able to show through crosslinking that the formation of higher molecular
weight multimers of EssC in S. aureus is not reliant on EsxB or EsxA. Therefore it seems that
oligomerisation of EssC may be controlled differently to that of EccC [3].
Bioinformatic analysis of EssC has indicated the presence of a conserved serine phosphorylation
site in the N-terminal region of the protein. My current work is focused on identifying whether EssC
is phosphorylated and if so whether this controls the activity of the type VII secretion system posttranslationally. My latest results will be presented.
1. M. J. Pallen, “The ESAT-6/WXG100 superfamily -- and a new Gram-positive secretion
system?,” Trends Microbiol., vol. 10, no. 5, pp. 209–12, May 2002.
2. O. S. Rosenberg, D. Dovala, X. Li, L. Connolly, A. Bendebury, J. Finer-Moore, J. Holton, Y.
Cheng, R. M. Stroud, and J. S. Cox, “Substrates Control Multimerization and Activation of the
Multi-Domain ATPase Motor of Type VII Secretion,” Cell, pp. 1–12, 2015.
3. F. Jäger, M. Zoltner, H. Kneuper, W. N. Hunter, and T. Palmer, “Membrane interactions and
self-association of components of the Ess/Type VII secretion system of Staphylococcus
aureus,” FEBS Lett., vol. 590, no. 3, pp. 349–357, 2016.
242
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
218 Biochemistry & Chemical biology
21/09/2016
Fast and specific detection of mycobacteria with a novel fluorogenic derivative
M. Kamariza2, C. Bertozzi1-3
1
Howard Hughes Medical Institute 2Biology Department 3Chemistry Department, Stanford
University, Stanford, United States
Tuberculosis (TB) is the leading cause of death from infectious disease worldwide. To reduce the
time-to-diagnosis of TB, better tools for the fast, sensitive detection of the causative agent M.
tuberculosis (Mtb) are urgently needed. Towards this end, we designed a fluorogenic derivative that
shows a greater than 700-fold enhancement when transitioning from aqueous to hydrophobic
environments and is biosynthetically incorporated into mycomembranes. Furthermore, the labeled
mycobacteria are detected within minutes with specificity to live mycobacteria. This combination of
properties facilitates the rapid no-wash visualization of a panel of mycobacterial and corynebacterial
species and enables the detection of live Mtb within sputum samples. The potential impact of this
work is vast and may lead to a complete modernization of the current state of TB diagnosis in high
burden regions.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
243
219 Biochemistry & Chemical biology
21/09/2016
Structural and mechanistic insight into 1-Cys peroxiredoxin AhpE from Mycobacterium
tuberculosis
A. Kumar, A. Balakrishna, W. Nartey, S. Manimekalai, G. Grüber
School of Biological Sciences, Nanyang technological University, Singapore, Singapore
Mycobacterium tuberculosis (Mtb) has the ability to persist within the human host for a long time in a
dormant stage and remerges when the immune system is compromised. One important condition,
which is responsible for dormancy, is reduced availability of oxygen. Mtb is constantly exposed to
numerous oxido-reductive stresses during its pathogenic cycle of transmission and infection.
However the unique ability of Mtb, to not only survive the redox stress manifested by host but also
to synchronize its metabolic pathway and expression of virulence factor accordingly, is central to its
survival. Under such severe conditions, Mtb employs an elaborate anti-oxidant defence mechanism
expressing a plethora of anti-oxidant proteins which includes peroxiredoxins, disulfide reductases,
superoxide dismutase etc. Among the family of Peroxiredoxins, Mtb expresses 1-cysteine
peroxiredoxin, known as alkylhydroperoxide reductase E (MtAhpE). The reduced MtAhpE (MtAhpESH) scavenges peroxides and peroxynitrite and is converted to MtAhpE-SOH. To provide
continuous availability of MtAhpE-SH, MtAhpE-SOH has to become reduced.
In the present study, we used NMR spectroscopy to delineate the reduced (MtAhpE-SH), sulphenic
(MtAhpE-SOH) and sulphinic (MtAhpE-SO2H) states of MtAhpE through cysteinyl-labelling, and
report for the first time evidence of a Mycoredoxin-independent mechanism of MtAhpE reduction.
This is confirmed by crystallographic studies, where a mycothiol-reduced- and an oxidized
conformation of MtAhpE was solved at 2.43 Å resolution. Combined with NMR-studies, the
crystallographic structures reveal conformational changes of important residues during the catalytic
cycle of MtAhpE (unpubl. data).
Reference:
1. Hugo, M. et al. Mycothiol/Mycoredoxin 1-dependent Reduction of the Peroxiredoxin AhpE
from Mycobacterium tuberculosis. J. Biol. Chem. 289, 5228-5239 (2014).
244
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
220 Biochemistry & Chemical biology
21/09/2016
Mycobacterial cell wall catabolism
A. Mery2, L. Shen2, A. Viljoen3, S. Villaume1, C. Mariller2, S. Vincent1, L. Kremer3, Y. Guerardel2
1
Université de Namur, Laboratoire de Chimie Bio-organique, Namur, Belgium 2Univ. Lille, CNRS,
UMR8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, Lille 3Mycobacterial
Pathogenesis and Novel Therapeutic Targets, CNRS-FRE3689, Montpellier, France
Arabinogalactan (AG) is a crucial element of the mycobacterial cell wall representing approximately
35 % of its components. AG is also remarkable because its sugars, D-Ara and L-Gal, are all under
furanose form but, unlike most bacterial polysaccharides, AG lacks repeating units and is instead
composed of a few distinct structural motifs. This polysaccharide has an essential function by
connecting the mycolic acid layer to the inner peptidoglycan layer to form the mycolylarabinogalactan-peptidoglycan (mAGP) complex. Because of its crucial importance in the complex
life style of mycobacterias, the biosynthesis of mAGP has always been essential for the
development of potential new drug targets. In contrast, earlier work on the existence of an
endogenous endo-D-arabinase in mycobacteria has also shown that the mycobacterial cell wall
catabolism and especially mAGP is an original and innovative route toward the discovery of new
anti-tuberculosis
drugs.
In this context, this work is aimed to prove that mycobacteria can degrade their own mAGP focusing
particularly on the enzymes capable of cleaving the arabinan and galactan portions of the AG.
By using ion chromatography coupled to mass spectrometry analysis of enzymatic products, we
developed a tool to investigate the glycosidase activities in mycobacteria. At the present moment
this methodology is used to monitor the endo-D-arabinase activity (in M. smegmatis) in order to
purify and identify it. We have also searched for putative glycoside hydrolase-coding genes by
mining the genome database and focusing focused our search on the identification of glycosyl
hydrolases that can use D-Araf and L-Galf as substrates. This second approach allowed us to
identify and express in E. coli the Rv3096 protein from M. tuberculosis. Functional assay showed
that this protein released exclusively free galactose when incubated with purified AG.
Altogether, we have successfully developed a straightforward methodology to screen for
glycosidase activities and identify enzymes. This enabled us to identify activities that degrade both
D-arabinan and L-galactan moieties of mAGP.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
245
221 Biochemistry & Chemical biology
21/09/2016
Mycobacterium tuberculosis inhibits human innate immune responses via the production
of TLR2 antagonist glycolipids
L. Blanc1, M. Gilleron1, J. Prandi1, P. Brodin3, M.S. Jang3, Y. Poquet1, O. Neyrolles1, D. Drocourt2,
G. Tiraby2, A. Vercellone1, J. Nigou1
1
Tuberculosis & Infection Biology, CNRS - Université de Toulouse 2Research Department,
Invivogen, Toulouse, France 3Institut Pasteur Korea, Seoul, South Korea
M. tuberculosis detection by human innate immune system involves different receptors called
"Pattern Recognition Receptors” (PRRs), including Toll-like Receptors (TLRs). The concerted action
of PRRs results, on one hand, in the phagocytosis of mycobacteria and, on the other hand, in the
activation of intracellular signaling pathways leading to nuclear translocation of transcription factors
(such as NF-κB and AP-1) and the expression of genes coding for cytokines and chemokines.
However, M. tuberculosis is able to inhibit innate immune responses, thus favoring its survival inside
the infected host. To identify the inhibitory mechanisms used by M. tuberculosis, we have screened
a transposition mutant library of M. tuberculosis on a human macrophage cell line THP-1,
expressing a NF-κB/AP-1 reporter system. We identified several mutants inducing a NF-κB
activation stronger than that of the wild-type strain. One of these mutants was found to be altered
for the synthesis of cell envelope glycolipids, namely sulfoglycolipids, suggesting that the latters can
interfere with innate immune responses. We further deciphered the molecular mechanisms involved
and determined that sulfoglycolipids are competitive antagonists of TLR2, thereby inhibiting the
recognition of M. tuberculosis by this receptor. Producing antagonists of PRRs is thus a strategy
used by M. tuberculosis to undermine innate immunity.
246
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
222 Biochemistry & Chemical biology
21/09/2016
ATP hydrolysis and ATPase domain motion in Mycobacterium tuberculosis DNA gyrase
G. Janvier3, C. Pissis4, L. Regad5, H. Munier-Lehmann2, S. Petrella3, C. Mayer3
1
INSERM 2Chimie Biocatalyse 3DBSC - Structural Microbiology Unit 4PF6, Institut Pasteur 5MTi,
University Paris Diderot, Paris, France
Mycobacterium tuberculosis DNA gyrase, a nanomachine involved in the regulation of DNA
topology, is the only type II topoisomerase present in this organism and hence is the sole target of
fluoroquinolones in the treatment of tuberculosis. This enzyme regulates DNA topology by creating
a double-stranded break in one DNA duplex and transporting another DNA duplex through this
break. The spread of FQ resistance represents a significant threat to public health and necessitates
the discovery of inhibitors that target DNA gyrase in novel ways. The DNA gyrase ATPase domain
provides the energy required for catalysis by ATP hydrolysis. In the present work, we discuss
several aspects of the Mtb DNA gyrase ATPase domain. On the basis of two crystal structures at
2.9 and 3.3 Å resolution and structural dynamics studies, we propose a mechanochemical scheme
to explain how ATP hydrolysis is coupled to domain motion. In this study we also validate a novel
assay for screening DNA gyrase inhibitors by the setup of an ATPase activity test. A library of 640
compounds representative of the chemical diversity of the “Chimiothèque Nationale” was screened
against the Mtb DNA gyrase ATPase domain. In the first campaign, eleven novel inhibitors were
identified but only one hit inhibits Mtb ATPase activity (> 90%). Finally, we developed
chemoinformatic-based statistical approaches to characterize the ATP-binding site in type II
topoisomerase to design a superligand.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
247
223 Biochemistry & Chemical biology
21/09/2016
Structural, functional and genetic studies of the BkaR regulon of Mycobacterium
tuberculosis
J. Pritchard1-2, N. Keep1, S. Kendall2
1
Institute of Structural Molecular Biology 2Royal Veterinary College, London, United Kingdom
The tetracycline repressor (TetR) family regulators are abundant in the Mycobacterium tuberculosis
genome and control the expression of genes involved in fatty acid metabolism, drug resistance and
virulence. Many genes in TetR regulons have been implicated in the survival of the bacteria, making
TetR regulators potential drug targets. TetR regulators bind to DNA to repress transcription and are
only removed from DNA in the presence of a ligand.
One member of the TetR family in M. tuberculosis is the branched-chain keto acid regulator (bkaR),
which is thought to control the expression of 12 genes in a regulon. The ligand for bkaR is unknown,
but the genes of the bkaR regulon have been shown to be involved in the catabolism of branchedchain amino acids (BCAA); leucine, isoleucine and valine. This regulon has been shown to be
important for the survival of the bacteria in macrophages. The physiological relevance of BCAA
catabolism in M. tuberculosis is unknown, but it is thought that utilisation may be conditional.
The overall goal of this study is to determine the physiological role of the bkaR regulon in M.
tuberculosis. We aim to use structural and biophysical approaches to characterise the regulation by
bkaR and to determine the promoter binding properties. This project also aims to identify the ligand
for the regulator and to determine the conditions under which the regulon is expressed.
248
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
224 Biochemistry & Chemical biology
21/09/2016
Siderophore monoxygenase: A potent drug target for Tuberculosis
S. Rao, J. Rao2
1
Dept of Biotechnology, Dayananda sagar College Of Engg, Bengaluru, India 2Richland college,
Dallas, United States
Iron is an essential nutrient and is typically present at concentrations too low to support proliferation
of microbial pathogens in mammalian hosts. To overcome this iron deficiency during infection many
invasive human pathogens such as Mycobacterium tuberculosis, Aspergillus fumigates, Shewanella
putrefaciens produce and secrete siderophores. The role of siderophores is to scavenge ferric iron
from the host in order for the bacteria to proliferate. Biosynthesis of these siderophores involves the
action of novel flavin –containing N-hydroxylating monoxygenases (NMO). Homologous NMOs are
present in a number of siderophore-producing organisms and disruption of these monooxygenases
in Burkholderia cepacia, and Aspergillus fumigates results in loss of effective persistence and
colonization and has been reported to be absolutely essential for virulence. Specifically, we are
targeting the enzymes that hydroxylate the siderophores to form a hydroxamate moiety, which is
essential for iron binding and the lack of homologous enzymes in humans suggest that these
enzymes are an attractive drug target for the treatment of tuberculosis and other bacterial diseases.
Putrescine monoxygenase (PMO) is a flavin –dependent monoxygenase that catalyzes the
NAD(P)H- and oxygen dependent hydroxylation of the putrescine to give N-hydroxyputrescine
involved in the biosynthetic pathway of the siderophore putrebactin is targeted. Thus, enzymes
involved in the biosynthesis of mycobactin/ putrebactin represent potential drug targets. With this
objective the cloning of pubA gene of Shewanella putrefaciens whose genomic seq is available from
Gen Bank is PCR synthesized based on the ORF sequence using genomic DNA of 4840251 bp as
template. The PCR amplicon is confirmed by sequencing and nucleotide BLAST analysis. The
amplicon is ligated wit pET Topo vector and the construct was propogated in TOP10 E coli cells and
confirmed by PCR analysis, sequencing and restriction digestion. Later transformed into E.coli BL21
cells for full length expression. IPTG induced protein expression of recombinant entry proteins fused
with 6-His and V5 epitope tags at their N termini and purified using metal affinity and ionic exchange
chromatographies. Recombinant PMO represents the first member of this class of enzymes isolated
in the active form, with a tightly bound FAD cofactor. The kcat value for formation of hydroxylated
putrescine under steady-state conditions was 8.0 min–1, and Km values of 0.5 mM for putrescine
1.8 mM for NADH, and 2.46mM for NADPH were calculated. Crystallization and drug binding
studies are in progress.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
249
225 Biochemistry & Chemical biology
21/09/2016
Metabolic labeling using azido pentoses: A novel and rapid tool to specifically target
Mycobacterium tuberculosis bacteria
K. Kolbe1-4, L. Möckl5, V. Sohst1, J. Brandenburg1, R. Engel3, S. Malm2, C. Bräuchle5, O. Holst3,
T. Lindhorst4, N. Reiling1
1
Microbial Interface Biology 2Molecular and Experimental Mycobacteriology 3Structural
Biochemistry, Research Center Borstel, Borstel 4Otto Diels Institute of Organic Chemistry,
Christiana Albertina University, Kiel 5Department of Physical Chemistry, Ludwig Maximilian
University, Munich, Germany
Tuberculosis, caused by Mycobacterium tuberculosis (Mtb) is the single leading bacterial cause of
death from infectious disease worldwide. The complex cell envelope of Mtb bacteria represents a
major virulence factor and contributes to the intrinsic difficulties to eradicate the pathogen and treat
the disease. However, the unique carbohydrate constituents of the mycobacterial cell envelope
provide the possibility to specifically target Mtb. Synthesized derivatives of selected carbohydrates
can be taken up, metabolized and subsequently be introduced into the cell envelope of Mtb
bacteria. In this study three new azido pentoses were synthesized and successfully used for
metabolic labeling of Mtb bacteria. The data strongly suggest the presence of arabinose and ribose
transporters in the Mtb cell envelope as well as unknown biosynthetic pathways for arabinose
derivatives. The investigated azido sugars exhibited no toxicity and high labeling efficiency also in
Mtb clinical isolates. Thus the newly developed metabolic method allows rapid fluorescent labeling
of different Mtb strains without time consuming genetic modifications. Metabolically labeled Mtb
bacteria were further used in infection studies with human primary macrophages, showing
comparable pathogen-specific virulence characteristics when compared to untreated bacteria. The
new azido pentoses can now be employed to identify unknown uptake and metabolic mechanisms
of Mtb bacteria and to study strain specific pathogenicity in host cells, which is strongly associated
with the clinical outcome of tuberculosis. In addition, carbohydrate derivatives may form a new and
unexpected way to specifically reach and target Mtb bacteria, which might facilitate the improvement
of tuberculosis therapy in future.
250
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
226 Biochemistry & Chemical biology
21/09/2016
Dynamics of trehalose mycolates in live mycobacteria
F. Rodriguez-Rivera2, C. Bertozzi2-1
1
Howard Hughes Medical Institute, Stanford University 2Chemistry, Stanford University, Stanford,
United States
Mycobacterium tuberculosis showcases a cell wall that serves as a formidable barrier against
targeted drugs and biological stresses. The highly conserved cell wall of mycobacteria benefits from
a mycomembrane, which has been correlated to intrinsic resistance to chemotherapeutics. Even
though extensive characterization of individual species within the plethora of glycolipids found in the
mycomembrane has been achieved, native organization and dynamics remain poorly understood.
Scarcity of tools has hampered the study of real-time mycomembrane dynamics in live bacterial
cells. Glycolipids with high degree of structural complexity and with significant roles in hostpathogen interactions remain elusive to biochemical and biophysical characterization in their native
environment. Metabolic engineering allowed rational design of unnatural trehalose reporters that
label trehalose mycolates selectively with a one-step procedure in bacterial cells. Our studies
demonstrate that trehalose glycolipids can be visualized during the course of infection in cellulo and
in the M. marinum/zebrafish model. This work illustrates the potential to identify relevant hostpathogen interactions in an in vivo tuberculosis pathogenesis model and enables visualization of the
dynamics of trehalose glycolipids with high spatial and temporal resolution.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
251
227 Biochemistry & Chemical biology
21/09/2016
Pyrazinoic acid exerts anti-mycobacterial activity via reduction of intracellular coenzyme A
level
J. Sarathy1, P. Gopal1, J. Sarathy2, F. Kaya2, V. Dartois2, T. Dick1
1
Department of microbiology and immunology, Yong Loo Lin School of Medicine, National
university of singapore, Singapore, Singapore 2Public Health Research Institute and New Jersey
Medical School, Rutgers, The State University of New Jersey, Newark, United States
Pyrazinamide (PZA) is a key drug used in the treatment of Tuberculosis (TB). Despite being in use
since the 1950s, its mode of action remains a mystery. In this study, a possible mechanism of action
was investigated: the depletion of the essential cofactor Coenzyme A (CoA). Through the use of a
fluorescence-based CoA assay and Liquid chromatography–mass spectrometry (LC-MS),
Pyrazinoic acid (POA), the bioactive form of PZA, was shown to reduce the CoA level in wild type
mycobacteria and this coincided with mycobacterial growth termination by the drug. However, POA
resistance mutations in the panD gene, a proposed POA target which functions in the CoA
biosynthesis pathway, was shown to prevent POA-induced CoA depletion. Wild type mycobacteria
supplemented with exogenous pantothenate mimicked this phenotype and from this it is
conceivable that the mechanistic basis of panD mutation-conferred resistance is that panD mutation
prevents POA-induced inhibition of the CoA biosynthesis pathway. Interestingly, POA resistance
mutations in the mas and ppsA-E genes, which are novel POA targets that function in the synthesis
of virulence factor phthiocerol dimycocerosate (PDIM), were shown to not prevent POA-induced
COA depletion. This provides further credence to POA modulating CoA levels through inhibition of
the CoA biosynthesis pathway instead via a different mechanism and it also suggests a possible
novel CoA-independent mode of action of POA where it targets the PDIM biosynthesis pathway.
Taken together, this study has helped provide a better understanding how PZA functions and this
will be crucial for the development of mechanistically-similar novel anti-TB drugs in the future.
Acknowledgements
This work was supported by the Singapore Ministry of Health’s National Medical Research Council
under its Translational Clinical Research Flagship Grant (NMRC/TCR/011-NUHS/2014) and its
Centre grant MINE / Research core no. 4 (NMRC/CG/013/2013) to A/Prof Thomas Dick, and is part
of the Singapore Programme of Research Investigating New Approaches to Treatment of
Tuberculosis (SPRINT-TB;www.sprinttb.org) led by Nick Paton. We would like to thank Sabai Phyu,
School of Medicine BSL3 core facility for support; Martin Gengenbacher, TB research laboratory, for
discussion and help, and Kristina Rutkute, SPRINT-TB, for management.
252
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
228 Biochemistry & Chemical biology
21/09/2016
Free trehalose is accumulated in dormant Mycobacterium smegmatis cells and is required
for their early resuscitation phase
M. Shleeva, G. Demina, K. Trutneva, D. Ostrovsky, A. Kaprelyants
A.N. Bach Institute of Biochemistry, Federal Research Centre “Fundamentals of Biotechnology” of
the Russian Academy of Sciences, Moscow, Russia, Moscow, Russia
The main aspect of the latent tuberculosis studies is elucidation of the molecular mechanisms
underlying M.tuberculosis dormancy in host organism.
Until now the role of trehalose in mycobacteria have been associated only with a cell wall glycolipid
known as “cord factor”. It consists of a unit of trehalose esterified to two mycolic acid residues,
which gives rise to α,α-trehalose 6,6′-dimycolates (bound form of trehalose).
In this work we show that under external gradual acidification resulted in formation of dormant
M.smegmatis cells, the content of accumulated free trehalose in dormant cells was greatly elevated.
The composition of low molecular weight compounds in cell-free extracts of dormant Mycobacterium
smegmatis cells was investigated with 1H-, 13C- and 31P-NMR spectroscopy. We found that up to
75% of total organic substances were represented by trehalose. The rest 25% contained unknown
minor components, dinucleotides, phosphomonoethers and other phosphates. In water extract of
active cells taken from early stationary phase it was detected only 15 % of trehalose.
During transition to dormant state the expression of genes involved in OtsA- OtsB and TreY- TreZ
pathways of trehalose synthesis (biosynthetic ways) was significantly increased. Gene expression of
TreS pathway (degradatory way) was negligible.
Trehalase activity was much higher in active cells compared to dormant forms although the amount
of the enzyme was approximately equal in these states of mycobacteria according to
electrophoresis data.
In the process of dormant form resuscitation the intracellular cAMP level was increased followed by
the decreasing of the free trehalose concentration during first 4-6 hours before RNA synthesis
started. The presence of trehalase inhibitor validamicin A slowed down considerably the reactivation
of dormant cells.
Thus, for the first time, we have shown that free trehalose is accumulated in significant amount in
dormant mycobacteria and participates in early molecular events leading to cell reactivation.
Consequently, the inhibitores of trehalose metabolism should possibly help to prevent the
dissemination of tuberculosis, lepra and other mycobacterial deseases by decreasing the
resuscitation of dormant pathogenic mycobacteria or even the development of dormancy.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
253
229 Biochemistry & Chemical biology
21/09/2016
A simple TLC-based method to determine the enzyme activity of two mycobacterial Nacetyl-muramyl-L-alanine amidases
N. Van Wyk3, T. Küssau3, K. Sørensen1, K. Jensen1, M. Thygesen1, M. Drancourt2, L. Kremer3-4,
M. Blaise3
1
Department of Chemistry, Faculty of Science, Centre for Carbohydrate Recognition and Signalling,
University of Copenhagen, Copenhagen, Denmark 2Unité de Recherche sur les Maladies
Infectieuses et Tropicales Emergentes, Centre National de la Recherche Scientifique (CNRS),
Institut de Recherche pour le Développement, Faculté de Médecine, Aix-Marseille Université,
Marseille 3Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé (CPBS), Centre
National de la Recherche Scientifique FRE3689, Université de Montpellier 4INSERM, CPBS,
Montpellier, France
As each bacterial replication event requires the partial hydrolysis of the cell wall peptidoglycan (PG),
screening for compounds that could inhibit the enzymes involved in the hydrolysis of PG could
prove a fruitful exercise. One set of enzymes responsible for PG hydrolysis is known as PG
amidases which includes N-acetyl-muramyl-L-alanine amidases that cleave the bond that exists
between the amino sugar moiety (N-acetyl-muramic acid) and the peptide stem of a PG subunit. To
our knowledge, the PG amidases are yet unexplored as a possible drug target. The caveat in
examining PG amidases is the lack of a suitable direct enzyme assay without the need of expensive
mass spectrometry equipment. Here, we describe a simple thin-layer chromatography method using
acetyl-muramyl-dipeptide (MDP) as the substrate for testing the activity of two mycobacterial Nacetyl-muramyl-L-alanine amidases – Rv3717 of Mycobacterium tuberculosis and Mab_0318 of M.
abscessus. Using a mobile phase consisting of ethanol, butanol and water at a ratio of 32:50:18
both the substrate and end products (acetyl-muramic acid and the dipeptide consisting of L-alanine
and isoglutamine) could be easily separated and visualized once stained with a 2% ninhydrin
solution and the TLC plate is charred. Due to the visual nature, cost-effectiveness, reproducibility
and relative ease this TLC-based assay bodes well for the future screening of compounds that
could inhibit these PG amidases.
254
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
230 Biochemistry & Chemical biology
21/09/2016
Mycobacterial Siderophore Biosynthesis Regulation through Post-Translational Acetylation
O. Vergnolle, H. Xu, J. Tufariello, L. Favrot, W.R. Jacobs, J. Blanchard
Albert Einstein College of Medicine, Bronx, United States
Mycobacterial siderophores are critical components for bacterial virulence in the host.
Mycobacterium tuberculosis (Mtb) synthesizes two related aryl-capped polyketide-polypeptide
siderophores, named mycobactin and carboxymycobactin to scavenge intracellular macrophage
iron. MbtA, the first enzyme of the mycobactin core biosynthesis catalyses two half reaction: 1)
salicylic acid activation as an acyladenylate before subsequent 2) ligation to the pantothenyl group
of the acyl carrier protein domain of MbtB. We demonstrated that MbtA can be reversibly posttranslationally acetylated by the mycobacterial protein lysine acetyltransferase (Pat, Rv0998)
leading to enzyme inhibition. MbtA acetylation can be reverted by the mycobacterial deacetylase
(DAc, Rv1151c) resulting in MbtA enzymatic reactivation. We also report the first crystal structure of
Mycobacterium smegmatis MbtA solved to 2.3 Å. Furthermore, deletion of Pat and DAc genes in
Mtb emphasize the significance of those specific two genes for normal mycobactin production
during iron starvation.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
255
231 Biochemistry & Chemical biology
21/09/2016
Clinical and genotypic profile of TB Diabetes patients at a tertiary hospital in Ghana
A. Wiredu, A. Baidoo, A. Forson
Department of Chest Diseases, Korle-bu teaching Hospital, Accra, Ghana
Background: We aimed to describe the prevalence of tuberculosis (TB) –Diabetes in Ghana and to
investigate its association with MTBC lineages.
Method: Seven hundred and sixty-seven sputum positive patients were recruited in this study The
study subjects were screened for DM and diagnoses were made on the basis of the WHO criteria
Species classification and sub lineage identification was by SNP-typing and Spoligotyping
respectively. Associations between the different phylogenetic lineages of MTBC and
epidemiological variables were assessed using Univariate and multivariate logistic regression.
Results: Six hundred and seventy nine: 476 (70.1 %) males and 203 (29.9%) females indicated
their diabetic status of which 85 (12.5%; 61 males and 24 females) were confirmed as diabetic with
HBA1C levels above 6.5mmol/L, 589, (86.8%) as non-diabetic and the remaining 5 (0.7%) as prediabetic patient. With the exception of one, all the diabetic participants were diagnosed with
pulmonary tuberculosis (TB). Seventy-seven (90.5%) patients were categorized as new cases with
6 relapse cases and 2 defaulted cases. Genotypic data showed 74 patients harboring M.
tuberculosis sensu stricto, 10 harboring M. africanum with the remaining 1 M. caprae. Four of the 74
(5.4%) had elevated blood pressure with a long history of hypertension. Nine patients (1.3%) were
co infected with HIV: 8 MTBss and 1 with Maf. All examined TB cases had abnormal chest X-ray
with the exception one particular patient showing severe cases of infiltration and presence of macro
nodules in all six zones of the chest coupled with cavitation at the right upper zone. Maf was
significantly associated with Rt upper mid and lower zone opacification (odds ratio=22.03, 95%
confidence interval (CI): 2.59-289, P<0.0014). Thirty-three patients (38.9%) presented with high
bacteria load (3+).
Conclusion: This study reports an association between diabetes and MTBC lineages in Ghana.
Given the substantial burden of DM and TB comorbidity, we recommend that TB be screened
routinely for DM to ensure the use of appropriate treatment regimen
256
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
POSTER SESSION 3
Thursday, September 22
SUMMARY
Non-tuberculous & Emerging mycobacterial pathogens ............. 259
Host-responses to mycobacteria /interaction .............................. 265
Host-Pathogen interaction and beyond ....................................... 307
232 Non-tuberculous & Emerging mycobacterial pathogens
22/09/2016
Discovery of novel anti-mycobacterial drug therapy in biofilm of pathogenic
nontuberculous mycobacterial keratitis
T. Aung2, J. Chor2, J. Yam1, Y. Liang1, R. Beuerman2
1
Singapore Centre for Environmental Life Science Engineering 2Anti-infective group, Singapore
Eye Research Institute, Singapore, Singapore
Bacterial keratitis is a leading cause of corneal opacification and contributing to the global burden of
blindness. Mycobacterial keratitis shows increasing incidence globally and is very difficult to
eradicate compared to the other common gram-positive and gram-negative infections of the cornea.
Mycobacteria easily forms biofilms in human tissues which may be a critical factor ifn sterilizing
these infections. Treatment often requires two or three different antibiotics in prolonged combination
treatment plan with accompanying surgical debridement. Thus, a new treatment strategy or
antibiotic or both would be welcomed. Our results suggest using the strong synergism of Amikacin
and Gatifloxacin for treating Mycobacterium fortuitum (ATCC 49404) while adding DNase to this
combination yielded the best efficacy in eradicating mycobacteria biofilms in vitro. In this study, we
reported the very first evidence of biofilm formation on the cornea by employing a novel neutropenic
mouse keratitis model using M. fortuitum (ATCC 49404). Our results indicated that mycobacterial
infections in keratitis were in mature biofilm modes with large amounts of extracellular DNA and
micro colony formation as evaluated by Confocal Laser Scanning Microscopy. We suggest that the
biofilm plays a critical role in this type of infection leading to the poor treatment outcomes in clinical
practice. Our results indicated that combination of Amikacin and Gatifloxacin, was more effective for
M. fortuitum (ATCC 49404) in murine keratitis model than the current standard treatment for atypical
mycobacteria, Amikacin and fourth generation fluoroquinolone, Gatifloxacin. In an in vivo murine
keratitis model, we successfully reported a novel treatment strategy, i.e., the destruction of biofilm
matrix component, extracellular DNA, increasing the efficacy of antibiotics for mycobacterial
keratitis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
259
233 Non-tuberculous & Emerging mycobacterial pathogens
22/09/2016
A new chemical entity active against Mycobacterium abscessus targets mycolic acid
transport
C. Dupont3-2, A. Viljoen3, F. Dubar1, M. Blaise3, A. Bernut3, A. Pawlik6, C. Bouchier5, R. Brosch6,
Y. Guerardel1, J. Lelièvre7, L. Ballell7, J.L. Herrmann2, C. Biot1, L. Kremer4-3
1
UMR 8576 - UGSF – Unité de Glycobiologie Structurale et Fonctionnelle, Université de Lille,
CNRS, Lille 2UMR 1173, INSERM, Université de Versailles St Quentin, Montigny Le Bretonneux
3
FRE 3689 Pathogénie mycobactérienne et nouvelles cibles thérapeutiques, CNRS, Université de
Montpellier 4FRE 3689 Pathogénie mycobactérienne et nouvelles cibles thérapeutiques, INSERM
CPBS, Montpellier 5PF1 Genopole, Institut Pasteur 6Unité de Pathogénomique Mycobacterienne
Intégrée, Institut Pasteur, Paris, France 7Diseases of the Developing World, GlaxoSmithKline Tres
Cantos, Madrid, Spain
Mycobacterium abscessus complex is an emerging pathogen responsible for severe lung infections
with parenchymal involvement especially in patients with cystic fibrosis. Highly resistant to most
antibiotics, the presence of M. abscessus is deleterious to pulmonary transplantation. Therefore,
there is an urgent need to identify new compounds active against this pathogen. M. abscessus
shares many biochemical pathways with Mycobacterium tuberculosis suggesting than antitubercular compounds may also be active against M. abscessus. Starting from a library of
compound previously validated for activity against M. tuberculosis, we identified a piperidinol-based
molecule, designated PIPD1. This compound exhibited a potent activity against a wide panel of
clinical M. abscessus strains with an MIC of 0.125 µg/ml. Moreover, PIPD1 was also found to be
active in M. abscessus-infected macrophages. In addition, infected zebrafish embryos treated with
PIPD1 displayed an increased survival rate that correlated with reduced bacterial loads. To gain
insights into the mechanism of action of PIPD1, whole genome sequencing of spontaneous M.
abscessus strains resistant to PIPD1 revealed several point mutations, all found in MAB_4508,
encoding a transmembrane protein homologous to MmpL3, a known mycolic acid transporter in M.
tuberculosis. Biochemical analyses confirmed that, while de novo biosynthesis of mycolic acids was
unaffected, PIPD1 treatment strongly inhibited the transport of trehalose monomycolate. This led to
impaired mycolylation of arabinogalactan, and to subsequent mycobacterial death. Thanks to a
predictive MAB_4508 tridimensional homology model, the mutations conferring resistance to PIPD1
were found to be clustered, defining a putative PIPD1-binding pocket.
PIPD1 represents a so far untapped class of chemical structures highly active against M. abscessus
and opens the way to future translational development opportunities by targeting transport of
mycolic acids.
260
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
234 Non-tuberculous & Emerging mycobacterial pathogens
22/09/2016
MgtC (MAB_3593), a host-induced factor and a vaccine candidate against Mycobacterium
abscessus infection
V. Le Moigne1, C. Belon2-4, C. Goulard1, G. Accard2-4, B. Pitard5, J.L. Gaillard1, L. Kremer3,
J.L. Herrmann1, A.B. Blanc-Potard2-4
1
EPIM, Université Versailles St-Quentin-en-Yvelines -INSERM U1173, Montigny-LeBretonneux 2UMR5235, CNRS 3Centre d'études d'agents Pathogènes et Biotechnologies pour la
Santé (CPBS), CNRS FRE 3689 4Laboratoire de Dynamique des Interactions Membranaires
Normales et Pathologiques, Université de Montpellier, Montpellier 5IN-CELL-ART, Nantes, France
Mycobacterium abscessus is an emerging pathogenic mycobacterium involved in pulmonary and
cutaneo-mucous infections, which constitutes a serious threat for cystic fibrosis (CF) patients. The
lack of an efficient treatment regimen and the emergence of multi-drug resistance in clinical isolates
require the development of new therapeutic strategies against this pathogen. Reverse genetics
revealed genes present in M. abscessus but absent from saprophytic mycobacteria, and potentially
involved in pathogenicity. Among them, MAB_3593 encodes MgtC, a known virulence factor
involved in intramacrophage survival and adaptation to Mg2+ deprivation in several major bacterial
pathogens. Herein, we first investigated the contribution of MgtC in M. abscessus virulence in
infected macrophages. Despite a strong induction of M. abscessus MgtC at both transcriptional and
translational levels when bacteria reside inside macrophages or upon Mg2+ deprivation, an M.
abscessus knock-out mgtC mutant exhibited a slight, but non significant, replication defect within
macrophages. However, our results indicated that inhibition of MgtC in vivo through immunization
with M. abscessus mgtC DNA, formulated with a tetra functional amphiphilic block copolymer,
exerted a protective effect against an aerosolized M. abscessus challenge in CF (ΔF508 FVB) mice.
The formulated DNA immunization was associated with the production of MgtC specific antibodies,
which may stimulate a protective effect by counteracting MgtC during M. abscessus infection. Our
results, thus, emphasize the importance of M. abscessus MgtC in vivo, and provide a basis for the
development of novel therapeutic tools against M. abscessus infections in CF patients.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
261
235 Non-tuberculous & Emerging mycobacterial pathogens
22/09/2016
The distinct fate of smooth and rough mycobacterium abscessus variants inside
macrophages
A. Roux3, A. Viljoen1, A. Bah6, R. Simeone5, A. Bernut4, T. Deramaudt2, M. Rottman3, J.L. Gaillard3,
L. Majlessi5, R. Brosch5, I. Vergne6, C. De Chastellier1, L. Kremer4, J.L. Herrmann3
1
Centre d'Immunologie de Marseille-Luminy, Marseille 2UMR1179, Montigny Le Bretonneux 3UMR
1173, Montigny-Le-Bretonneux 4Centre d’études d’agents Pathogènes et Biotechnologies pour la
Santé, Montpellier 5Unité de Pathogénomique mycobactérienne, Paris 6IPBS, Toulouse, France
Mycobacterium abscessus is a pathogenic rapid growing mycobacterium responsible for pulmonary
and cutaneo-mucous infections in immunocompetent patients and in patients with cystic fibrosis. It
transitions from a smooth (S) morphotype with cell surface-associated glycopeptidolipids (GPL) to a
rough (R) morphotype lacking GPL. Herein, we show that the M. abscessus S and R variants are
able to grow inside macrophages and are present in morphologically distinct phagosomes. The S
variantsare usually found as single bacteria within phagosomes characterized by a tightly apposed
phagosomal membrane and the presence of an electron translucent zone (ETZ) surrounding the
bacilli. Conversely, infection with the R variantleads to phagosomes harbouring more multiplebacilli,
surrounded by a loose phagosomal membrane and lacking the ETZ. In contrast to the R variant, the
S variant is capable to restrict the intraphagosomal acidification of macrophages and limits
apoptosis and autophagy. Unexpectedly, the phagosomal membrane enclosing the S forms showed
signs of alteration such as breaks or partial lysis. Although such events were not frequently
encountered, they suggest that the S form is capable to maintain phagosome-cytosol
communications. This view support M. abscessus S as sharing several traits relevant to slowgrowing mycobacterial species with respect to pathogenicity and virulence.
262
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
236 Non-tuberculous & Emerging mycobacterial pathogens
22/09/2016
Association of genetic markers with the pattern of antibiotic resistance of Mycobacterium
avium subsp. hominissuis clinical isolates in South Korea
S.Y. Kim1, W.J. Koh1, S.J. Shin2
1
Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine
2
Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
The aim of this study was to genetically characterize clinical isolates from patients diagnosed with
Mycobacterium avium lung disease and to investigate the clinical significance. Multi-locus
sequencing analysis (MLSA) and pattern of insertion sequence analysis of M. avium isolates from
92 Korean patients revealed that all isolates were M. avium subspecies hominissuis. In hsp65
sequevar analysis, codes 2, 15, and 16 were most frequently found (88/92) with similar proportions
among cases additionally two isolates belonging to code N2 and an unreported code were
identified, respectively. In insertion element analysis, all isolates were IS1311 positive and IS900
negative. Four of the M. avium subsp. hominissuis isolates did not harbor IS1245 and 1 of the M.
avium isolates intriguingly harbored DT1, which is thought to be a M. intracellulare-specific element.
M. avium subsp. hominissuis harboring ISMav6 are prevalent in Korea. No significant association
between clinical manifestation and treatment response in patients with hsp65 code type and ISMav6
has been found, indicating that any specific strain/genotype among M. avium subsp. hominissuis
organisms was not a major source causing M. avium lung disease. Interestingly, the presence of
ISMav6 was significantly related to trends of more resistance to moxifloxacin. Conclusively, the
genotype of Korean M. avium subsp. hominissuis isolates is not a disease determinant responsible
for lung disease and further investigation of specific virulent factors of M. avium subsp. hominissuis
needs to be investigated further.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
263
237 Non-tuberculous & Emerging mycobacterial pathogens
22/09/2016
MAB_3551c encodes the primary triacylglycerol synthase involved in lipid accumulation in
Mycobacterium abscessus
A. Viljoen2-1, M. Blaise2, C. De Chastellier1, L. Kremer2-3
1
Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS
UMR7280, Marseille 2Centre d'études d'agents Pathogènes et Biotechnologies pour la
Santé 3INSERM, CNRS FRE3689, Montpellier, France
Mycobacterium abscessus (Mabs) is a fast-growing mycobacterial species and an emerging
infective agent. The presence of seven genes encoding putative triacylglycerol (TAG) synthases
(Tgs) in Mabs, suggests that during infection this mycobacterial species may accumulate TAG as a
source of carbon and energy in the form of intracellular lipid inclusions (ILI), which could be used for
long term survival within patients suffering from a chronic Mabs infection.
Targeted gene deletion and gene-complementation studies were used to investigate the importance
of the seven putative Tgs-encoding genes in TAG production in Mabs. Analysis of the TAG content
in the different strains was carried out by lipid extraction followed by thin layer chromatography
analysis. The most important TAG-producing Tgs’s identified by the complementation studies were
purified, allowing characterization of their substrate preference. Residues critical to Tgs activity were
identified by site-directed mutagenesis. Subsequently, a well-defined foamy macrophage (FM)
model was used for quantitative electron microscopy to monitor and quantify ILI formation in Mabs.
Deletion of MAB_3551c (tgs1) resulted in a pronounced decrease of TAG production in Mabs.
However, complementation studies of this mutant with tgs1 to tgs7 revealed that Tgs4 also
expressed considerable Tgs activity. Recombinant Tgs1 and Tgs4 were purified and among the
two, Tgs1 exhibited the greatest TGS activity in the presence of diacylglycerol and acyl-CoA.
Subsequently, H144 and Q145 of the putative acyltransferase domain were identified to be critical
to the enzymatic activity. Importantly, when macrophages were infected with Mabs and stimulated to
become foamy, Mabs accumulated TAG in the form of ILI. In addition, the tgs1 deletion mutant
produced less and smaller ILI within FM than the parental strain, while over-expression of Tgs1
resulted in increased ILI size.
We demonstrate, for the first time, ILI accumulation in Mabs inside FM. We propose Tgs1 as the
major factor in this phenotype and provide novel biochemical features of this family of enzymes in
mycobacteria, including substrate preferences for long acyl-CoA chains and residues critical to its
activity. Future developments of in vivo animal models are required to address the importance of
tgs1 in survival and persistence of Mabs during latent infection.
264
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
238 Host-responses to mycobacteria /interaction
22/09/2016
Profiling the early immune response of macrophages to Mycobacterium tuberculosis
infection using RNA-seq
N. Andreu2, J. Phelan2, P.F. De Sessions1, T.G. Clark2, M.L. Hibberd2-1
1
Genome Institute of Singapore, A*STAR, Singapore, Singapore 2London School of Hygiene and
Tropical Medicine, London, United Kingdom
Tuberculosis disease kills 1.5 million people every year. Latent Mycobacterium tuberculosis
infection persists within an estimated third of the world’s population, and 10% lead to active
tuberculosis. Understanding how the host immune system is able to contain or even eradicate the
infection in the remaining 90% of the infected cases is crucial to the development of new therapies
and vaccines aimed at boosting these defence mechanisms. Macrophages play an essential role in
the early immune response against M. tuberculosis and are the cell type preferentially infected in
vivo. In vitro, naïve macrophages are adept at killing at least some bacilli during the first two days of
infection, although this ability wanes with time. To explore the early response of macrophages to M.
tuberculosis infection we have characterised their transcriptomes using RNA-seq. In particular, we
define the transcriptomes of macrophages infected with live and dead M. tuberculosis at 4 and 24
hours post-infection. Using this approach, we have detected many differentially expressed genes
and pathways previously described as important during M. tuberculosis infection, confirming the
ability of this approach to detect relevant processes. In addition, we have also identified novel genes
whose function in resistance to M. tuberculosis infection has not been investigated. The results
presented here detail the immune response of macrophages to M. tuberculosis infection at greater
depth and confidence than previously achieved.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
265
239 Host-responses to mycobacteria /interaction
22/09/2016
Impact of helminth infection on antimycobacterial immune responses in UK migrants
S. Anwar1, H. Fletcher1, M. Brown2, H.M. Dockrell1, F. Toulza1
1
Infection and Immunity, London School of Hygiene and Tropical Medicine 2University College of
London, London, United Kingdom
Latent tuberculosis and helminth infections are co-endemic in many parts of the world. Chronic
helminth infections can modulate the immune responses against Mycobacterium tuberculosis (Mtb).
The consequent immunomodulation may affect the ability of the host to control the growth or killing
of Mtb. This study aims at investigating the modulations of the immune response profile of latent
tuberculosis (LTBI) and helminth co-infected patients and whether these modulations are
associated with a decrease in mycobacterial growth inhibition using a mycobacterial growth
inhibition assay (MGIA). UK migrants attending University College Hospital London, UK with or
without diagnosed helminth infection (Strongyloides spp and Schistosoma spp) and/or latent TB
were bled at recruitment and 4 months after completing anthelmintic treatment. PPD specific IFN-γ
responses in PBMC are tested by ex-vivo ELISpot assay and flow cytometry. We have previously
shown the frequency of CD4+IFNγ+T cells following PPD stimulation is lower in helminth and latent
TB coinfected UK migrants, compared to LTBI only, while antihelminth treatment resulted in a
reduction in the frequency of CD4+FoxP3+T cells and increased Mtb specific CD4+IFNγ+ T cell
responses (Toulza et al, Eur J Imm 2016). Helminth infected patients showed reduced
mycobacterial growth inhibition compared to healthy controls which improved after anthelmintic
treatment. Modulations of cytokine profile were also observed in these patients following
treatment. Mycobacterial growth inhibition and multiple immune parameters simultaneously may
provide immune signatures indicative of protective immunity against Mycobacterium tuberculosis, as
well
as
modulation
following
treatment
of
helminth
co-infected
patients.
Acknowledgement: Funded by EU consortia; TBVAC2020; # 643381, IDEA; # 241642 and
Commonwealth Scholarship Commission, UK.
266
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
240 Host-responses to mycobacteria /interaction
22/09/2016
Complement activation by Mycobacterium tuberculosis is phenotype-dependent
T. Keating3-1, S. Thomas2, L. Alderwick1, S. Taylor2, J. Bacon3
1
School of Biosciences, University of Birmingham, Birmingham 2Pathogen Immunology Group 3TB
Research, Public Health England, National Infection Service, Salisbury, United Kingdom
Pathogenesis of pulmonary TB is highly complex, with the measurement of cellular responses often
used to assess protection in human and animal models. Compared to other major bacterial
pathogens, relatively little is known about the nature of Mycobacterium tuberculosis-Complement
interactions. The deposition of C3b on bacterial surfaces is thought to be beneficial to the pathogen
by enhancing uptake by alveolar macrophages, an environment within which M. tuberculosis is
adapted to survive. The M. tuberculosis cell wall is rich in mannose-containing structures and these
have been shown to be important for complement activation with MBL levels and have been linked
with susceptibility to infection in humans. We have investigated how bacterial phenotype,
specifically biofilm and planktonic growth, alters complement pathway activation.
Using IgG-depleted human plasma as the complement source, significantly enhanced deposition of
C3b and C5b-9 was detected on the surface of planktonic M. tuberculosis, compared to cells from
pellicle biofilms or planktonic cells from the non-pathogenic Mycobacterium smegmatis. This was
seen at 10% complement levels and at levels physiologically relevant to the lung where the
alternative pathway is less active (2%). Use of a blocking monoclonal to C1q resulted in reductions
in C3b deposition on both phenotypes, showing direct activation of the classical pathway, and this
was more reduced in biofilms. At 2% complement, C1q blocking completely abrogated C3b
deposition on M. smegmatis. Direct measurement of C1q and MBL deposition on bacterial surfaces
showed higher levels on planktonic compared to biofilm M. tuberculosis cells, with no MBL
detectable on M. smegmatis.
Biochemical analyses of planktonic and biofilm cells showed altered proportions of glucose in cell
wall associated alpha-glucans of the biofilm cells. Carbohydrate extracts are being used in a
complement C3b activation ELISA determine whether these alpha-glucan changes are leading to
reduced C3b generation on the biofilm-derived polysaccharide.
Currently IFN-y is used as an indicator of vaccine protection in TB. However, this has failed to
predict protection of a vaccine in recent clinical trials. A greater understanding of the interplay
between antibody, complement, and T cell responses could lead to additional correlates that span
both the humoral and cell-mediated responses.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
267
241 Host-responses to mycobacteria /interaction
22/09/2016
Mycobacterium abscessus-induced granuloma formation is strictly dependent on TNF
signaling and neutrophil trafficking
A. Bernut2, J.L. Herrmann1, G. Lutfalla3, L. Kremer2-4
1
UMR1173, Université de Versailles St Quentin, INSERM, Montigny Le Bretonneux 2CPBS,
FR3689 3DIMNP, UMR5235, CNRS 4CPBS, FR3689, INSERM, Montpellier, France
Mycobacterium abscessus (Mabs) is considered the most common respiratory pathogen among the
rapidly growing non-tuberculous mycobacteria. Infections with Mabs are increasingly found in
chronic lung diseases, especially with cystic fibrosis, and they are often refractory to antibiotic
therapy. Mabs has two morphotypes with distinct effects on host cells and biological responses. The
smooth (S) variant is recognized as the initial airway colonizer while the rough (R) is known to be a
potent inflammatory inducer associated with invasive disease, but the underlying
immunopathological mechanisms of the infection remain unsolved.
We conducted a comparative stepwise dissection of the inflammatory response in S and R
pathogenesis by monitoring transparent infected zebrafish embryos. Quantitative RT-PCR, reporter
zebrafish lines and antisense technology (morpholinos) were used to determine the contribution of
the innate immune system and the role of inflammation in Mabs infection.
An intense TNFα and IL8 pro-inflammatory response was measured by qRT-PCR. Through the use
of mpeg1:mCherry or mpx:eGFP transgenic zebrafish lines harboring fluorescent macrophages and
neutrophils, we found that, like macrophages, neutrophils interacted with Mabs at the initial site of
infection. Loss of IL8 function resulted in increased mortality with both variants and was associated
with impaired of neutrophils mobilization to the infection foci. Next, the role of TNFα in the control of
Mabs infection was subsequently investigated using TNFR1 morpholinos. TNFR1 morphants were
found to be extremely susceptible to both Mabs infections. Impaired TNF signaling disrupted the
IL8-dependent neutrophil mobilization and the defect in neutrophil trafficking led to the formation of
aberrant granulomas, extensive mycobacterial cording, unrestricted extracellular bacterial growth
and subsequent larval death. The role of neutrophils for the granuloma formation was confirmed in
IL8- or in neutrophil-depleted embryos.
These results indicate that the TNF/IL8 inflammatory axis provides a protective sanctuary for Mabs.
Our unanticipated findings indicate that neutrophils are critical for the establishment and
maintenance of Mabs granulomas and play an important protective role against Mabs. These
observations are particularly relevant to infections in cystic fibrosis patients, which are characterized
by severe inflammation dominated by neutrophil influx.
268
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
242 Host-responses to mycobacteria /interaction
22/09/2016
Human Xenobiotic Nuclear Receptor PXR Augments Mycobacterium tuberculosis Survival.
E. Bhagyaraj, R. Nanduri1, A. Saini1, H. Kitdorlang Dkhar1, N. Ahuja1, V. Chandra1, S. Mahajan1,
R. Kalra1, D. Tiwari1, C. Sharma1, A. Janmeja2, P. Gupta1
1
IMTECH, CSIR 2Pulmonary medicine, Government Medical College and Hospital, Chandigarh,
India
The current challenge in tuberculosis (TB) treatment is the rapid increase in the emergence of drug
resistant TB and this necessitates alternative therapeutic strategies that are less likely to induce
drug resistance. Identification and targeting of host factors that could modulate the outcome of M.
tuberculosis infection is a promising strategy. Human nuclear receptor Pregnane X receptor (hPXR)
plays a crucial role in drug metabolism, drug resistance, energy homeostasis and immune
response. So far its role in M. tuberculosis infection has not been explored. In this study, we
investigated hPXR role in M. tuberculosis infection in human monocyte derived macrophages
(hMDMs) generated from human peripheral blood mononuclear cells (PBMCs) isolated from healthy
donors. Here, we demonstrate that PXR augments M. tuberculosis survival inside the host
macrophages by promoting the foamy macrophage (FM) formation and abrogating phagolysosomal
fusion, inflammation and apoptosis. Additionally, M. tuberculosis cell wall lipids, particularly mycolic
acids (MAs) crosstalk with hPXR by interacting with its promiscuous ligand binding domain. To
confirm our in vitro findings and to avoid the reported species barrier in PXR function, we adopted
an in vivo mouse model expressing hPXR wherein expression of hPXR in mice promotes M.
tuberculosis survival. Therefore, pharmacological intervention and designing antagonists to hPXR
may prove to be a promising adjunct therapy for TB.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
269
243 Host-responses to mycobacteria /interaction
22/09/2016
The Wnt ligand Wnt6 promotes the survival of Mycobacterium tuberculosis in macrophages
by modulating host cell metabolism
J. Brandenburg1, D. Schwudke1, T. Goldmann1, S. Marwitz1, M. Leitges3, A. Kispert2, N. Reiling1
1
Research Center Borstel, Leibniz Center for Medicine and Biosciences, Borstel 2Institut für
Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany 3The Biotechnology
Centre of Oslo, University of Oslo, Oslo, Norway
The Wnt signaling pathway, an ancient and highly conserved signaling network, controls embryonic
development and tissue homeostasis and has also been shown to exert immuno-regulatory
functions during inflammatory and infectious diseases including tuberculosis. We recently showed
that the Wnt ligand Wnt6 is expressed in the lung of Mycobacterium tuberculosis (Mtb)-infected
mice and demonstrated that Mtb-induced Wnt6 shifts macrophage polarization towards a M2-like
phenotype. Here, the functional role of Wnt6 and downstream factors during Mtb infection was
analyzed in primary murine and human macrophages by use of a variety of methods, ranging from
gene expression to mass spectrometry-based lipidome and CFU analyses. We were able to show
that Wnt6 expression promotes the survival of Mtb within its main host cell by targeting cellular
metabolism.
270
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
244 Host-responses to mycobacteria /interaction
22/09/2016
Th22 cells in mycobacterial immunity and the effect of HIV co-infection
R. Bunjun2, C. Riou2, F. Omondi2, R. Wilkinson1-4-3, W. Burgers2
1
Clinical Infectious Diseases Research Initiative 2Division of Virology, Institute of Infectious Disease
and Molecular Medicine, Cape Town, South Africa 3Department of Medicine, Imperial College
London 4Mill Hill Laboratory, The Francis Crick Institute, London, United Kingdom
HIV-1 infected individuals are at greater risk of developing tuberculosis (TB), irrespective of CD4
count. We investigated mycobacterial CD4+ T cell responses during early HIV infection, to identify
defects present prior to profound CD4 depletion.
T cell immunity was measured in the blood of healthy persons with evidence of immune
sensitisation to M. tuberculosis (Mtb) who were either HIV-1-infected (median CD4 count 619
cells/mm3, ART naïve) or uninfected (n=24 in each group). Whole blood was stimulated with BCG
and Mtb PPD and analysed for cytokine production using multiparameter flow cytometry.
Characterisation of CD4+ T cell responses in HIV-1-uninfected persons after BCG stimulation
revealed a high frequency of IL-22-producing CD4+ cells, that was significantly greater than the
magnitude of the IFN-g response (median 0.91% versus 0.55% of CD4+ T cells, respectively;
p=0.024). By comparison IL-17 responses were of low frequency (median 0.11%). There was a
positive correlation between the IFN-γ and IL-22 response to BCG (r=0.83, p<0.0001), suggesting
co-expression of these cytokines. However, the majority (80%) of IL-22+ cells did not co-express
either IFN-γ or IL-17, suggesting rather that these cells are a distinct ‘Th22’ subset. Further
characterisation of these Th22 cells revealed a memory differentiation phenotype similar to Mtbspecific Th1 cells, with 80% of cells displaying an early-differentiated phenotype (CD45RO+CD27+)
and 20% of cells being late-differentiated (CD45RO+CD27-). Like Th17 cells, mycobacteriaresponsive Th22 cells expressed the chemokine receptor CCR6 (median 96% of cells producing IL22 alone), and had an intermediate phenotype between Th1 and Th17 cells for CXCR3 (28%),
CCR4 (26%) and CCR10 (3%) expression. Interestingly, in HIV-1 infected individuals the IL-22+
response was 3-fold lower than in the uninfected group (p=0.0007), despite well preserved CD4
counts. This decrease was similar to the depletion of IFN-g+ CD4+ T cells in the same individuals
during HIV-1 infection (p=0.0006).
Thus, Th22 T cells make up a substantial and under-recognised part of the mycobacterial response,
that together with the Th1 response is significantly decreased even in early HIV-1 infection. These
findings highlight the effect of HIV on multiple immune subsets that may be important for control of
Mtb.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
271
245 Host-responses to mycobacteria /interaction
22/09/2016
Acid sphingomyelinase is required for immune control of Mycobacterium tuberculosis
infection
M. Burmeister1, N. Reiling2, D. Adam3, U. Schaible1
1
Cellular Microbiology 2Microbial Interface Biology, Research Center Borstel, Borstel 3Institute for
Immunology, Christian-Albrechts-University, Kiel, Germany
Dysfunctional lysosomes interfering with phagocyte functions can alter host defense, antigen
processing and downstream immune responses. The etiological agent of tuberculosis (Tb),
Mycobacterium tuberculosis (Mtb), manipulates the macrophage's phagosomal system to escape
elimination and modulate inflammation. The lysosomal acid sphingomyelinase (aSMase) generates
biologically active ceramide species, which modulate autophagy, vesicle formation and apoptosis,
thereby linking lipid mediator signalling, inflammation and membrane trafficking.
Here, we analyze the functions of aSMase in lipid signalling and vesicle trafficking during Mtb
infection. The murine Tb model of aerosol infection revealed an increased mycobacterial burden in
lung, spleen and liver of aSMase KO vs. wild type (WT) mice. Furthermore, Mtb-infected aSMase
KO mice showed enhanced systemic as well as pulmonary production of the proinflammatory
cytokines MCP-1 and TNF-α and an exacerbated histopathology characterized by lipid storing
foamy macrophages and multinucleated giant cells. We also observed alterations in immune cell
recruitment with less CD4+ T cells, DCs and NK T cells in lungs of KO mice, while there was an
increase in macrophages and CD8+ T cells. Currently, we analyze whether increased susceptibility
to Mtb is due to a loss of macrophage function associated with intracellular lipid accumulation,
which promotes Mtb growth or either hampers TH1 and NK T cell recruitment or antigen
presentation / T cell activation.
Understanding the function of aSMase in Mtb infection may identify signalling pathways and target
structures for host directed therapeutic (HDT) strategies, which can accompany classical antimycobacterial treatment.
272
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
246 Host-responses to mycobacteria /interaction
22/09/2016
Mycobacterium tuberculosis RipA induces a prominent production of IL-10 in dendritic
cells though activating TLR4-ROS-MAPK signaling
H. Choi, J.S. Kim, H. Kim, W.S. Kim, S.J. Shin
Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21
PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
Currently the biggest problem for tuberculosis (TB) control worldwide is preventing reactivation of
latent TB. In this context, we hypothesized that some Mycobacterium tuberculosis (Mtb) antigens
could be involved in the reactivation process of TB by altering the immune status, especially downregulating immune signaling and immune cell function towards a pathogen-favoring environment.
RipA, a key enzyme in Mtb cell division, may be actively involved in this process by interacting with
various immune cells playing an important role in the regulation of T cell immunity. In this study, we
investigated how RipA manipulates the host immune system toward pathogen-favored conditions by
interacting with dendritic cells (DCs). RipA-treated DCs significantly up-regulated the expression of
cell surface molecules (CD80, CD86, MHC class I and II) along with remarkable secretions of proinflammatory cytokines, IL-6, IL-1β, and TNF-α, in a dose-dependent manner. Interestingly, unlike
LPS, RipA significantly produced IL-10 in DCs through TLR4 binding and subsequent activation of
the p38 MAPK and NF-κB signaling pathways. Furthermore, we found that intracellular reactive
oxygen species (ROS) is required for the p38 MAPK activation and for RipA-induced IL-10
generation in DCs. Collectively, our results suggest that RipA induces a prominent production of IL10 through TRL4-dependent ROS and p38 MAPK signaling pathways and may contribute to the
reactivation process by regulating host immune response.
This study was supported by grants from the Basic Science Research Program through the Ministry
of Science, ICT, and Future Planning (NRF-2014R1A2A1A11053330).
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
273
247 Host-responses to mycobacteria /interaction
22/09/2016
RNA-seq transcriptional profiling of PPD-b-stimulated peripheral blood from cattle infected
with Mycobacterium bovis
C. Correia3, K. Mcloughlin3, N. Nalpas3-1, D. Magee3, J.A. Browne3, K. Killick3, H. Vordermeier5,
B. Villarreal-Ramos5, S. Gordon4-2, D. Machugh3-2
1
Proteome Center Tübingen, University Of Tübingen, Tübingen, Germany 2UCD Conway Institute
of Biomolecular and Biomedical Research 3UCD School of Agriculture and Food Science, Animal
Genomics Laboratory 4UCD School of Veterinary Medicine, University College Dublin, Dublin,
Ireland 5Animal and Plant Health Agency, Weybridge, United Kingdom
Mycobacterium bovis infection, the cause of bovine tuberculosis (BTB), costs an estimated $3 billion
to global agriculture annually. The impacts of M. bovis infection are manifold, including risks to
animal and public health, disruptions to trade, and reduced agricultural productivity. During the last
decade, the maturation of high-throughput sequencing technologies coupled with well-annotated
genome resources, has provided an unprecedented opportunity to gain a deeper understanding of
host-pathogen interactions for many infectious diseases. Within this context, transcriptional profiling
of the host immune response to M. bovis infection is a powerful approach for identifying host genes
and cellular pathways important to disease pathology and for biomarker development. For the
present study, ten age-matched male Holstein-Friesian calves were infected endobronchially with
M. bovis (~2,000 CFU). Peripheral blood samples were collected in duplicate at four time points (-1
wk pre-infection, +1 wk, +2 wk, and +10 wk post-infection) and used for a) an overnight stimulation
with purified protein derivative of bovine tuberculin (PPD-b) at 37°C or b) a control overnight
incubation at 37°C without PPD-b stimulation. After isolation of total RNA, poly(A)+ purified RNA
was used to generate strand-specific RNA-seq libraries for high-throughput sequencing (HTS).
Sequencing reads were quality checked, adapter and quality filtered, and then aligned to the Bos
taurus reference genome UMD3.1.1. Following summarisation of gene counts, lowly expressed
transcripts were removed prior to subsequent gene annotation and differential expression analyses.
This computational workflow revealed 929 differentially expressed (DE) genes at -1 wk pre-infection,
1,619 DE genes +1 wk post-infection, 1,170 DE genes at +2 wk, and 5,535 DE genes at +10 wk
(when compared to the non-PPDb-stimulated group at each time point; FDR correction threshold ≤
0.05). DE gene lists were used for functional enrichment with Ingenuity® Systems Pathway Analysis
(IPA) to identify canonical cellular pathways perturbed across the infection time course in peripheral
blood due to PPD-b stimulation.
274
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
248 Host-responses to mycobacteria /interaction
22/09/2016
Mycobacterium tuberculosis induces a quiescent bioenergetic phenotype in infected
macrophages through irreversible suppression of oxidative phosphorylation
B. Cumming1, K. Addicott1, A.J.C. Steyn1-2
1
KwaZulu-Natal Research Institute for Tuberculosis and HIV, Durban, South Africa 2Department of
Microbiology, University of Alabama at Birmingham, Birmingham, United States
Immunometabolism has become increasingly important in the development of new host-directed
therapies against human diseases such as atherosclerosis, diabetes and some cancers. About one
third of the world’s population is latently infected with Mycobacterium tuberculosis (Mtb) and
infection with Mtb does not confer immunity against future infections demonstrating Mtb has
definitive evasion strategies to manipulate the immune response and prevent complete eradication.
To explore the use of immunometabolism as a potential target for adjunctive host-directed therapy
in the treatment of tuberculosis, we investigated the effects of pathogenic Mtb and non-pathogenic
M. bovis BCG on the energy metabolism of the infected macrophage. Using extracellular flux
analysis, Mtb infection was shown to decrease the rates of both ATP-generating pathways,
oxidative phosphorylation (OXPHOS) and glycolysis, in macrophages, shifting the bioenergetic
phenotype of the host cell to one synonymous with a state of quiescence. In contrast, macrophages
infected with M. bovis BCG maintained OXPHOS and glycolysis at levels similar to those in
uninfected macrophages. Heat-killed Mtb decreased OXPHOS rates of macrophages comparable to
that of viable Mtb infections, but maintained the glycolytic rate present in uninfected macrophages.
Pharmacological killing of intracellular Mtb by treatment with combinations of rifampicin and
isoniazid, or bedaquiline and clofazimine, increased the rates of glycolysis to levels observed in
uninfected macrophages, however it did not improve host OXPHOS. Thus, the presence of dead
Mtb in the macrophages still suppresses OXPHOS, which suggests that the infected cells have a
greater dependency on glycolysis for their ATP requirements. This was confirmed by treatment of
Mtb infected macrophages with 2-deoxyglucose, which inhibits the enzyme catalysing the first step
in glycolysis, namely hexokinase. Unlike the therapeutic effects of 2-deoxyglucose in cancers using
aerobic glycolysis for ATP generation, 2-deoxyglucose did not reverse the bioenergetic phenotype
of Mtb infected macrophages. Metformin, an AMPK-agonist that has been shown to inhibit
gluconeogenesis and promote OXPHOS in diabetics, did not restore OXPHOS in Mtb infected cells.
Going forward, we plan to discover the mechanisms whereby Mtb subdues host OXPHOS; if this
has detrimental effects on macrophage mitochondria and whether or not this suppression can be
pharmacologically reversed or inhibited.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
275
249 Host-responses to mycobacteria /interaction
22/09/2016
Calcimycin inhibits intracellular mycobacterial growth by regulating autophagy
S. Mawatwal3, S. Kidwai2, A. Kapuria3, A. Singh1, S. Agarwal2, R. Singh2, R. Dhiman3
1
Structural Biology Laboratory, G. N. Ramachandran Protein Centre, CSIR-Institute of Microbial
Technology, Chandigarh 2Vaccine and Infectious Disease Research Centre, Translational Health
Science and Technology Institute, Faridabad, Haryana 3Department of Life Science, National
Institute of Technology, Rourkela, India
Mycobacterium tuberculosis (M. tb), the causative agent of tuberculosis resides and multiplies in
macrophages by inhibiting various host defensive pathways like autophagy. Increased autophagy
by different inducers like adenosine triphosphate (ATP) has been shown to be antimycobacterial
through calcium-dependent mechanism that decreased M. bovis BCG viability. Our preliminary
screening experiment using whole cell based approach has identified Calcimycin, which increases
intracellular calcium as a potent inhibitor of M. tb growth. We observed that Calcimycin was
bactericidal in action and inhibited M. tb growth in vitro by 99% at 1.25 M. So, present study was
undertaken to decipher the role of Calcimycin on intracellular mycobacteria in THP-1 cells. We
found by MTT assay that 0.4 µM of calcimycin is non-toxic even after 72 h of treatment. Viability
findings were further corroborated by Trypan blue dye exclusion assay where calcimycin treatment
for 72 h reduced cell viability to 86.4 ± 1.8%, 73.0 ± 11.9% and 62.2 ± 10.2% at 0.6 µM, 0.8 µM and
1µM respectively. Time kinetic experiment was performed with 0.4 µM of calcimycin to study its
effect on the autophagy in THP-1 cells. We found optimum up-regulation of different autophagy
markers like Beclin-1, Atg 7 and Atg 3 after 12 h of treatment through western blotting. Increased
conversion of LC3-I to LC3-II, widely used marker of autophagy induction was also observed in
treated cells suggesting increased autophagy. Addition of 3-methyadenine (3-MA), an autophagy
inhibitor, abrogated the effect of calcimycin on autophagy. We found down-regulation of Beclin-1,
Atg 7, Atg 3 and LC3-II expression in calcimycin treated cells in the presence of 3-MA. Increased
autophagy in calcimycin treated cells led to decrease in intracellular M. bovis BCG viability and
addition of 3-MA reversed this effect. Studies are currently underway to determine the mechanism
of how calcimycin induced autophagy exerts its antimycobacterial effect.
276
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
250 Host-responses to mycobacteria /interaction
22/09/2016
Insights into modulatory roles of vitamin C on human monocytic cell line, THP-1: An ex
vivo model for studying host-pathogen interaction
S. Dhingra, M. Nandi, K. Sikri, J.S. Tyagi
Biotechnology, All India Institute of Medical Sciences, Delhi, India
Macrophages are considered to be the first line of defence in response to any pathogen. The THP-1
cell line is widely used for studying macrophage functions and for analyzing host cell-pathogen
interactions. Vitamin C (vit C) is established as a free radical scavenger, an essential cofactor of
many enzymes and is required for immune effector cell function of macrophages.
We performed a genome-wide temporal gene expression and functional enrichment analysis of
THP-1 cells treated with 100 μM of vit C, a physiologically relevant concentration of the vitamin.
Extensive modulation of gene expression was observed from 8 hours to 96 hours as reflected by an
increase in the number of differential regulated genes. Further, functional enrichment analysis
based on statistically stringent criteria revealed a gamut of functional responses, namely,
‘Regulation of gene expression’, ‘Signal transduction’, ‘Cell cycle’, ‘Immune system process’,
‘Regulation of cAMP metabolic process’, ‘Cholesterol transport’ and ‘Ion homeostasis’ were
enriched. A comparative analysis of vit C-mediated modulation of gene expression data in THP-1
cells and human skin fibroblasts disclosed an overlap in certain functional processes such as
‘Regulation of transcription’, ‘Cell cycle’ and ‘Extracellular matrix organization’ and THP-1 specific
responses, namely, ‘Regulation of gene expression’ and ‘Ion homeostasis’. It was noteworthy that
vit C modulated the ‘Immune system’ process throughout the time-course. We have reported earlier
that tubercle bacilli treated with vit C develop an isoniazid-tolerant phenotype that is widely
considered to be an indicator of bacterial dormancy. This vit C-induced drug tolerant response
occurred in vitro as well as in infected THP-1 cells. In view of the ability of vit C to induce a ‘dormant’
phenotype in Mtb, this temporal transcriptome profiling study of baseline gene expression to assess
the response of THP-1 cells to vit C would pave the way for utilizing the vit C-based THP-1 infection
model to study biochemical and cellular responses of host cells to ‘dormant’ Mtb infection and other
intracellular pathogens.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
277
251 Host-responses to mycobacteria /interaction
22/09/2016
Mycobacterial N-Glycolyl Muramyl Dipeptide Drives Cell-Mediated Immunity from Complete
Freund’s Adjuvant
J. Dubé2-4-1, D. Montamat-Sicotte4-1, F. Mcintosh4-1, M. Behr3-1-4-2
1
McGill International TB Centre 2Dept. of Microbiology & Immunology, McGill University 3Dept. of
Medicine 4Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
The capacity for the mycobacterial cell to act as an adjuvant has long been recognized, notably in
the form of complete Freund’s adjuvant (CFA, killed M. tuberculosis in mineral oil). However, work
remains to define the molecular entities unique to mycobacteria that contribute to cell-mediated
immunity (CMI), as was first demonstrated by Chase and Landsteiner circa 1940. Subsequent work
by Lederer and colleagues suggested the peptidoglycan fragment N-acetyl muramyl dipeptide
(MDP) was the minimal component for CFA adjuvancy. MDP was later shown to elicit innate
immune responses via the pattern recognition receptor NOD2. Together, this has led to two related
hypotheses: 1) the CMI adjuvant effect is in part mediated by NOD2, and 2) mycobacteria, instead
of other more conventional bacteria, were used by Freund because of their unique N-glycolylated
MDP.
To determine the contribution of N-glycolyl MDP, acting via NOD2, to the CMI response elicited by
CFA, we have investigated these two hypotheses at two levels: the whole animal, using an in vivo
murine model of immunization, and the cell, by contrasting the effect of N-glycolyl MDP with Nacetyl MDP in different ex vivo models of innate and adaptive immunity. Ultimately, we aim to
‘complete’ Freund’s incomplete adjuvant (mineral oil without mycobacteria) by adding specific
mycobacterial pathogen-associated molecular patterns (PAMPs).
Data indicate that the majority of CMI elicited by CFA is dependent on the pattern recognition
molecule NOD2, strongly implicating MDP in CFA adjuvancy. At the cellular level, N-glycolyl MDP
augments dendritic cell (DC) functions as compared to N-acetyl MDP, when measuring cytokine
secretion, upregulation of costimulatory molecules and T-cell activation. Complementing the PAMP
data, we observe that these DC read-outs are diminished with either of two genetic manipulations:
1) using Nod2-/- DCs, or 2) infecting DCs with mutated mycobacteria that are unable to produce Nglycolyl MDP. This improved understanding of mycobacterial cell adjuvancy will inform our
perspective on the immunomodulatory capacity of mycobacterial pathogens and may provide
insights about BCG as a vaccine and an immunotherapeutic agent.
278
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
252 Host-responses to mycobacteria /interaction
22/09/2016
Insights into the role of a Mycobacterium abscessus specific MmpL transporter involved in
the macrophage-mycobacterium interaction
V. Dubois3, L. Laencina3, A. Viljoen1, V. Le Moigne3, G. Sapriel3, E. Dumas2, E. J Rubin4,
M. Blaise1, L. Kremer1, J.L. Herrmann3, F. Girard-Misguich3
1
FRE3689, Montpellier 2UMR8079, CNRS, Paris Saclay 3U1173, UVSQ, Saint-Quentin-EnYvelines, France 4Department of Immunology and Infectious Diseases, Massachusetts, United
States
Mycobacterium abscessus (Mabs) is a non-tuberculous rapidly growing mycobacterium mainly
responsible for lung infections. Prevalence of Mabs infections has been increasing worldwide, and
specifically among Cystic Fibrosis patients. Mabs has been shown to grow and survive in eukaryotic
cells like macrophages. Benefiting of a transposon mutant library obtained in a Mabs subsp
massiliense smooth clinical Korean isolate, we aimed at deciphering the genes allowing Mabs to
survive intracellularly. 6000 Tn mutants were screened individually for their survival impairment in
macrophages. One specific mutant was identified with a Tn insertion within MAB_0855
(MAB_0855::Tn), encoding a mycobacterial membrane protein Large (MmpL) protein. MmpL protein
members are mycobacterial transporters involved in the maturation and export of cell wall lipids and
can also act as drug efflux proteins.
These proteins are well-described in Mycobacterium tuberculosis: some of them play a role in M.
tuberculosis virulence such as MmpL8 and MmpL10. Although no orthologs of MAB_0855 are found
in M. tuberculosis by Bidirectional Best Hit (BBH), a Blast analysis and phylogenetic studies
revealed the presence of MAB_0855 within the M. tuberculosis mmpL8, 10 and 12 gene cluster.
A Thin Layer Chromatography of the extractible lipids unraveled subtle changes in its parietal lipids
composition. One consequence of this cell wall defect results in impaired adhesion to macrophages
and a subsequent diminution of MAB_0855::Tn internalization by the cells. In addition, the intramacrophage survival of MAB_0855::Tn is highly reduced in comparison to the parental strain. To
further confirm the phenotype of MAB_0855::Tn, the MAB_0855 gene has been inactivated in both
the smooth and rough reference strains of Mabs.
Functional complementations are currently being carried out in these two gene deletion mutants to
confirm and validate the contribution of MAB_0855 in the observed phenotypes. The impact of the
gene deletion will also be studied in whole infected zebrafish embryos.
Here, we identified and characterized a new MmpL protein implicated in shaping the cell-wall and in
the Mabs/macrophage interactions, further supporting the importance of these transporters in Mabs
virulence.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
279
253 Host-responses to mycobacteria /interaction
22/09/2016
Ancient and modern M. tuberculosis lineages differently modulate the innate immune
response and autophagy in human macrophages
E. Petruccioli3, A. Romagnoli3, I. Palucci2, E. Carata1, L. Petrone3, S. Camassa2, L. Dini1,
E. Girardi3, G. Delogu2, D. Goletti3, G. Fimia3-1
1
Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali, University of Salento,
Lecce 2Department of Microbiology, Catholic University 3Epidemiology and preclinical research,
Inmi l. Spallanzani irccs, Rome, Italy
Introduction: Recent studies highlighted the impact of M. tuberculosis (Mtb) genetic diversity on
relevant pathogenetic properties, with Mtb-modern lineages associating with reduced immune
responses that consequently led to higher Mtb intracellular replication and, as such, potentially
linked to rapid progression from infection to disease. Moreover, laboratory strain H37Rv reduces
macrophage autophagy flux, which can be fully restored by the host specific immune response,
leading to a reduction of the Mtb intracellular survival.
Aim: To in vitro evaluate the impact of the Mtb genetic diversity on the immune response, Mtb
replication and modulation of the autophagic flux in human primary macrophages.
Methods: Different Mtb clinical isolates belonging to strains from different lineages (1-5), in addition
to H37Rv, were used to infect human macrophages. We overtime evaluated Mtb replication (CFU);
cytokines (cytometric bead array) in cell supernatants; autophagy flux (immunoblot).
Results: Mtb strains belonging to modern lineage 4 show high rate of replication, associated to a
significant production of inflammatory cytokines (IL1, IL6, TNFa), but not of IL-8. Moreover, high
autophagy flux was observed in macrophages infected with lineage 4 Mtb, which is stimulated via
an IL1-dependent signaling. Instead, Mtb strains belonging to the ancient lineage 1 and 5, were
characterized by a significant lower cytokine production, reduced bacterial replication and
decreased autophagy flux.
Conclusion: Mtb belonging to modern lineages show an unpredicted high capability to stimulate
the innate immune responses and the autophagy flux of in vitro infected macrophages. However,
activation of both processes is not effective in limiting the intracellular replication of these MTB
strains. Conversely, these patterns may be correlated to their increased pathogenic properties with
respect to ancient ones.
280
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
254 Host-responses to mycobacteria /interaction
22/09/2016
Investigating the role of rv2660c and rv2661c during mycobacterial stress repsonses and
non-replicating persistence
N. Hafneh, G. Mukamolova
Dept. Of infection, immunity & inflammation, University of Leicester, Leicester, United Kingdom
One key aspect of Mycobacterium tuberculosis (Mtb) pathogenesis is its ability to survive stress
conditions encountered during macrophage infection and transit into a state of non-replicating
persistence (NRP) which is characterised by low metabolic activity and alteration of gene
expression. This forms the basis of global risk of latent tuberculosis and justify efforts to identify key
latency-specific antigens as component of vaccines for prevention of TB reactivation. rv2660c
encoding a conserved hypothetical protein was shown to be significantly up-regulated in several
NRP models and in late stages of both murine and macaque infections. The multistage vaccine H56
which included a recombinant Rv2660c protected cynomolgus macaques against active TB and
reactivation of latent TB (1), while Rv2660c peptides induced specific T-cell responses in TB
patients (2). Although the existence of Rv2660c has been recently challenged by identification of a
small non-coding RNA, ncrv12659 overlapping with a coding sequence of rv2660c (3), the
importance of the genomic region annotating rv2660c, rv2661c and ncRv12659 in Mtb pathogenesis
and stress response has not been investigated. To address this question, we have generated Mtb
H37Rv strains with deleted or over-expressing rv2660c and rv2661c and characterise their
biological phenotypes. ∆rv2661c, ∆rv2660c, ∆rv2661c+rv2660c mutants strains showed no growth
defect in either 7H9 Middlebrook or Sauton’s media. In addition, these strains survived similarly to
the wild type Mtb during starvation and under hypoxic conditions. However, survival of
∆rv2661c+rv2660c mutant was significantly impaired during oxidative stress, while over-expression
of rv2661c+rv2660c from a strong hsp60 promoter improved Mtb survival in the same condition. Our
findings suggest that Rv2660c and Rv2661c may facilitate Mtb response to reactive oxygen and
nitrogen species. Further experiments will elucidate the role of Rv2660c and Rv2661 in Mtb
infection and persistence in vivo.
References:
1. Lin et al, J Clin Invest. 2012,122: 303-14.
2. Govender et al, Vaccine. 2010;29: 51-7
3. Houghton et al, PLoS One. 2013, 8(12): e80047.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
281
255 Host-responses to mycobacteria /interaction
22/09/2016
Mycobacterium tuberculosis peptidyl-prolyl isomerases modulates host immune response
and aids in intraphagosomal survival
D. Tripathi1, S. Pandey2-3, M. Khubaib3-2, N.Z. Ehtesham2, S.E. Hasnain1
1
Kusuma School of Biological Sciences, Indian Institute of Technology 2Inflammation Biology and
Cell Signaling Laboratory, National Institute of Pathology, Delhi 3School of Life Sciences, University
of Hyderabad, Hyderabad, India
Tuberculosis caused by the intracellular pathogen Mycobacterium tuberculosis (M.tb), remains a
potential threat regardless of strong efforts to alleviate its toll on humanity.
Intracellular/intraphagosomal survival plays a critical role in the infection cycle of the pathogen, a
process which majorly relies on array of virulence factors to colonize and replicate within the host
macrophages. Studies involving immunological characterization of these effector molecules can
bridge huge gaps in our understanding of M.tb biology and facilitate better therapeutic and
diagnostic interventions.
M.tb is known to possess two Ppiases (cyclophilins), PpiA and PpiB. M.tb PpiA is a part of the
secretome and is known to interact with host proteins involved in immune defense mechanism and
signal transduction, while PpiB has been reported in membrane fraction and mannosylation
enriched culture filtrate. In the present study, we describe the potential role of Ppiases in
intraphagosomal survival of the pathogen. We present evidence that M.tb Ppiases play role in
modulating host immune responses. ELISA results revealed presence of antibodies to M.tb Ppiases
in the patient sera as compared to the sera of healthy individuals. Treatment of THP-1 cells with
increasing dose of rPpiA induced secretion of pro-inflammatory cytokines. Alternatively, treatment
with rPpiB inhibited secretion of TNFα and induced secretion of IL-10. Furthermore, heterologous
expression of M.tb PpiA and PpiB in Mycobacterium smegmatis increased its survival in THP-1 cells
as compared to the vector control. Our results demonstrate that M.tb Ppiases are immunogenic
proteins that can possibly modulate host immune response and enhance persistence of the
pathogen within the host by subverting host cell generated stresses.
282
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
256 Host-responses to mycobacteria /interaction
22/09/2016
Pulmonary Microbiota: Impact on Lung Morphology, Respiratory Diseases and Immune
Responses
M. Hauptmann, Y. Yun, U. Schaible
Cellular Microbiology, Research Center Borstel, Borstel, Germany
With the advent of high throughput sequencing tools, it became evident that the healthy lung is not
sterile as considered so far, but contains a diverse repertoire of aerobic and anaerobic bacterial
species. In contrast, standard microbiological techniques only encompassed a small percentage of
total microbiota in healthy lungs. These lung microbiota isolates however can now be used for
murine lung colonization experiments.
Colonizing germ-free mice with Lactobacillus spec. isolated from lungs of wild mice, had profound
influence on lung morphology. We observed higher numbers of alveoli and increased mucus
production. As research on the impact of pulmonary microbiota is still in its beginning, it is still an
open question how pulmonary microbiota influence immune responses and the outcome of
respiratory diseases. To address these questions, we have established an in vivo model that allows
manipulations of pulmonary microbiota in adolescent mice and subsequent infection with
Mycobacterium tuberculosis (M. tb). Our results revealed that the course of M. tb infection was
similar between specific pathogen free (SPF), antibiotic-treated, and antibiotic-treated mice that
were recolonized with bacterial isolates from wild mice lungs. However, a tendency towards higher
percentage of IFN-γ+ CD4+ T cells was observed in recolonized mice. At present, we do not know
whether this response was due to non-physiologically high burden of commensal bacteria during M.
tb infection, or whether immune responses in the lung can be trained by microbial exposure, as has
been suggested from models of asthma and allergies. Therefore, we currently investigate the effect
of microbiota-alterations during early life on M. tb infection by the use of germ-free mice.
The emerging field of lung microbiota research will provide new insights on how environmental
factors shape the disposition for respiratory disorders like infections, allergies, COPD and acute
respiratory distress syndrome.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
283
257 Host-responses to mycobacteria /interaction
22/09/2016
Characterization of IgG responses and isolation of B cells from healthy individuals and TB
patients
S. Hwa2-1, D. Mahamed2, A. Sigal2-1
1
Systems Infection Biology, Max Planck Institute for Infection Biology, Berlin, Germany 2Sigal Lab,
K-RITH, Durban, South Africa
Isolation of human monoclonal antibodies is a powerful tool for understanding the humoral response
against pathogens. In both humans and nonhuman primates with active TB, anti-Mycobacterium
tuberculosis (Mtb) serum titers increase and the reactivity toward specific antigens shifts.
Transcriptomic data also suggest a B cell role in health versus TB disease states. We hypothesize
that immunoglobulins do play a role in protection against TB disease and aim to recover antibodies
with functional activity. We developed an automated image analysis method to screen
immunoglobulin samples for effects on phagocytosis and growth of Mtb in primary human
monocyte-derived macrophages. Polyclonal serum IgG from healthy South African volunteers
restricted the growth of Mtb in macrophages compared to IgG derived from TB patients or US-origin
normal serum. We also showed proof of concept that B cells can be sorted by binding fluorescently
labelled whole bacteria and/or purified antigens, and constructed a feeder cell line which supports
proliferation and IgG secretion from primary B cells without further cytokine supplementation. Single
cells sorted into microtiter plates containing feeder cells yielded secreted IgG up to several hundred
ng/mL, as well as cDNA sequences of heavy and light chain mRNA. Monoclonal antibodies
recovered from the single-cell cultures will be screened for effects on phagocytosis as above. We
are currently enrolling volunteers undergoing lung resection for TB or non-TB lung disease controls
which will allow us to characterize the antigen specificities and functional activities of antibodies
from B cells in the draining lymph nodes.
284
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
258 Host-responses to mycobacteria /interaction
22/09/2016
Interferon gamma production upon mycobacterial stimulations is controlled by major loci
on chromosome 8q and 3q in various populations exposed to tuberculosis
F. Jabot-Hanin3-6, A. Cobat3-6, J. Feinberg3-6, J. Bustamante3-6, S. Boisson-Dupuis9-3-6,
J.L. Casanova8-9-3-4-6, E.G. Hoal7, E. Schurr1-2, A. Alcaîs9-3-6, C. Delacourt5, L. Abel9-3-6
1
McGill International TB Centre 2Department of Human Genetics and Department of Medicine,
McGill University, Montreal, Canada 3Laboratory of Human Genetics of Infectious Diseases,
Necker Branch,U1163, INSERM 4Pediatric Hematology-Immunology Unit 5Pediatric Pneumology
Unit, Necker Hospital for Sick Children, AP-HP 6Imagine Institute, Paris Descartes University,
Sorbonne Paris Cité, Paris, France 7Molecular Biology and Human Genetics, MRC Centre for
Molecular and Cellular Biology, DST/NRF Centre of Excellence for Biomedical TB Research,
Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa 8Howard Hughes
Medical Institute 9St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller
Branch, Rockefeller University, New York, Ny, United States
More recent and more specific than Tuberculin Skin Test (TST), Interferon Gamma (IFN-γ) release
assays (IGRAs) provide an in vitro measurement of antimycobacterial immunity that is widely used
as a test for tuberculosis (TB) infection. IGRA outcomes are highly heritable in various populations,
but the nature of the involved genetic factors remains unknown.
We conducted a genome-wide linkage analysis of IGRA phenotypes in 143 families from a TB
household contact study in France with the new Maximal Likelihood Binomial method which
considers the sibship as a whole and makes no assumptions about the distribution of the
phenotype. A replication study in 135 families
from South Africa was also conducted to confirm the loci identified. We identified a major locus on
chromosome 8q controlling IFN-γ production in response to stimulation with live BCG (LOD
score=3.81, p=1.40x10-5). We also detected a second locus on chromosome 3q that controlled
IFN-γ levels in response to stimulation with ESAT-6 antigen when accounting for the IFN-γ
production shared with that induced by BCG (LOD score=3.72, p=1.8x10-5). Both loci were
replicated in South African families, where TB is hyperendemic, and do not overlap with the TST1
and the TST2 loci controlling TST positivity per se and the intensity of TST reactivity, respectively.
Refined identification of the new identified loci is ongoing by ultra-fine association studies of each of
the two phenotypes within the corresponding linked regions, with interesting preliminary results. The
identification of these two new linkage signals in populations of various ethnic origins living in
different M. tuberculosis exposure settings provides new clues to the genetic control of human
antimycobacterial immunity.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
285
259 Host-responses to mycobacteria /interaction
22/09/2016
Cord-forming Mycobacterium tuberculosis causes the formation of macrophage
extracellular traps
S. Kalsum, C. Braian, M. Lindroth, M. Lerm
Dept. of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
In 1882, Robert Koch observed microscopic cords as one of the first phenotypic characteristics in
Mycobacterium tuberculosis, the causative agent of tuberculosis. When grown in a liquid medium
without detergent, they form pellicle-like structures, or cords, in which the orientation of the long axis
of each cell is parallel to the long axis of the cord. The formation of cords has been shown to
correlate with increased virulence. Like neutrophils, the formation of extracellular traps has been
recently recognized in macrophages and referred as macrophage extracellular traps (METs). METs,
like neutrophil extracellular traps, are composed of DNA and histones. We found that cord-forming
mycobacteria (H37Rv) can induce MET formation in human monocytes-derived macrophages
(hMDMs). Formation of METs in hMDMs is independent of ROS production but dependent on M.
tuberculosis virulent factor ESAT-6. Now, we want to evaluate the response of cord-forming
mycobacteria in a more complex system such as our recently developed human lung tissue model.
Knowledge on how different phenotypes of M. tuberculosis interacts with human cells and tissues is
fundamental for the understanding of the devastating disease that this pathogen is causing.
286
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
260 Host-responses to mycobacteria /interaction
22/09/2016
Mycobacterium smegmatis Δespg3 mutant a potential live bacterial vaccine vector for
antigen delivery
N. Kannan, M. Haug, T. Flo, M. Steigedal
CEMIR,, NTNU, Trondheim, Norway
Background and Objective: Mycobacteria possess a family of protein secretion systems, ESX-1 to
ESX-5, which has been demonstrated to aid in altering host pathways and promote bacterial
survival. One of them, ESX-3, is ubiquitously present in all mycobacterial species and is regulated
by iron and zinc; also it has been shown that ESX-3 is involved in siderophore mycobactin based
iron uptake. Recently, a mutant strain of the non-pathogenic M. smegmatis lacking ESX-3,
complemented with Mycobacterium tuberculosis esx-3 has shown promise as a vaccine candidate.
We wanted to investigate how the vaccine effect was created so efficiently.
Methods: To study the host response to mutant M.smegmatis, we investigated the survival of the
mutant within in macrophages and dendritic cells. In addition we measured host response as ability
to produce various cytokines and other immune modulators. Finally, we also investigated the
antigen presenting ability of the M. smegmatis strains using a model ovalbumin system.
Results and conclusion: Preliminary data indicate that mutant M. smegmatis has reduced survival
when compared to the wild type when added to macrophages and dendritic cells. As a
consequence of this we observe a significant change in expression profile of a panel of immune
modulators. Further, we observed improved antigen presentation upon exposure to the mutant
vector when compared to the wild type. Currently, we are conducting mouse experiments, to check
if the mutant strain may provide protective immunity when compared to BCG.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
287
261 Host-responses to mycobacteria /interaction
22/09/2016
Rv0753c, a novel dendritic cell-activating antigen of Mycobacterium tuberculosis, drives
Th1-type T-cell immunity
J.S. Kim, W.S. Kim, H.H. Choi, H.M. Kim, K.W. Kwon, S.B. Cha, S.N. Cho, S.J. Shin
Department of Microbiology and Institute for Immunology and Immunological Diseases, Brain
Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South
Korea
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, is an outstanding pathogen
that modulates the host immune response. This inconvenient truth drives the continual identification
of antigens that generate protective immunity, including Th1-type T cell immunity. Here, the
contribution of methylmalonate semialdehyde dehydrogenase (MmsA, Rv0753c) of Mtb to immune
responses was examined in the context of dendritic cell (DC) activation and T cell immunity both in
vitro and in vivo. The results showed that MmsA induced DC activation by activating the MAPK and
NF-κB signaling pathways. Additionally, MmsA-treated DCs activated naïve T cells, effectively
polarized CD4+ and CD8+ T cells to secrete IFN-γ and IL-2, and induced T cell proliferation. These
results indicate that MmsA is a novel DC maturation-inducing antigen that drives the Th1 immune
response. Thus, MmsA was found to potentially regulate immune responses via DC activation
toward Th1-type T cell immunity, enhancing our understanding of Mtb pathogenesis.
288
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
262 Host-responses to mycobacteria /interaction
22/09/2016
Epigenetic reprogramming of peripheral blood mononuclear cells in BCG-vaccinated
individuals
M. Lerm
Dept of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
Bacille Calmette-Guerin (BCG) was developed in the 1920s as a vaccine against tuberculosis (TB)
and albeit having variable efficacy against TB, it is one of the world’s most widely used vaccines.
The mechanisms responsible for the protective effects of BCG are not well understood. Here, we
isolated peripheral blood mononuclear cells (PBMCs) from BCG-vaccinated subjects and performed
global DNA methylation analysis in combination with functional assays to address epigenetic
changes and functional responses induced through BCG vaccination. Enhanced containment of
Mycobacterium tuberculosis replication could be observed in monocyte-derived macrophages from
a sub-group of BCG-vaccinated individuals (classified as responders). A stable and robust
differential DNA methylation pattern in response to BCG could be observed in PBMCs isolated from
the responders but not from the non-responders. Gene ontology analysis revealed that promoters
with altered DNA methylation pattern were strongly enriched among genes belonging to immune
pathways in responders but no enrichment could be observed in the non-responders. Our findings
suggest that BCG-induced epigenetic reprogramming of immune cell function can enhance antimycobacterial immunity. Understanding why BCG induces this response in responders but not in
non-responders could provide clues to improvement of TB vaccine efficacy.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
289
263 Host-responses to mycobacteria /interaction
22/09/2016
B-lymphocytes forming follicle-like structures in the lung tissue of TB-infected mice:
phenotypic and functional features
I. Linge, E. Kondratieva, V. Avdienko, A. Dyatloff, E. Petrova, A. Apt, T. Kondratieva
Laboratory for Immunogenetics, Central Institute for Tuberculosis, Moscow, Russia
In the course of tuberculosis (TB) infection, B cells form follicle-like aggregates in close vicinity of TB
lung granuloma. Such aggregates are known in TB patients, as well as in mycobacteria-infected
mice and monkeys; however, their role in TB immunity yet remains obscure. To shed more light on
this problem, we assessed the surface phenotype and function of lung B cells during TB in
genetically TB-susceptible I/St mice in considerable detail. Our results demonstrate that the majority
of lung B cells closely resemble classical B2 (surface phenotype CD19+IgMloIgDhiCD21/35intCD5CD11b-CD43-) but differ from the latter by the absence of B2-cell marker CD23. Although it is
speculated that the CD23 expression may drop after B-cell activation, in our system the
overall proportion of activated B cells, as measured by CD80 and CD69 expression and the
numbers of the CD138+ plasma cells, was less than 10 percent, suggesting that CD23 expression
does not reflect the activation status.
Thus, we expected that lung B-cells’ functions should not differ much from B cells populating
lymphoid organs. Indeed, lung B-cells constitutively expressed MHC II molecules, with a slight
elevated after infection. Accordingly, lung B-cells presented mycobacterial antigens to immune
CD4+ T-cells as effectively as their splenic counterparts. We also assessed the secretion level of key
pro- and anti-inflammatory cytokines by lung B-cells and observed production of pro-inflammatory
IL-6 (very high) and IL-11 (moderate), whereas marginal to no production of classical type 1 (TNF-α,
IFN-γ), or anti-inflammatory IL-10 and TGF-β.
Assessment of antibodies evidenced that a very large proportion of the total antibody response in
tuberculous lung shows no specificity to mycobacteria. A major proportion of immunoglobulins
produced by lung B cells from infected mice did not react with mycobacterial antigens, despite a
high reactivity of sera from the same mice. A panel of monoclonal antibodies obtained from lung B
cells contained only few clones demonstrating anti-mycobacterial specificity or reactivity with the
infected lung tissue extract, whereas the majority of clones’ specificity remained unidentified. Taken
together, our results suggest that B cell response against TB infection in the lung has a clear
pathological and a doubtful protective role.
This work was financially supported by the Russian Scientific Foundation (grant 15-15- 30020).
290
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
264 Host-responses to mycobacteria /interaction
22/09/2016
Human and bovine tubercle bacilli elicit distinct innate responses during early stage
infection of bovine alveolar macrophages
K.M. Malone6, K. Rue-Albrecht1, D. Magee1, K. Conlon6, N. Nalpas1, J.A. Browne1, A. Smyth6,
E. Gormley5, D.E. Mc-Hugh1-2, S. Gordon6-4-3-2
1
Animal Genomics Laboratory, School of Agriculture and Food Science 2Conway Institute of
Biomolecular and Biomedical Research 3School of Biomolecular and Biomedical Science 4School
of Medicine 5Tuberculosis Diagnostics and Immunology Research Centre, School of Veterinary
Medicine 6Tuberculosis Molecular Microbiology group, School of Veterinary Medicine, University
College Dublin, Dublin, Ireland
Members of the Mycobacterium tuberculosis complex (MTBC) exhibit a high degree of genetic
identity and yet show distinct host preference; for example Mycobacterium bovis is highly virulent for
cattle while the human pathogen M. tuberculosis is attenuated in the bovine host. Differential
immunopathology exhibited by MTBC members may have a basis in host innate immune
mechanisms; hence the initial interaction between pathogen and host may be key for the host
preference observed within the MTBC. As a route towards defining the differential innate immune
response to host-adapted tubercle bacilli, we have performed the first in-depth comparative
transcriptomic analysis of the bovine alveolar macrophage during infection with M. bovis AF2122/97
and M. tuberculosis H37Rv. In addition, both the transcriptomic and proteomic profiles of M. bovis
AF2122/97 and M. tuberculosis H37Rv were evaluated during in vitro growth to identify differential
expressed effectors between the pathogens that could drive divergent interactions with the host cell.
The bovine alveolar macrophage response to infection with M. bovis and M. tuberculosis was found
to be highly similar over the first 24 hours of infection, with substantial differentially expressed genes
between the M. bovis- and M. tuberculosis-infected macrophages only observed 48 hours post
infection. Differences observed in cell death and autophagy responses, eicosanoid metabolism and
type I interferon signalling suggest a distinct engagement by M. bovis with the bovine innate
immune system, and suggest increased cytosolic access during the first 48 hours compared to M.
tuberculosis. In addition, profiling of both pathogens revealed increased expression of the ESX-1
secretion system in M. bovis in contrast to M. tuberculosis; this latter observation presents a
potential mechanism to explain the increased cytosolic access of the bovine bacillus during the
initial stages of macrophage infection. The work presented here therefore provides insight into
factors that may underlie host preference between the bovine and human tubercle bacilli during the
early stages of infection.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
291
265 Host-responses to mycobacteria /interaction
22/09/2016
Characterization of the repertoire of lipid-derived epitopes presented by CD1 proteins
during mycobacterial infection
J. Mazurek2-3, C. Carrat2, G. Gaibelet1, E. Fabre2, E. Layre2, M. Gilleron2
1
Institut de Génomique Fonctionnelle, Montpellier 2Institut de Pharmacologie et de Biologie
Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France 3Karolinska Institutet,
University Hospital Huddinge, Stockholm, Sweden
Lipid-containing antigens are presented to T cells in the complex with CD1 molecules. Several
glycolipids derived from mycobacterial cell wall have been found among CD1 ligands. Moreover,
CD1-restricted T cells recognizing mycobacterial lipid antigens have been isolated from the blood of
Mycobacterium tuberculosis (Mtb)-infected patients. Such cells secrete the Th1 cytokines and are
cytotoxic, which indicates that the lipid antigen-mounted immune response plays a role in controlling
mycobacterial infections.
Particularly, Mtb-derived mycolic acid, glycerol and glucose monomycolates, phosphatidyl-myoinositol mannosides (PIM) and diacylated sulfoglycolipids (Ac2SGL) have been found to be
associated with CD1b molecules. Whilst some lipid antigens, e.g. Ac2SGL, are presented to T cells
without being processed, others, like PIM6, require prior processing to activate T cells. Up to now,
identification of a lipid antigen depended on the availability of the specific T cell clone(s) recognizing
the lipid epitope which severely limits the identification of the new CD ligands.
In this project we aim at characterizing the entire repertoire of Mtb-derived lipid antigens bound by
CD1b. We employ a novel strategy to define such antigens that is non-biased by the availability of T
cell clones. Furthermore, our methodology allows for isolation of CD1b-bound lipids without need for
using detergents, which severely impact the readout by dissociating lipids from the CD1b binding
groove. We constructed antigen presenting cells (APCs) expressing protease-cleavable CD1b
molecules (pCD1b). Such pCD1b molecules could be proteolitically cleaved and purified on an
affinity chromatography column without destabilization of their antigen binding capacity. Next, lipids
separated from CD1b proteins by the organic solvent extraction are identified by liquid
chromatography followed by mass spectrometry analysis. Using this strategy we will identify the lipid
epitopes associated with CD1b molecules upon exposure of the APCs to the Mtb cell wall-derived
lipid mixture, and also upon infection with Mtb. Identified lipid epitopes will next be tested for their
antigenicity on the blood obtained from the Mtb-infected individuals and the immune response
triggered by such epitopes will be characterized.
292
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
266 Host-responses to mycobacteria /interaction
22/09/2016
Establishment of a cell culture method to functionally study the TB-helminth co-infection
M. Méndez1, N. Angulo1, R. Gilman3-1, M. Verástegui1, O. Stendahl2, M. Lerm2, R. Blomgran2
1
Laboratorio de Investigación en Enfermedades Infecciosas, Universidad Peruana Cayetano
Heredia, Lima, Peru 2Department of Clinical and Experimental Medicine, Division of Medical
Microbiology, Linköping University, Linköping, Sweden 3Department of International Health, The
Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
Helminth infections are among the most common infections worldwide and are associated with an
increased host susceptibility to develop tuberculosis (TB) disease. By inducing a strong Th2
immune response, helminths impair Th1 responses that are necessary for protection against TB.
This reduction in Th1 response has been mostly studied at the systemic level by assessing the
cytokine profiles and in adaptive immunity by analyzing T-cells. However, M. tuberculosis can
replicate within innate immune cells, especially the macrophage, and manipulate the fate of these
cells contributing to the severity of the disease. Within this context, functional studies on
macrophages from TB-helminth coinfected individuals are lacking. Our aim is to investigate the
immunological effect of helminth infection on the macrophages of TB patients. For this, we start by
optimizing a cell-culture system in which we can obtain monocyte-derived macrophages from
cryopreserved PBMCs and differentiated with autologous serum. In this phase we work with healthy
donors to later, once established the method, carry out an observational study on a Peruvian
Amazon region that is endemic for both helminthiasis and TB. By using autologous serum to
differentiate macrophages ex-vivo, we intend to provide conditions closer to their original
environment and determine macrophage polarization, ability to control M. tuberculosis infection and
cytokine secretion pattern.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
293
267 Host-responses to mycobacteria /interaction
22/09/2016
Factors Affecting TB transmission from adult to children within households in the Gambia
A. Mendy1, L. Tientcheu1, U. Egere1, E. Coker1, A. Ayorinde1, B. Kampmann1-2, D. Leopold1
1
Vaccines and Immunity Theme, TB Immunology Lab, Medical Research Council (MRC) Gambia
unit, Banjul, Gambia 2Faculty of Medicine, Imperial College London, London, United Kingdom
Background: Childhood tuberculosis (TB) has significant impact on public health worldwide and it
is believed that most children acquire TB from an adult smear-positive index case within their
household. To further examine this hypothesis and to investigate transmissibility of strains within the
household setting, we compared strain-types in adults and their child contacts. We also examined
the influence of bacillary burden and strain-type on clinical outcome of contacts.
Materials and Methods: Stored isolates from smear positive adult TB cases (n =136) were
selected according to clinical outcomes of their household contacts (children ˂ 15 years old).
Mycobacteria were isolated from both adult and –where available- children samples via culture, and
typed using spoligotyping to enable strain classification.
Results: The AFB grade of adult index cases correlated with clinical outcome of the children with
microbiologically confirmed TB, clinically diagnosed probable TB, asymptomatic but TST positive
and asymptomatic, TST negative children showed 60%, 35%, 34% and 33% highest AFB grade
(3+) levels respectively.
Strain-type determination by spoligotyping showed that 93% of children had acquired EuroAmerican lineages, while 7% had M. Africanum lineage. Combining results for adult contacts of
children with confirmed TB and probable TB, 76% showed Mtb-Euro-American, 17% M. africanum
and 7% Mtb-Indo-Oceanic. Contacts of TST positive children showed 59% Mtb-Euro-American,
32% M. africanum, 8% Mtb-Indo-Oceanic and 2% Mtb-Beijing. Those of TST negative children
showed 63% Mtb-Euro-American, 26% M. africanum, 9% Mtb-Indo-Oceanic and 2% Mtb-Beijing.
Conclusion: The data so far support other published data, which show that a higher bacillary
burden in the index case increases the likelihood of TB transmission to child contacts. Adult patients
appear to be more likely to transmit TB if they were carrying Euro-American lineages rather than
West African strains.
294
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
268 Host-responses to mycobacteria /interaction
22/09/2016
Antigen HLA-DR at patients with different clinical manifestations of tuberculosis
T. Morozova, T. Salina
Department of Tuberculosis, Saratov State Medical University, Saratov, Russia
HLA-DR - antigen MHC (major histocompatibility complex) class II needed to generate an adequate
immune response.
Aim: Study of expression HLA-DR on immune cells of patients with different clinical manifestations
of tuberculosis.
Materials: Examined 54 patients newly diagnosed pulmonary tuberculosis (TB) and 22 healthy.
Depending on the clinical manifestations of TB patients divided into 2 groups) Groupa 1 included 18
patients with severe and complicated forms of tuberculosis. Groupa 2 included 36 patients with
limited and favorable flowing TB. Assessed the relative number of cells HLA-DR, in the peripheral
blood using immunophenotyping method with monoclonal antibodies (anti-HLA-DR).
Results: All patients with TB prior to antibiotic treatment were found an increase number of cells
HLA-DR - (M ± δ) (medium-arithmetic and standard deviation) - 16 ± 41,8% Me (median) - 40.6 Mo
(moda) - 17.4 interval 17,4-75) compared with healthy (M - 32,5,0 ± 12%, Me - 33.3 Mo - 33 3, the
range 14,4-50,3), p = 0.0017. The expression level of marker HLA-DR significantly higher in Group
1 (M - 17 ± 49,3% Me - 45 Mo - 65,4, 24,4-75 interval) compared with Group 2 (M - 38,2 ± 15%, Me
- 36.4 Mo - 17.4, range 17,4-64,2, p1-2 = 0,0122) and healthy (M - 32,5 ± 12%, Me - 33.3 Mo - 33.3,
interval - 14,4-50,3, p1-3 = 0,0086, p2-3 = 0,0495). All patients at baseline and after 2 months of TB
treatment found decrease in the number of cells of HLA-ДР after treatment of M - 41,8 ± 16% vs
30,4 ± 9%,respectively, p = 0.0009.
Conclusions:
1. In patients TB in acute phase of disease is observed elevated levels of cells HLA-DR in blood
compared with healthy.
2. The level of expression HLA-DR depends on the severity TB and is most pronounced in
patients with severe forms of disease.
3. In course of antibacterial therapy HLA-DR cells is reduced, which may indicate the
relationship of its expression with the level of the bacterial population M. tuberculosis. These
results open prospects for a targeted use of immune preparations
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
295
269 Host-responses to mycobacteria /interaction
22/09/2016
Human plasma lipidome variations during Mycobacterium tuberculosis infection
H. Neo3-2-1, S. Ji3, F. Torta3, A. Bendt3, C. Chee4, Y. Wang4, A. Singhal5, G. De Libero6, M. Wenk3
1
Agilent Technologies Singapore Pte Ltd 2Department of Biological Sciences, Faculty of
Science 3Singapore Lipidomics Incubator (SLING), National University of Singapore 4Tuberculosis
Unit, Tan Tock Seng Hospital, Singapore 5Singapore Immunology Network (SigN), A*STAR,
Singpaore, Singapore 6Department of Biomedicine, University Hospital Basel, Basel, Switzerland
Eradication of tuberculosis (TB) remains challenging with lengthy treatment durations and the
existence of drug resistant Mycobacterium tuberculosis (Mtb). As human pathogens continue to
infect and acquire resistance to current anti-microbial drugs, recent focus has turned to potential
host-directed therapeutics in search for novel treatment strategies. Pathogens such as Mtb make
use of host lipids as building blocks and influence the host cell physiology to enable their survival
and replication. Hence, detection of changes in the host lipidome during TB progression may enable
novel and precise diagnostic tools as well as potentially providing insights into host-pathogen
interactions. To test this hypothesis, we aimed to identify changes in the human plasma lipidome of
active TB patients (n=88) in comparison with the lipidome of healthy controls (n=62) and latent
individuals (n=36). After lipid extraction, analyses were performed by liquid chromatography mass
spectrometry (LC/MS) using targeted analysis for the major classes of lipids. The lipid classes that
showed the most significant differences (p < 0.05) were molecular species of ceramides,
sphingomyelins, plasmalogen and ether phosphatidylethanolamines. These lipids were significantly
lower in the plasma of active TB patients. Validation of these potential disease biomarkers in
longitudinal studies with human plasma samples from active TB patients (n=19) who were on
established TB drug treatment over a period of six months was carried out and will be discussed.
296
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
270 Host-responses to mycobacteria /interaction
22/09/2016
An interferon-γ-independent antimycobacterial mechanism
M. Resende1-2-4, M. Cardoso4, M. Borges3, A. Gil Castro1-2, R. Appelberg4
1
Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of
Minho, Braga 2ICVS/3B’s – PT Government Associate Laboratory, Braga/guimarães 3Faculty of
Pharmacy, University of Porto 4Institute for Molecular and Cell Biology (IBMC)/ I3S – Instituto de
Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
Granuloma formation is a hallmark of several infectious diseases, including those caused by
Mycobacterium sp. These structures are composed of accumulations of inflammatory cells and it
has been shown that cytokines such as IFNγ and TNFα are required for granuloma assembly during
M. avium infections in mice. MIIG mice have macrophages, monocytes and dendritic cells that are
unresponsive to IFNγ. We observed that although IFNγ-ko mice present an exacerbated infection,
the same is not true for MIIG animals, where the same levels of protection as the wild type (WT)
animals were observed in the liver and partial protection in the spleen. Moreover MIIG mice still
develop well defined granulomas (as opposed to IFNγ-ko mice), suggesting that IFNγ-mediated
macrophage activation is not required for granuloma assembly. This work also shows that MIIG
animals exhibit increased iron accumulations within the granulomas and increased cell recruitment
with higher CD4+ T cells numbers as well as increased IFNγ and TNFα expression, suggesting that
TNFα may have a role on protection and may compensate the lack of macrophage response to
IFNγ in the MIIG model. Indeed blocking TNFα with monoclonal antibodies in M. avium infected
MIIG animals induced an increased bacterial burden accompanied by a reduction of iron
accumulation in the granulomas. How the axis TNFα expression/iron accumulation overcomes the
lack of IFNγ signalization controlling the bacteria proliferation is being studied.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
297
271
Host-responses to mycobacteria /interaction
22/09/2016
Selective reduction of IFN-γ single positive mycobacteria-specific CD4+ T cells in HIV-1
infected individuals with latent tuberculosis infection
C. Riou2, R. Bunjun2, T.L. Muller2, A. Kiravu2, Z. Ginbot2, T. Oni3-1-4, R. Goliath3-1, R. Wilkinson3-1-6-5,
W. Burgers3-2
1
Clinical Infectious Diseases Research Initiative 2Department of Pathology, Division of Medical
virology 3Institute of Infectious Disease and Molecular Medicine 4School of Public Health and
Family Medicine, Division of Public Health Medicine, University of Cape Town, Cape Town, South
Africa 5Department of Medicine, Imperial College London 6Mill Hill Laboratory, The Francis Crick
Institute, London, United Kingdom
While in HIV-uninfected persons the risk of progression from latent to active tuberculosis (TB) is 210% in a lifetime, it increases to a 5-10% annual risk in HIV-infected individuals. Moreover, HIV-1 is
recognized to increase the risk for TB even before CD4+ T cell deficiency is profound. In order to
understand the mechanisms involved in the maintenance and/or impairment of TB latency, it is of
interest to define in detail the extent to which HIV affects TB immune responses.
Using flow cytometry, we compared the frequency, polyfunctional (IFN-γ, TNF-α and IL-2) and
memory profile (CD45RO, CD27) of ESAT-6/CFP-10-, PPD- and BCG-responding CD4+ T cells in
Mycobacterium tuberculosis-sensitized HIV-uninfected individuals (n=25) and HIV-infected subjects
(n=24) with well-preserved CD4 counts (median: 625 cells/mm3).
Our data revealed a significant and selective reduction of IFN-γ single positive CD4+ T cells, for all
mycobacterial antigens tested in HIV-infected participants, compared to HIV-uninfected individuals
(median fold reduction of 3.2, 10 and 5.6 for ESAT-6/CFP-10, PPD and BCG, respectively). The
proportion of IFN-γ single positive cells correlated inversely with viral replication (p=0.007, r=-0.66
for BCG responses and p=0.03, r=-0.4 for PPD and ESAT-6/CFP-10 responses). Interestingly, no
changes were observed in the magnitude of polyfunctional cells (producing IFN-γ+IL-2+TNF-α+)
between the two groups. Moreover, IFN-γ single positive cells were significantly enriched in a late
differentiated phenotype (CD45RO+CD27-, median: 22%, IQR: 17-39), compared to polyfunctional
cells (median: 12%, IQR: 12-18). As differentiated CD4+ T cells are more susceptible to HIV
infection; this could, in part, contribute to the preferential depletion of this subset.
Overall, we found that HIV-1 infection impairs TB immunity not only by numerically reducing
mycobacteria-specific CD4+ T cells, but also alters the balance of their functional profile by
selectively depleting IFN-γ single positive cells. Our results provide new insights into potential
mechanisms that perturb immune control of latent TB infection during HIV infection.
298
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
272
Host-responses to mycobacteria /interaction
22/09/2016
Antimycobacterial activity in whole blood from type I diabetes patients
S. Raposo-García3, S. García-García5, J. Guerra-Laso3, J. Juan-García5, C. Diez-Tascón2,
T. Nebreda-Mayoral4, E. López-Fidalgo1, R. López-Medrano6, O. Rivero-Lezcano1-7
1
Unidad de Investigación, Leon 2Anatomía Patológica 3Medicina
Interna 4Microbiología 5Neumología, Complejo Asistencial Universitario de León,
León 6Microbiología, Hospital El Bierzo, Ponferrada 7Instituto de Estudios de Ciencias de la Salud
de Castilla y León, Soria, Spain
In contrast with isolated phagocytes (usually monocytes and neutrophils), which are difficult to
activate against pathogenic mycobacteria, whole blood has shown to exhibit a strong
mycobactericidal activity. Diabetic patients are recognized in epidemiological studies as a
population susceptible to tuberculosis. We have used the whole blood model for in vitro infection to
analyze the susceptibility to tuberculosis in type I diabetes patients.
We have applied a factorial design to analyze the effect of four cytokines (IFNγ, TNFα, GM-CSF
and IL-15) and their interactions on the mycobactericidal activity of whole blood. Samples were
obtained from 18 volunteers: 9 type I diabetes patients and 9 non-diabetic controls, matched by age
and gender, and 18 in vitro infections with different cytokine combinations were performed with each
sample. Upon infection with Mycobacterium tuberculosis and incubation for two days in a rotary
shaker, antimicrobial activity was estimated by colony counting. Additionally, we studied the
phagocytic activity of neutrophils by flow cytometry, using a Mycobacterium bovis BCG transformed
with the gene of the fluorescent protein DsRed.
Mycobactericidal activity was significantly lower in blood from diabetic patients (P*=0.005). When
patients and controls were analyzed independently, it was observed that IL-15 stimulated the
antimicrobial activity in patients, but not in controls. In addition, the phagocytic activity of neutrophils
from diabetic patients was significantly lower (P*=0.006). Monocytes from patients showed also a
lower phagocytic activity, although it did not reach statistical significance (P*=0.066).
These results prompt further research to assess whether the whole blood model may be suitable for
testing the tuberculosis susceptibility of different groups as exemplified by type I diabetic patients
that present a lower capacity to kill M. tuberculosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
299
273
Host-responses to mycobacteria /interaction
22/09/2016
The impact of influenza coinfection on the control and progression of tuberculosis
S. Ring1, L. Eggers1, G. Gabriel2, B. Schneider1
1
Coinfection Unit, Priority Area Infections, Research Center Borstel, Borstel 2Viral Zoonosis and
Adaptation, Heinrich Pette Institute, Hamburg, Germany
Mycobacterium tuberculosis (Mtb) can establish long-term infection in the host even in the presence
of an otherwise intact immune system. The problem with persistent infections like tuberculosis (Tb)
is that they usually have to be controlled in the host for decades. Coinfections acquired throughout
life may impact on the balance of the immune response and skew it into one direction that is
disadvantageous for the host. Influenza virus establishes pulmonary infection and is known to
impair host responses to secondary bacterial infections. Epidemiological data raise concerns about
a possible impact of influenza on patients with Tb. In an experimental coinfection model we study
the impact of an early influenza A virus (IAV) coinfection on mycobacterial control and the initiation
of the Mtb-specific adaptive immune response. We found that control of Mtb replication is impaired
shortly after IAV challenge whereas viral clearance is not hampered. Importantly, the long-term
control of Mtb after IAV coinfection differs between males and females, indicating a role for sexrelated factors. While female mice regain control over Mtb infection after 7 weeks, males are unable
to restrict mycobacterial replication and show severely increased lung pathology after IAV
coinfection. Our goal is to elucidate the influenza-induced changes in the lung and to identify sexdependent factors that render males more susceptible to coinfection than females.
300
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
274
Host-responses to mycobacteria /interaction
22/09/2016
Mycobacterium tuberculosis PE27 activates dendritic cells and contributes to Th1-polarized
memory immune responses during in vivo infection
W.S. Kim, J.S. Kim, H. Kim, K.W. Kwon, S.J. Shin
Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
A gradual understanding of the proline-glutamate (PE) and proline-proline-glutamate (PPE) families,
which compromise 10% of the coding regions in the Mycobacterium tuberculosis (Mtb) genome, has
uncovered unique roles in host–pathogen interactions. However, the immunological function of
PE27 (Rv2769c), the largest PE member, remains unclear. Here, we explored the functional roles
and related signaling mechanisms of PE27 in the interaction with dendritic cells (DCs) to shape the
T cell response. PE27 phenotypically and functionally induces DC maturation by up-regulating
CD80, CD86, MHC class I and MHC class II expression on the DC surface to promote the
production of TNF-a, IL-1B, IL-6, and IL-12p70 but not IL-10. Additionally, we found that PE27mediated DC activation requires the participation of mitogen-activated protein kinases (MAPKs) and
nuclear factor κB (NF-κB) signaling pathways. Interestingly, PE27-treated DCs directed naïve CD4+
T cells to secrete IFN-g and activate T-bet but not GATA-3. These DC also induced PE27-specific
IFN-g-producing memory T cells in Mtb-infected mice indicating that PE27 contributes to Th1polarization. Taken together, these findings suggest that PE27 possesses Th1-polarizing potential
through DC maturation and could be useful in the design of TB vaccines.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
301
275
Host-responses to mycobacteria /interaction
22/09/2016
Blockade of the Kv1.3 K+ channel enhances BCG vaccine efficacy by expanding central
memory T lymphocytes
D.K. Singh1-2, V.P. Dwivedi2, A. Ranganathan2-1, G. Das2
1
Recombinant Gene Products Group, ICGEB 2Center for Molecular Medicine, Jawaharlal Nehru
University, New Delhi, India
Tuberculosis (TB) is the oldest known infectious disease, yet there is no effective vaccine against
adult pulmonary TB. Emerging evidence indicates that T helper (Th)1 and Th17 cells play important
roles in host protection against TB. However, TB vaccine efficacy in mice is critically dependent on
the balance between antigen-specific central memory T (Tcm) to effector memory T (Tem) cells.
Vaccine efficacy is determined on the generation of Tcm cells, which are a continuous source of
Tem cells. BCG induces strong Th1 effector cell responses and these cells function effectively in
neonates. However, over time these cells become gradually exhausted by recurrent exposure to the
pathogen, rendering the host again susceptible to infection. Therefore, to provide the host with a
continuous supply of Tem cells, a large pool of Tcm cells is needed. The generation of Tem cells
increases with the severity of bacterial pathogenesis and these cells rapidly produce copious
amount of IFN-γ upon antigenic challenge. Tem cells, however, are terminally differentiated effector
cells with low or no proliferative capacity. Therefore, maintenance of long-term protective memory
responses is thought to rely on Tcm cells with high proliferative capacity. Thus, increasing the pool
of Tcm (CD4+CD62LhiCD44hi) cells by concomitant regulation of Tem (CD4+CD62LloCD44hi) cells
during vaccination may be an effective strategy to develop a vaccine that induces long-lasting and
robust recall responses. In our study we found that pharmacological inhibition of Kv1.3, a potassium
channel preferentially expressed by Tem cells, with clofazimine selectively expands Tcm cells
during vaccination with Bacillus Calmette Guerin (BCG) which, therefore, have the potential to
continuously replace effector T cells at the site of infection. Specifically, a high Tcm to Tem cell ratio
is essential for optimal vaccine efficacy. Further, animals that received clofazimine following BCG
vaccination exhibited significantly enhanced resistance against TB as well as robust Th1 and Th17
responses. This superior activity could be transferred to naïve syngeneic animals by CD4+ T cells.
Thus, Kv1.3 blockade via inclusion of clofazimine during BCG vaccination is a promising approach
for enhancing the efficacy of the BCG vaccine in humans.
302
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
276 Host-responses to mycobacteria /interaction
22/09/2016
Host and Mycobacterium tuberculosis determinants of immune cells activation
J. Sousa7-8, H. Novais Bastos1-2-5, N. S. Osório1-2, M. Isabel Veiga1-2, H. Machado1-2, F. Cardoso1-2,
J. Gaifem1-2, A. Ramos4-6, T. Carvalho4-6, J. Vieira7-8, C. Vieira7-8, F. Rodrigues1-2, J. Tiago
Guimarães4-3-6, A. Gil Castro1-2, M. Saraiva7-8
1
Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of
Minho, Braga 2ICVS/3B’s-PT Government Associate Laboratory, Braga/guimarães 3Department of
Biochemistry, Faculty of Medicine, University of Porto 4Department of Clinical Pathology, São João
Hospital Center, Porto 5Department of Pneumology, São João Hospital Center, Porto 6EPIUnit Institute of Public Health, University of Porto 7i3S - Instituto de Investigação e Inovação em
Saúde 8IBMC, University of Porto, Porto, Portugal
A striking feature of tuberculosis (TB) is the variability of disease outcomes, ranging from pathogen
clearance or latency establishment to active disease. Immune imbalances clearly contribute to this
heterogeneity, assuming a rising importance as therapeutic targets. Several studies show a central
role of the cross-talk between host and bacteria in defining the immune response and the outcome
of disease. To underpin the molecular mechanisms regulating this cross-talk, we are studying a
cohort of 192 TB patients, showing different severity of disease at presentation. Thirty
Mycobacterium tuberculosis (Mtb) clinical isolates associated with different clinical severity of TB
were selected and PBMCs obtained from healthy IGRA+ donors (HD) or TB patients infected.
Cytokine production by the infected cells was then measured. Independently of the host genetics,
we identified two distinct groups of Mtb isolates: high vs low inflammatory triggers. Although a
straight relation of the cytokine profile with TB severity was not found, PBMCs from patients
responded differently than those from HD. These results show that although the profile of PBMC
activation is Mtb-dependent, host factors can also contribute to different disease outcomes.
Resorting to whole-genome and functional analysis, we are now investigating the host and
pathogen determinants underlying the observed differences. Our findings highlight the importance
of considering both host- and bacteria-associated characteristics when designing immunemodulatory interventions for TB.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
303
277 Host-responses to mycobacteria /interaction
22/09/2016
Therapeutic targeting of PPM1A to eliminate macrophages persistently infected with
Mycobacterium tuberculosis
K. Schaaf1, S.R. Smith1, O. Kutsch1, J. Sun2
1
Medicine 2Microbiology, University of Alabama at Birmingham, Birmingham, United States
The ability to suppress host macrophage apoptosis is essential for M. tuberculosis (Mtb) to replicate
intracellularly and evade the cell-mediated immune response. Thus, reversal of the anti-apoptotic
response during Mtb infections would have therapeutic benefits. However, a complete
understanding of the host factors responsible for the signaling pathways that control apoptosis
during Mtb infection remains elusive. We recently described that Mtb infection upregulates
expression of the host phosphatase PPM1A, which reduces the antibacterial and antiviral response
of macrophages1. Here we established a central role for PPM1A in the control of macrophage
apoptosis during Mtb infection. Overproduction of PPM1A protected macrophages from etoposide
(intrinsic) and tumor necrosis factor-α (extrinsic) induced apoptosis, and more importantly
suppressed macrophage apoptosis during Mtb-infection. Conversely, targeted depletion of PPM1A
by shRNA restored the ability of Mtb-infected macrophages to undergo apoptosis. Kinome analysis
and subsequent studies revealed that PPM1A-controlled inhibition of apoptosis is mediated by a
mechanism that involves abrogation of the c-Jun N-terminal kinase (JNK) activity. Accordingly,
activation of JNK by subtoxic concentrations of anisomycin restored the ability of macrophages to
undergo apoptosis, and most importantly, induced selective killing of primary human macrophages
infected with Mtb. Our data indicate that therapeutic targeting of PPM1A or its downstream signaling
network could provide novel host directed therapies for the elimination of persistent bacterial
infections such as Mtb. This strategy could be particularly beneficial for Mtb/HIV co-infected patients
since we also describe that HIV-infected macrophages display upregulated PPM1A expression
leading to arrest of the cellular innate antiviral response and apoptosis pathways.
Reference:
1.
304
Sun J, Schaaf K, Duverger A, Wolschendorf F, Speer A, Wagner F, Niederweis M, Kutsch O.
Protein Phosphatase, Mg2+/Mn2+-dependent 1A controls the innate antiviral and antibacterial
response of macrophages during HIV-1 and Mycobacterium tuberculosis infection. Oncotarget
2016.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
278 Host-responses to mycobacteria /interaction
22/09/2016
Immature granulocytes are increased in early clearance of Mycobacterium tuberculosis
infection
A. Verrall5, L. Apriani1, K. Sharples2, J. Ussher3, A. Indrati1, A. Nurani1, E. Diandini1,
B. Alisjahbana1, R. Van Crevel6, P. Hill4
1
TB-HIV Working Group, Universitas Padjadjaran, Bandung, Indonesia 2Department of
Medicine 3Department of Microbiology and Immunology 4Department of Preventive and Social
Medicine, Dunedin 5Department of Pathology and Molecular Medicine, University of Otago,
Wellington South, New Zealand 6Department of Internal Medicine and Radboud Center for
Infectious Diseases, Radboudumc, Nijmegen, The Netherlands
Background: Early clearance (EC) is the eradication of inhaled Mycobacterium tuberculosis (Mtb)
by innate mechanisms before an adaptive immune response develops. Understanding EC could
lead to novel Mtb vaccines or host directed therapies. Neutrophils may have a protective role in the
early response to Mtb infection. Hence, we tested the hypotheses that peripheral blood total
neutrophil and immature granulocyte (IG) counts are associated with remaining uninfected after
household exposure to a smear positive tuberculosis (TB) case.
Methods: A study of household contacts of TB cases in Indonesia recruits contacts within 2 weeks
of the case starting treatment. Mtb infection is determined by Interferon Gamma Release Assay
(IGRA) at baseline (and if negative, again at 14 weeks). At baseline the duration and intensity of
exposure to the index case is recorded, a full blood count is performed using a Sysmex XE 5000
analyser, and other specimens are archived. We compared neutrophils and IG in contacts who
remained persistently IGRA negative, a group that contains a high proportion of ECs, to susceptible
contacts who converted from negative at baseline to positive at 14 weeks. Adjusted risk ratios
(ARR) for IGRA conversion were estimated using generalised estimating equations that accounted
for confounders and the clustered nature of household data.
Results: Of 941 contacts, 545 (57.9%) were IGRA positive at baseline, 222 (23.6%) were
persistently IGRA negative, and 75 (8.0%) converted. Immature granulocytes were detected in 39%
of persistently IGRA negative contacts compared to 31% of converters. The median neutrophil
count was 3.95 (IQR 3.3 - 5.0) for persistently IGRA negative contacts and 4.05 (Inter Quartile
Range (IQR): 2.9 - 5.1) for converters. IGRA conversion was independently associated with lower
IG (P=0.003) but the relationship of lower neutrophils with conversion was not statistically significant
(P=0.06). The relative risk of conversion decreased sharply as IG and neutrophils increased (figure).
Conclusion: Immature granulocytes are associated with EC in this cohort of Mtb exposed case
contacts. We will elaborate key elements of this signal through RNA sequencing and flow cytometry
sub-studies that are currently underway.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
305
279 Host-responses to mycobacteria /interaction
22/09/2016
Identification of genes required for BCG survival in bovine lymph nodes.
K. Williams3, B. Villarreal-Ramos4, H. Wu3, S. Manjari2, A. Prasad2, M. Veerasami1, G. Stewart3,
M. Vordermeier4, J. Mcfadden3
1
Tamil Nadu Veterinary and Animal Sciences University (TANUVAS), Chennai 2Centre for
Development of Advanced Computing (C-DAC), Pune, India 3Faculty of Medical and Health
Sciences, University of Surrey, Guildford 4Animal and Plant Health Agency (APHA), Weybridge,
United Kingdom
Although bovine tuberculosis is a major global problem, BCG vaccination is not used to control
bovine tuberculosis, despite its proven efficacy in cattle, as vaccination interferes with the current
diagnostic PPD test. This project aims to generate a new recombinant BCG strain and
complementary diagnostic test, which would allow the vaccination of cattle to greatly reduce bovine
tuberculosis prevalence.
Our approach:
1. Identify genes required for in vivo survival of BCG by Tn-seq
2. Remove several antigens from BCG not required for in vivo survival (new vaccine)
3. Construct protein cocktail comprising these antigens (new skin test)
A saturated BCG Danish Tn library was constructed and injected into the prescapular lymph nodes
of 3 cows. After 3 weeks the 6 lymph nodes were harvested and Tn mutants recovered. Tn-seq data
from input and output libraries was analysed by TRANSIT. Known skin test antigens (such as
MPB70 & MPB83) were found not to be essential for in vivo survival, validating the removal of these
antigens from BCG for our new vaccine. Conversely, BCG genes found to be essential or
attenuated in the bovine lymph node model could represent genes important for host survival.
306
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
280 Host-Pathogen interaction and beyond
22/09/2016
The expired microbiome sampled by use of face masks
M. Abdulwhhab, M. Barer, C. Williams, K. Haldar, M. Ramsheh, P. Haldar, N. Garton
Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
We have been evaluating the use of modified face masks as a means of sampling the microbial
content of exhaled air particles. We are using this approach to study respiratory diseases such as
tuberculosis, pneumonia and chronic obstructive pulmonary disease. We have shown that the
approach is effective in detecting tubercle bacilli in TB patients (PLoS One 9:e104921) and are now
assessing its value in providing samples from the lower airways in other conditions.
We report here our preliminary results from a study of over 100 samples from healthy volunteers
and consenting patients with respiratory disease. Subjects were invited to wear adapted face masks
for periods of up to 1 hour while undertaking specified respiratory efforts including coughing, normal
breathing, instructed tidal breathing and reading out loud. As with our TB study, samples were
acquired on gelatine filters from which we then obtained extracts for DNA and protein analysis.
Bacterial 16SrDNA-directed analyses indicate contamination rates of between 104 and 106 copies
per hour of phyla including Firmicutes, Gamma- and Beta-proteobacteria, Actinobacteria and
Bacteroidetes. Evidence of mask contamination with particles from the lower and upper respiratory
tracts has been obtained from these phylogenetic assignments and from quantifying surfactant
protein A and albumin. Mask sampling provides important new opportunities to characterise the
microbial content of human expired air in tuberculosis and other lung infections.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
307
281 Host-Pathogen interaction and beyond
22/09/2016
Deciphering the role of shape regulators in growth and survival of mycobacteria
D. Arora2, Y. Chawla2, B. Malakar2, A. Singh1, V. Nandicoori2
1
CSIR-IGIB 2Signal Transduction Laboratory-I, National Institute of Immunology, New Delhi, India
Establishment of successful bacterial infection in host requires undeterred cell growth and division.
Mycobacterial cell wall, the prime barrier protecting the cell from immune assault undergoes
substantial remodeling events during its survival in host. How mycobacteria regulate cell shape
maintenance during this process is an unexplored facet. Continuous cell integrity and shape
maintenance will require coordination of biosynthetic, structural and regulatory elements of cell
division. Studies on genes involved in maintaining bacterial cell shape during mycobacterial division
are scant. Here, we report the role of mycobacterial shape regulators genes in the growth and
survival of the pathogen in vitro and in host. Overexpression and deletion studies suggests critical
role for these genes in maintaining normal cell morphologies. Furthermore, our studies delineate the
significance of the previously uncharacterized active domains of these proteins and suggest
a phosphorylation-mediated regulation in conserving their normal activity. Animal infection studies
further suggests critical role for these genes in survival of the pathogen in host. Together, our data
indicate that these shape regulators are required for mycobacterial cell fitness during host-induced
stress.
308
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
282 Host-Pathogen interaction and beyond
22/09/2016
Breaking fat!
How mycobacteria become obese in Dictyostelium mutants lacking lipid droplets
C. Barisch, T. Soldati
University of Geneva, Geneva, Switzerland
Lipid droplets (LDs) store energy in form of neutral lipids and exist in virtually every cell type.
Induction of foamy, LD-filled macrophages is a general host response to mycobacterial infection and
develops shortly after granuloma formation. Intracellular mycobacteria have been seen in the
vicinity or even “plunging” inside LDs, possibly corroborating that the bacteria are able to exploit
host lipids for their own metabolism. The mechanism by which the bacteria get access to LDs is still
unknown.
Using the Dictyostelium/Mycobacterium marinum infection system as a model for foamy
macrophages, we have found that M. marinum exploits lipids from host LDs to build up its own lipid
inclusions (ILIs). Strikingly, the Dictyostelium homologue of perilipin and the murine perilipin 2
surround bacteria that had escaped to the cytosol of Dictyostelium or microglial BV-2 cells,
respectively. In addition, bacterial growth was inhibited in perilipin mutants.
To test which lipids are preferentially transported into the mycobacterium-containing compartment,
we interfere with host enzymes involved in triacylglycerol (TAG) synthesis. Interestingly, the Long
Chain Fatty Acid CoA Synthase 1, that activates fatty acids (FAs), is recruited to the mycobacterium
niche. In addition, LDs decorated with diacylglycerol acyltransferase (Dgat) 2 were seen clustering
around cytosolic bacteria at late infection stages.
Dictyostelium Dgat mutants are unable to generate TAGs and LDs. Instead, the exogenous FAs are
esterified into phospholipids, inducing uncontrolled proliferations of the ER-membrane. Strikingly, M.
marinum is able to exploit this source of host phospholipids resulting in rapid reversal of ERproliferations. Moreover, the bacteria build up many more ILIs compared to the wild type. This
excessive “fattening” is not accompanied by a change in intracellular growth and metabolic activity,
and thus provides evidence that the storage of neutral lipids does not necessarily induce dormancy.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
309
283 Host-Pathogen interaction and beyond
22/09/2016
Cloning, expression and characterization of Mycobacterium tuberculosis antigen, HBHA,
produced in the yeast Pichia pastoris
C. Benabdessalem2, R.Y.M. Ouni2, C. Rouanet1, F. Haj Aissa2, M. Loyens1, D. Raze1, C. Locht1,
R. Barbouche2
1
INSERM U1019 - CNRS UMR 8204, Univ Lille Nord de France, CIIL - Center for Infection and
Immunity of Lille, Institut Pasteur de Lille, Lille, France 2Laboratoire de Transmission, Contrôle et
Immunobiologie des Infections, Institut Pasteur de Tunis, Tunis, Tunisia
Identification and preventive treatment of latently infected cases (LTBI) with Mycobacterium
tuberculosis (Mtb) are currently essential components of the global Post-2015 tuberculosis (TB)
strategy. Since commercially available Interferon Gamma Release Assays (IGRAs) cannot, alone,
discriminate active TB from LTBI individuals it will be crucial to identify alternative antigens to use
for IGRA. The test HBHA-IGRA is a novel test that shows a high specificity for LTBI. This assay is
based on the HBHA (Heparin Binding Haemagglutinin Antigen) a latency associated antigen of Mtb
which induces a strong IFNg response during latent but not active TB. It has been shown that the
methylation on its C-terminal domain is crucial for an effective T cell response to this antigen. The
current HBHA-IGRA is based on the use of the native antigen purified from BCG strain since the
conventional expression system, E.coli, is not able to carry out methylation.
The development of a recombinant methylated form of this antigen would be crucial for the
commercialization of HBHA-IGRA. Herein we used the yeast Pichia (P.) pastoris to express a
recombinant form of the HBHA. Indeed, P. pastoris similar to Mtb has a GC rich codon usage and
also retains protein post-translational events, which may better mimic native antigens. In previous
studies we showed that P. pastoris could be a good alternative expression system for mycobacterial
genes. Three, already developed monoclonal antibodies (MoAbs), two of which are specific for the
methylation of the native form of HBHA were used to recognize the produced recombinant form as
well.
Using shaker flasks, we generated a P. pastoris clone expressing a recombinant form of HBHA
(rHBHA). Interestingly, we showed that the rHBHA is methylated since it is well recognized by both
methylation specific MoAbs. We are studying by mass spectrometry whether the methylation
performed by the yeast is similar to mycobacterial one.
310
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
284 Host-Pathogen interaction and beyond
22/09/2016
The initial encounter of pathogenic mycobacteria with tissue resident macrophages
determines infection outcome
C. Cambier2, L. Ramakrishnan1
1
University of Cambridge, Cambridge, United Kingdom 2Chemistry, Bertozzi, Stanford, Stanford,
United States
Despite the notoriety of tuberculosis as a human killer, epidemiological studies suggest that some
exposed individuals can clear tuberculosis independently of adaptive immune responses. Using the
zebrafish, we provide direct mechanistic evidence for early clearance of infection, and show that it is
mediated by the innate microbicidal activity of tissue-resident macrophages. We find that resident
macrophages are first-responders to bacterial pathogens, including mycobacteria. However, virulent
mycobacteria deploy an escape strategy after phagocytosis by these microbicidal cells. This escape
requires mycobacterial phenolic glycolipid (PGL), which programs the infected resident
macrophages to recruit permissive monocytes, into which the mycobacteria transfer. Thus infection
outcome - clearance versus progression - depends on the relative strengths of two competing
programs in the resident macrophage - intrinsic microbicidal activity versus PGL-induced
recruitment of permissive monocytes. Our findings suggest immunological or pharmacological PGLblocking strategies as interventions to prevent tuberculosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
311
285 Host-Pathogen interaction and beyond
22/09/2016
Mycobacterium tuberculosis LppM displays an original structure and domain composition
linked to a dual localisation
M. Cohen-Gonsaud
Centre de Biochimie Structurale, Montpellier, France
Mycobacterium tuberculosis (Mtb) encodes several bacterial effectors impacting the colonization of
phagocytes. LppM (Rv2171) is both implied in phagocytosis and able to efficiently block
phagosomal acidification in the macrophage, two key features contributing to Mtb persistence. We
showed that LppM is anchored to the mycobacterial cell wall by a C-terminal membrane domain.
But the protein also exists as a truncated protein secreted into the culture medium. The LppM
solution structure we solved here displays no similarity with other Mtb lipoproteins also involved in
phagosomal maturation (ie LprG). In addition, we demonstrated the protein is able to bind scarce
phosphatidylinositol mannosides derivatives, molecules involved in the host processes
manipulation. Therefore, our data demonstrate a dual localization and open new perspective on Mtb
secretion and protein localization regulation.
312
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
286 Host-Pathogen interaction and beyond
22/09/2016
Developing a persistent zebrafish embryo infection model
S. Commandeur1, N. Iakobachvili2, M. Nur2, G. Mukamolova2, A. Van Der Sar1, W. Bitter1
1
Department of Medical Microbiology and Infection Control, VUmc, Amsterdam, The
Netherlands 2Department of Infection, Immunity, and Inflammation, University of Leicester,
Leicester, United Kingdom
Mycobacterium tuberculosis (Mtb) is able to reside in the host without inducing any clinical
symptoms of the disease. A so-called latent tuberculosis (TB) infection can be present for decades.
Mtb is able to adapt to the harsh environmental conditions it encounters in the host, resulting in a
persistent phase of Mtb. Such persisters are phenotypically drug resistant and require long-term
treatment of a combination of antibiotics. The rise of multidrug-resistant (MDR) and extensively
drug-resistant (XDR) Mtb is fueled by this long-term therapy and thus dramatically shortening of the
length of TB treatment is urgently required. New drugs are needed that directly target the persister
population to reach this goal.
In this work we develop a model that can be used to identify compounds that efficiently kill such
persistent mycobacteria. We use Mycobacterium marinum (Mm) and the zebrafish embryo host as
model system because (i) Mm infection of zebrafish embryos induces similar pathology as observed
for Mtb infection in humans and (ii) it is useful for high throughput screens in search for novel
compounds.
Because latent infections cannot be reached in the zebrafish embryo model due to time constraints,
we have to establish the bacterial persistent phase in vitro prior infection. To create persistent Mm
we analyzed whether Mm is able to adapt to in vitro stress conditions and identified that carbon-rich
starvation conditions leads to a persister phase in Mm. However, the persister phenotype was not
maintained in vivo. Mtb expresses resuscitation promoting factors (Rpfs) which are described to aid
in resuscitation of persister mycobacteria. We hypothesized that Rpf knock-outs will prevent outgrowth in the zebrafish embryos. Single and double Rpf knock-outs were generated and analyzed
for their growth abilities after persister formation both in vitro and in vivo.We conclude that Mm is
able to adapt to stress conditions and that the absence of Rpfs seem to play a role in maintaining
the persister phenotype in the zebrafish embryo model.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
313
287 Host-Pathogen interaction and beyond
22/09/2016
Mycobacterium tuberculosis survival upon efferocytosis of infected neutrophils by
macrophages requires ESAT-6
T. Dallenga1-2, U. Repnik4, J. Eich2, G. Griffiths4, R. Reimer3, U.E. Schaible1-2
1
German Centre for Infection Research 2Cellular Microbiology, Research Center Borstel,
Borstel 3Core Facility Microscopy & Image Analysis, Heinrich-Pette-Institute, Hamburg,
Germany 4Department of Biosciences, University of Oslo, Oslo, Norway
Extensively and multidrug-resistant isolates of Mycobacterium tuberculosis are on the rise
worldwide. Novel host-directed therapies are promising adjunct measures to antibiotics treatment.
Neutrophils represent the main infected cell population in sputum and bronchoalveolar lavages from
patients with active tuberculosis, but fail to kill M. tuberculosis. Instead, virulent but not RD1deficient mycobacteria induce necrotic cell death in a reactive oxygen species-dependent manner,
facilitating mycobacterial escape from killing. Dead neutrophils are removed by macrophages in a
process termed efferocytosis. We studied the role of efferocytic removal of mycobacteria-infected
apoptotic vs. necrotic neutrophils in macrophage defense against M. tuberculosis. We found that
efferocytosis of necrotic neutrophils infected with wild type M. tuberculosis promoted survival and
growth of the mycobacteria, whereas mutants lacking either RD1 or the RD1-encoded ESAT-6
alone failed to grow in efferocytes. In contrast, mycobacteria directly entering macrophages required
other RD1-encoded genes for intracellular growth, additionally to ESAT-6. The decision whether
efferocytosed M. tuberculosis survived in macrophages was determined by differential intracellular
trafficking and cell death pathways upon infection by either wild type or attenuated mutant strains.
By combined time-lapse live cell imaging, correlative confocal-based 3D-reconstruction and electron
microscopy we monitored infection, cell death and efferocytosis. Data suggests differential
phagosomal sorting of dead PMN material from Mtb within macrophages early after uptake. Taken
together, efferocytosis of neutrophils infected with virulent M. tuberculosis promotes infection in an
ESAT-6-depending manner. Our data further emphasize the detrimental role of neutrophils for
tuberculosis pathology and disease outcome, making them a predestined candidate for both, point
of care testing as well as target for host-directed therapies.
314
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
288 Host-Pathogen interaction and beyond
22/09/2016
Regulation of phagocyte triglyceride by a STAT-ATG2 pathway controls mycobacterial
infection
C. Péan2, M. Schiebler1, S. Tan2, K. Brown1, M. Maserumule1, S. Menezes3, M. Pilátová3,
K. Bronda3, P. Guermonprez3, B. Stramer3, A. Floto1, M. Dionne2
1
University of Cambridge, Cambridge 2Dept of Life Sciences and MRC CMBI, Imperial College
London 3King's College London, London, United Kingdom
Macrophages are essential to the host defense against mycobacteria; however, macrophages are
often inefficient at killing virulent mycobacteria and constitute a niche for mycobacterial persistence
and proliferation. We use a Drosophila model to identify host mechanisms that regulate
mycobacterial growth within phagocytes. We show that, in this model, mycobacterium-induced
cytokine-STAT signaling is detrimental to the host, reducing resistance to Mycobacterium marinum
partly by reducing Atg2 expression and thus facilitating pathogen-induced deregulation of lipid
droplets. Flies with cytokine-STAT signaling inhibited live longer when infected and exhibit markedly
lower mycobacterial burden and improved macrophage survival; this is accompanied by significant
and specific overexpression of Atg2. Atg2 overexpression is sufficient to reduce proliferation of
mycobacteria in Drosophila phagocytes. Increased Atg2 appears not to affect bulk autophagy;
instead, Atg2 overexpression partially normalizes lipid deposition in infected phagocytes. Direct
inhibition of macrophage triglyceride biosynthesis likewise reduces intracellular lipid droplets and
mycobacterial numbers. Similarly, in human macrophages, IL-6 reduces ATG2A levels and
contributes to mycobacterial growth by deregulating macrophage lipid metabolism. Our results
reveal a new mechanism by which cytokines control lipid droplet homeostasis and consequently
resistance to mycobacterial infection. That inhibition of macrophage triglyceride synthesis reduces
mycobacterial growth and the identification of ATG2 as a key cytokine effector highlight potential
host-directed therapies in tuberculosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
315
289 Host-Pathogen interaction and beyond
22/09/2016
Peptidyl prolyl isomerase family of Mycobacterium tuberculosis mediates the modulation of
host macrophages aiding the infection process
N. Dubey2, M. Gupta1, A.K. Sharma1, Y. Singh2
1
Institute of genomics and integrative biology 2Department of Zoology, University of Delhi, Delhi,
India
Mycobacterium tuberculosis (Mtb), the etiological agent of the disease tuberculosis can evade the
host immune system to either develop an active infection or to stay in the latent form for years. This,
in part, is achieved by the secretion of virulence factors which not only assist in initial invasion but
also in the progression of the disease towards the granulomatous form. The focus of our study is
mycobacterial secretory proteins Peptidyl prolyl isomerases (PPiases) and we are trying to
determine their role in mycobacterial pathogenesis. PPiases mediate the inter-conversion of cisand trans- conformers of protein, conferring the stability and activity to the cellular proteome. Mtb
encodes two PPIases, PPiA and PPiB. Though the family executes many housekeeping functions,
recent literature suggests a probable role of PPiases as potent virulence factors in many pathogens
such as Listeria, Salmonella and Brucella. Interestingly, Mtb PPiA is a secretory isomerase and its
secretion is restricted to the pathogenic species of Mycobacteria genus. Also, the transcriptional upregulation of Mtb PPiA is observed in intra-phagosomal bacteria. Our studies with the gene deletion
strain of PPiA in H37Rv strain indicate towards its role during the late time point of mice infection. To
explore the mechanism of action of PPiA, we have utilized in-silico tools to identify the novel
interacting partners of Mtb PPiA in the host. These targets were further confirmed by co-immuno
pull down and ELISA based interaction assays. Two interacting partners that we chose to study are
cell surface receptor protein Integrin and an innate immune response protein, Lipocalin. While
Integrins mediates the extracellular matrix modulations via matrix metallo-proteases, Lipocalins are
host siderocalins, which are known to inhibit the iron translocation required for bacterial survival by
competing with the Mtb carboxymycobactins. Our study, (i) elaborates the role of Mtb PPiA in
employing Integrin as a strategy for host invasion (ii) the implication of PPiA & lipocalin interaction
on intracellular bacterial iron requirements.Mtb PPiB is a membrane bound protein which undergoes
mannosylation. Its role during bacterial phagocytosis by host is also being studied.
This study, for the first time, delineates the functional significance of the PPiase family during
tuberculosis pathology. Our research on Mtb PPIases will provide us better understanding of the
cues utilized by the bacteria to evolve over its host.
316
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
290 Host-Pathogen interaction and beyond
22/09/2016
Pst/SenX3-RegX3 directly regulates ESX-5 secretion in Mycobacterium tuberculosis
S. Elliott, A. Tischler
University of Minnesota, Minneapolis, United States
The Type VII secretion system ESX-5 has been implicated in Mycobacterium tuberculosis (Mtb)
virulence, though both the regulatory mechanism and the signal inducing ESX-5 activity were
unknown. We recently discovered that ESX-5 is activated during phosphate (Pi) limitation, an
environmental signal Mtb likely experiences during infection. ESX-5 activity is directly regulated at
the transcriptional level by the Pst/SenX3-RegX3 system, which controls gene expression in
response to Pi availability. Pi limitation resulted in RegX3-dependent overexpression of esx-5
transcripts, as well as overproduction and hyper-secretion of ESX-5 proteins. We propose that
regulation of ESX-5 activity in response to Pi limitation is a critical function of the Pst/SenX3-RegX3
system that promotes Mtb virulence. Deletion of pstA1, which encodes a Pst component, causes
constitutive activation of RegX3, hyper-secretion of ESX-5 substrates, and attenuation in the mouse
aerosol infection model. We hypothesize that disruption of ESX-5 regulation is responsible for
attenuation of the ΔpstA1 mutant. Since EsxN is one of the hyper-secreted ESX-5 substrates and is
highly antigenic, we tested whether deletion of esxN could reverse the ΔpstA1 virulence defect. We
infected C57BL/6 and NOS2-/- mice with a ΔpstA1ΔesxN mutant, and evaluated bacterial replication
and persistence in the lungs. The ΔpstA1ΔesxN mutant did not reverse the virulence defect,
suggesting that hyper-secretion of all, or some combination of, ESX-5 substrates causes
attenuation. To further investigate this possibility, we first determined the precise regulatory
mechanism governing ESX-5 secretion. We previously demonstrated that RegX3 directly binds to a
region within the esx-5 locus. Using electrophoretic mobility shift assays (EMSA), we screened a
series of unlabeled competitor probes and identified a direct repeat sequence that is essential for
RegX3 binding. Subsequent mutation of this region to evaluate the importance of RegX3-dependent
regulation of ESX-5 secretion for Mtb virulence is currently in progress.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
317
291 Host-Pathogen interaction and beyond
22/09/2016
Mycobacterium tuberculosis subverts autophagosome formation by inducing miR-155
expression in human dendritic cells
M.P. Etna3, A. Sinigaglia2, A. Grassi2, E. Giacomini3, M. Pardini3, M. Severa3, M. Cruciani3,
A. Romagnoli4, L. Barzon2, B. Di Camillo2, G.M. Fimia4-1, R. Manganelli2, E.M Coccia3
1
Department of Biological and Environmental Science and Technology, University of Salento,
Lecce 2Department of Molecular Medicine, University of Padua, Padua 3Department of Infectious,
Parasitic and Immune-mediated diseases, Istituto Superiore di Sanità 4National Institute for
Infectious Diseases "L. Spallanzani”, Rome, Italy
Given the re-emergence of tuberculosis (TB), increasing efforts are devoted to understand the
events occurring along Mycobacterium tuberculosis (Mtb)-host interaction, w ith a partic ular interest
for the strategies engaged by Mtb to escape immune response.
In this study, we sought to characterize the interaction of Mtb w ith human dendritic cells (DC), a key
cell type involved in the regulation of innate and adaptive immune responses against pathogens. In
particular, we analyzed autophagy, as an immune process exploited by Mtb to evade the host
antimicrobial response and, in this context, we focused on microRNAs (miRNAs), whose role in
controlling the Mtb-DC interplay remains unclear.
Microarray analysis w as conducted at different time points after the Mtb infection thus unveiling the
modulation of a specific pattern of cellular miRNAs. Among cellular miRNAs de-regulated in Mtbinfected DC, w e analy zed the function of miR-155 for which target predic tion analysis identified as a
putative target Atg3, the E2 ubiquitin-like-conjugating enzyme involved in LC3-lipidation.
Interestingly, we observed that at late time points of infection Mtb impairs both ATG3 protein levels
and the number of LC3 puncta per cell, thus supporting the hypothesis of a miR-155-dependent
mechanism for Atg3 regulation.
Accordingly, dual luciferase 3’UTR reporter assay demonstrated that miR-155 dow n-regulated the
3’UTR of Atg3. All these observations are in line w ith the reduction of ATG3 protein level detected in
miR-155-transfected DC and, as a consequence, w ith a poor conversion of LC3 in its
autophagosome-associated form.
These data further underlined the importance of autophagy in controlling the fate of Mtb infection,
indeed in addition to the well-characterized block of the autophagic flux mediated by Mtb, here we
illustrated how this pathogen takes advantage from cellular miRNA regulation to impair
autophagosome formation thus escaping host immune response. These evidences provided new
intriguingly information on host pathogen interaction involved in Mtb-driven control of autophagy flux
to favor its own intracellular survival and persistence in the host.
This w ork was supported by grant RF-2010235199 from the Italian Ministry of Health (to EMC).
318
Tubercul osis 2016 – September 19-23, 2016 – Paris, Franc e
292 Host-Pathogen interaction and beyond
22/09/2016
Mycobacterium avium interfering with phagosome maturation evades an antibacterial
program activated by phagolysosomal TLR7/8/MyD88 in human primary macrophages
A. Gidon, S. Åsberg, C. Louet, L. Ryan, T. Flo
Centre of Molecular Inflammation Research, and Dept of Cancer Research and Molecular
Medicine, Norwegian University of Science and Technology, Trondheim, Norway
In developed countries the prevalence of non-tuberculous mycobacterial infections caused by M.
avium (Mav) is increasing in individuals who are immunocompromised due to underlying disease or
use of immunosuppressant drugs. Mav lacks several of the key virulence factors of Mtb, but
nevertheless can establish chronic infections. Pathogenic mycobacteria reside in macrophages and
are thought to avoid lysosomal targeting and degradation through poorly understood mechanisms
involving secreted phosphatases and cell wall lipids. We have previously demonstrated that Mav is
associated with Rab11-related compartments accessible to transferrin four days post infection.
However, how trafficking of mycobacteria relates to inflammatory signalling is poorly characterized,
especially for Mav. Using high-resolution confocal imaging of human primary macrophages infected
with Mav we show evidence that mycobacterial phagosomes are not arrested at an early endosomal
stage but rather progress to a late endosomal/lysosomal stage where inflammatory signalling is
initiated through TLR7/8 recruitment of MyD88 and nuclear translocation of NF-kB and IRF-1,
inflammatory cytokine production and Mav growth restriction. A fraction of the mycobacteria
escapes and re-establish in a less hostile "stealth" compartment, evading destruction, TLR detection
and growth restriction.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
319
293 Host-Pathogen interaction and beyond
22/09/2016
Role of VirS in acid induced responses of Mycobacterium tuberculosis
G. Khare, S. Singh, A.K. Tyagi
Department of Biochemistry, University of Delhi South Campus, Delhi, India
More than five decades of tuberculosis (TB) control programmes using potentially efficacious drugs
have not succeeded in reducing the burden of this disease in most parts of the world.
Mycobacterium tuberculosis is one of the most successful pathogens that effectively counteracts the
host antimicrobial responses and is able to reside in macrophages, the very cells involved in the
killing of the incoming pathogens. Phagosomal acidification is considered as the major host defense
mechanism against the microbial pathogens in addition to the production of reactive oxygen
intermediates (ROI) and reactive nitrogen intermediates (RNI). However, M.tuberculosis is endowed
with various properties that help its survival in the macrophages by subverting the phagosomal
maturation pathways. The ability of mycobacterium to resist the killing in these harsher acidified
conditions highlights the importance of maintaining its pH homeostasis for its long term persistence
in macrophages during infection, however, the underlying mechanisms for this are poorly
understood. Till to date, the identity of a specific sensor of acidic pH in M.tuberculosis is not known.
In this context, our laboratory and others have identified the specific induction of an AraC/XylS-type
transcriptional regulator VirS (Rv 3082c) in response to acidic pH. The fact that acidic conditions led
to its up-regulation suggested that the genes regulated by VirS (VirS-regulon) might be associated
with the maintenance of pH homeostasis in the cell. Further, the infection of guinea pigs with the M.
tuberculosis virS mutant was shown to result in an ~800 fold reduction in the splenic bacillary load.
The present study describes the role of VirS in the acid induced responses of M.tuberculosis by
employing the gene knockouts in in vitro broth cultures and macrophages. Purification of VirS was
carried out and the DNA binding region for VirS was determined by using EMSA and footprinting
studies. Moreover, microarray studies were carried out to identify the genes regulated by VirS in
response to acidic stress. The study further focused on understanding the gene regulation under
acidic conditions faced by the pathogen and the role of acid induced transcriptional regulator VirS in
this process.
320
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
294 Host-Pathogen interaction and beyond
22/09/2016
Imaging NADH-NAD+ redox homeostasis of Mycobacterium tuberculosis during infection
S. Bhatt, I. Iqbal, A. Kumar
CSIR-Institute of MIcrobial Technology, Chandigarh, India
The NADH:NAD+ ratio is the primary indicator of the metabolic state of bacteria. NAD(H)
homeostasis is critical for Mycobacterium tuberculosis (Mtb) survival and is thus considered an
important drug target, but the spatio-temporal measurements of NAD(H) remain a challenge. In this
study, we have generated reporter strains of fast and slow growing mycobacterium species for
measurement of the NADH:NAD+ levels in vitro and in vivo. Genetically encoded fluorescent
biosensors of the NADH:NAD+ ratios were recently described, paving the way for investigations of
the metabolic state of pathogens during infection. Here we have adapted the genetically encoded
biosensor Peredox for measurement of the metabolic state of Mtb in vitro and during infection of
macrophage cells. Using Peredox, here we show that inhibition of the electron transport chain,
disruption of the membrane potential and proton gradient, and treatment with antimycobacterial
drugs lead to the accumulation of NADH in mycobacterial cells. Interestingly, we observed that
activation of resting macrophages with interferon-gamma results in higher NADH:NAD+ levels in
resident Mtb cells. Furthermore, we observed that the sub-vacuolar localization dictates the
metabolic state of Mtb; Mtb cells residing in phagolysosomes have a higher NADH:NAD+ ratio and
Mtb cells colocalizing with the early endosome have lower NADH:NAD+. Thus in this study we report
novel engineered reporter strains of mycobacteria capable of measuring NADH:NAD+ redox
homeostasis in response to antimycobacterial drug and other physiologically relevant stresses.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
321
295 Host-Pathogen interaction and beyond
22/09/2016
Genome Wide Mutagenesis strategies in Dictyostelium discoideum and Mycobacterium
marinum to decipher the conserved genetic basis of mycobacteria intracellular infections
L. Lefrançois1, T. Mendum2, G. Stewart2, T. Soldati1
1
Dept of Biochemistry, University of Geneva, Geneva, Switzerland 2FHMS, University of Surrey,
Guildford, United Kingdom
This study aims at understanding how Mycobacterium bovis and Mycobacterium marinum
manipulate the fundamental processes of innate immunity, in particular the phagosome
environment. Because genetic analysis of host factors is difficult in diploid mammalian cells, we
propose to use Dictyostelium discoideum as a model phagocyte. This social amoeba exhibits
evolutionarily conserved mechanisms of cell-intrinsic defence and has a haploid genome opened to
forward genetics. Our plan is to develop the host-pathogen system D. discoideum-M. marinum as a
powerful genetically tractable model for mycobacterial infection.
To that purpose, we will apply genome-wide mutagenesis and high throughput sequencing.
Transposon mutagenesis (Tn-Seq) in M. marinum and M. bovis will be performed using the
MycoMar transposon. Restriction Enzyme-Mediated Insertional Mutagenesis (REMI-Seq) will be
performed in D. discoideum using a blasticidin resistance cassette. The complexity of the libraries
generated is about 100,000 mutants, with an insertion every 100bp in M. marinum and in 92% of
protein coding genes in D. discoideum. The precise identification and relative abundance of
insertions before and after a given selection allows us to quantitatively compare the compositions of
pools with a high dynamic range.
As a proof of principle, we are validating the approach by applying the Tn-Seq method for the first
time to M. marinum. Then, we want to test whether M. marinum can use similar carbon sources as
Mycobacterium tuberculosis, namely fatty acids and sterols, by performing selections on Tn-seq
pools in different media. In addition, we will also investigate whether accumulation of propionate
produced by cholesterol catabolism is toxic for M. marinum and can be alleviated by adding acetate.
Moreover, we will infect D. discoideum and macrophages with both M. marium and M. bovis Tn-seq
pools to identify genes required for survival in these two phagocytes. Finally, infection of D.
discoideum REMI-seq pools with M. marinum will reveal host genes implicated in resistance and
susceptibility to infection.
These promissing and innovative approaches will allow a comprehensive definition of the host and
pathogen genes important for the intracellular host-pathogen interactions and an overview of
mycobacterial virulence mechanims conserved during macrophages and Dictyostelium infection.
322
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
296 Host-Pathogen interaction and beyond
22/09/2016
Beyond macrophages: the role of lymphatic endothelial cells in tuberculosis
T. Lerner7, C. De Souza Carvalho-Wodarz2, U. Repnik3, M. Russell7, S. Borel7, C. Diedrich5,
M. Rohde1, H. Wainwright4, L. Collinson7, R. Wilkinson7-5-6, G. Griffiths3, M. Gutierrez7
1
Helmholtz Institute for Infection Research, Braunschweig 2Helmholtz Institute for Pharmaceutical
Research Saarland, Saarbrucken, Germany 3University of Oslo, Oslo, Norway 4UCT Faculty of
Health Sciences and NHLS Laboratories 5University of Cape Town, Cape Town, South
Africa 6Imperial College 7The Francis Crick Institute, London, United Kingdom
Lymphatic tuberculosis (TB) is the second most common manifestation of TB in humans but the role
of lymphatics in TB is poorly characterised. By analysing the localization of Mycobacterium
tuberculosis (Mtb) in lymph node samples from human TB patients, we identified lymphatic
endothelial cells (LECs) lining the lymphatic vessels as a niche for Mtb. In cultured primary human
LECs (hLECs), the uptake of mycobacteria via phagocytosis was mediated through the mannose
receptor. Moreover, Mtb formed distinctive intracellular cords within the cytosol, a process that
required the type VII secretion system ESX-1. Interestingly, these M. tuberculosis cords were not
recognised by the autophagy machinery. Surprisingly, by using correlative light electron microscopy
we demonstrated that Mtb also replicated in autophagosomes. However, upon activating hLECs
with interferon gamma (IFN-γ), the growth of the bacteria was restricted. This restriction of bacterial
growth was via the induction of autophagy and also via nitric oxide production. Thus, Mtb localises
in distinct sub-populations in LECs that differentially interact with the autophagy pathway resulting in
either Mtb growth or restriction. These findings highlight the importance of LECs in the pathogenesis
of tuberculosis, and implicate LECs as a potential reservoir for M. tuberculosis in humans.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
323
297 Host-Pathogen interaction and beyond
22/09/2016
ESX-1 exploits type I IFN-signaling to inhibit IFNg-mediated autophagy and growth
restriction of intracellular mycobacteria
J. Lienard1, E. Movert1, C. Valfridsson1, E. Sturegård2, F. Carlsson1
1
Experimental Medical Science, Immunology section, Host-pathogen interactions, Lund 2Laboratory
Medicine, Medical Microbiology section, Lund University, Malmö, Sweden
The ability of macrophages to eradicate intracellular pathogens is normally greatly enhanced by
IFNg, a cytokine produced mainly after onset of adaptive immunity. However, adaptive immunity is
unable to provide sterilizing immunity against mycobacteria, suggesting that mycobacteria have
evolved virulence strategies to inhibit the bactericidal effect of IFNg-signaling in macrophages. Still,
the host-pathogen interactions and cellular mechanisms responsible for this feature have remained
elusive. Our data suggest that the ESX-1 type VII secretion systems of Mycobacterium tuberculosis
and Mycobacterium marinum exploit type I IFN-signaling to promote an IL-12low/IL-10high regulatory
macrophage phenotype characterized by secretion of IL-10, IL-27 and IL-6. This mechanism has no
impact on intracellular growth in the absence of IFNg, but suppresses IFNg-mediated autophagy
and growth restriction, indicating that the regulatory phenotype extends to function. The IFNgrefractory phenotype is partly mediated by IL-27-signaling, establishing functional relevance for this
downstream cytokine. These findings identify a novel macrophage-modulating function for the ESX1 secretion system that might contribute to suppress the efficacy of adaptive immunity, and provide
mechanistic insight into the antagonistic cross talk between type I IFNs and IFNg in mycobacterial
infection.
324
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
298 Host-Pathogen interaction and beyond
22/09/2016
The anti-inflammatory role of C-type lectin DC-SIGN in human alternatively activated
macrophages and the response against Mycobacterium tuberculosis
A. Troegeler2, L. Balboa1, C. Lastrucci4, C. Duval2, I. Mercier2, A. Bénard2, T. Al Saati3,
R. Poincloux2, I. Kondova5, F. Verreck5, I. Maridonneau-Parini2, M.C. Sasiain1, C. Cougoule2,
O. Neyrolles2, G. Lugo-Villarino2
1
IMEX-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina 2Tuberculosis and
Infection Biology, CNRS, IPBS (Institut de Pharmacologie et de Biologie Structurale), UPS
(Université Paul Sabatier) 3INSERM/UPS/US006 CREFRE, CHU Purpan, Toulouse, France
4
EMBL/CRG, Barcelona Institute of Science and Technology, Barcelona, Spain 5Biomedical
Primate Research Centre, Rijswijk, The Netherlands
DC-SIGN is a C-type lectin receptor (CLR) that serves as a biological marker for human
alternatively activated macrophages, M(IL-4). Although DC-SIGN plays important roles in M.
tuberculosis (Mtb) interactions with dendritic cells, its contribution to the mycobacteria-macrophage
interaction remains poorly understood. Here, we investigate the pertinence of DC-SIGN in the
control of Mtb intracellular growth and inflammatory response by M(IL-4). We found that siRNAmediated inactivation of DC-SIGN renders M(IL-4) less permissible to Mtb intracellular growth
compared to DC-SIGN-expressing cells, despite the equal levels of the initial Mtb uptake.
Transcriptomic analysis in DC-SIGN-deficient M(IL-4) revealed a pro-inflammatory gene signature
characterized by up-regulation of IL-6, TNF and CXCL1, which was confirmed by RT-qPCR and
ELISA analyses. Conditioning of human monocytes with supernatants from Mtb-infected DC-SIGNdeficient M(IL-4) resulted in a shift towards a pro-inflammatory phenotype, supporting the role of
DC-SIGN in controlling macrophage inflammation. At the molecular level, we show that DC-SIGN
antagonizes the pro-inflammatory response mediated by Dectin-1. Treatment of M(IL-4) with
mannosylated lipoarabinomannan (an Mtb-derived glycolipid and ligand for DC-SIGN) induces
STAT3 activation in a DC-SIGN-dependent manner, suggesting a novel signaling pathway in the
DC-SIGN-Dectin-1 crosstalk. Finally, we demonstrate that DC-SIGN expression can be accentuated
in macrophages under different contexts using samples from TB patients, and become abundant in
TB pulmonary lesions of non-human primates, arguing for the pertinence DC-SIGN+ macrophages
in TB pathology. Collectively, this study highlights a dual role for DC-SIGN as, on the one hand,
being a host factor granting advantage for Mtb to parasitize macrophages and, on the other hand,
representing a molecular switch to turn off the pro-inflammatory response in these cells to prevent
potential immunopathology.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
325
299 Host-Pathogen interaction and beyond
22/09/2016
Growth of Mycobacterium tuberculosis in dead macrophages drives a host cell death
cascade
D. Mahamed2-3, M. Boulle2-3, S. Skroch2-3, L. Oom2-3, G. Sreejit2-3, K. Pillay2-3, O. Catinas2,
M. Naicker2-3, S. Rampersad2-3, C. Mathonsi2-3, J. Hunter2-3, A. Pym2-3, G. Lustig2-3, A. Sigal2-1
1
Max Planck Institute for Infection Biology, Berlin, Germany 2KwaZulu Natal Research Institute for
TB-HIV 3University of KwaZulu-Natal, Durban, South Africa
Active Mycobacterium tuberculosis (Mtb) infection leads to extensive host cell death and bacterial
growth, releasing Mtb into the airways to be transmitted to another host. Here we set out to
determine the factors contributing to bacterial growth and host cell death at the level of individual
macrophages, the primary Mtb host cells. Primary human macrophages, differentiated from
peripheral blood monocyes, were infected with a fluorescently labelled strain of Mtb H37Rv and
tracked using live cell time lapse microcopy. To facilitate quantitative analysis of complex
macrophage-Mtb interactions, custom written image analysis software was developed to track cell
fate. We quantified the number of bacteria in each pickup of Mtb by a macrophage, whether the
bacilli were cell-free or inside a dead infected cell, and the fate of the internalizing macrophage after
pickup. Macrophages internalizing clumps of Mtb died with a frequency which increased with total
Mtb uptake and number of Mtb in a single clump. Interestingly, macrophage death did not eliminate
intracellular bacteria. Instead, it led to rapid Mtb multiplication inside dead host cells, with a median
doubling time of 25±10 hours. Growth in dead infected cells was significantly faster than
extracellular Mtb growth (doubling time 38±11 hours), while live macrophages controlled Mtb
replication efficiently. Bacteria grew as a clump inside dead macrophages, creating a locally high
Mtb concentration. Internalization of these dead infected cells resulted in nearly 90% macrophage
death, indicating that once an infected cell dies, additional cycles of cell death will occur. Such a
cascade of host cell death provides a mechanism for the propagation of TB infection at the cellular
level which, once initiated, would be difficult to contain.
326
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
300 Host-Pathogen interaction and beyond
22/09/2016
Inhibition of matrix metalloproteinases interfere with the formation of tuberculosis
granuloma in a human lung tissue model
V.R. Parasa1, J. Muvva2, J. Jeronimo Frias Rose1, S. Brighenti2, M. Lerm1
1
Division of Medical Microbiology and Molecular Medicine, Linköping University, Linköping 2Center
for Infectious Medicne, Karolinksa University Hospital, Karolinska Institute, Stockholm, Sweden
Granulomas, organized cluster of immune and non-immune cells, are the hallmarks of pulmonary
tuberculosis (TB). Matrix metalloproteinases (MMPs), a family of proteases involved in extracellular
matrix destruction and tissue remodeling, have been implicated in the pulmonary pathology of TB,
specifically during extensive tissue damage and disseminated TB. However, the exact stage of the
disease at which the specific MMPs act in human TB is uncertain.
We hypothesize that MMPs are required for early granuloma formation and that by blocking the
activity of specific MMPs, the process of granuloma formation could be altered, thus perturbing the
initial growth of M. tuberculosis.
Studying TB at its initial stages, like granuloma formation, requires the assessment of early events
during infection in a three-dimensional system. Therefore, we developed an in vitro lung tissue
model [1,2] to investigate the role of MMPs during TB granuloma formation. Confocal microscopy
was used for the visualization of granulomas. Analysis of MMPs 1,2,3,7,8,9,11,12 and13 is done in
the different cell types extracted from the M. tuberculosis-infected tissue models using quantitative
PCR and immunohistochemistry. Validation of the MMPs by comparison with tissues from human
TB patients are under investigation.
Our results show that the granuloma formation observed in the in vitro model upon infection with
virulent M. tuberculosis was inhibited in the presence of a global MMP inhibitor. Identification of the
specific MMPs involved in the process of TB granuloma formation is underway. We further plan to
investigate the effect of MMP inhibitors on the inflammatory response and bacterial load in the
tissue model.
Our project will give insight into how MMP inhibitors may help managing TB.
1. Parasa VR et al. Dis. Model. Mech. 2014;7:281-8.
2. Braian C et al. J. Vis. Exp. 2015;104
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
327
301 Host-Pathogen interaction and beyond
22/09/2016
Enhanced presence of proteins related to type I interferon signalling at the Mycobacterium
tuberculosis phagosome
N. Reiling1, M. Hiller1, C. Steinhäuser1, D. Linke3, V. Tchikov2, A. Tholey3, S. Schütze2,
J. Brandenburg1
1
Microbial Interface Biology, Research Center Borstel, Borstel 2Institute of Immunology 3Systematic
Proteomics and Bioanalytics, Christiane Albrechts University, Kiel, Germany
This project explores the molecular mechanisms operating at the interface of Mycobacterium
tuberculosis strains of clinical relevance and their first intracellular habitat, the phagosome. This
compartment determines intracellular survival and growth of the pathogen, host cell destruction and,
ultimately, impacts on the outcome of tuberculosis (TB). Recently we identified clade-specific
virulence pattern of M. tuberculosis complex (MTBC) strains: Exclusively human-adapted M.
tuberculosis lineages, also termed clade I, comprising modern lineages showed a significantly
enhanced capability to replicate inside in macrophages, when compared to clade II strains, which
include ancient lineages. This project now focusses on the sequelae differentially induced by MTBC
clade I and II strains at the level of the phagosome. A recently developed immunomagnetic isolation
procedure to isolate pathogen-containing phagosomes was used to characterize live and heat
inactivated MTBC clade I strain-containing phagosomes from macrophages to comparatively
evaluate their protein content by mass spectrometry. These data show that viable M. tuberculosis
induces the presence of proteins related to type I interferon signalling to its phagosome.
Experiments, which focus on the induction and the presence of these factors in M. tuberculosisinfected macrophages will be discussed.
328
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
302 Host-Pathogen interaction and beyond
22/09/2016
The pro-apoptotic factor Bax regulates macrophage necrosis during Mycobacterial
infection
F. Roca Soler, S. Redmond, L. Ramakrishnan
Department of Medicine, University of Cambridge, Cambridge, United Kingdom
In the zebrafish-Mycobacterium marinum model for tuberculosis infection, excess TNF triggers
necrosis of infected macrophages which leads to exuberant extracellular bacterial growth and host
susceptibility (Tobin et al.,2012; Roca and Ramakrishnan, 2013). It has been shown that in infected
macrophages, excess TNF induces production of mitochondrial reactive oxygen species (ROS)
which activate two death pathways that converge in macrophage necrosis: cyclophilin D-dependent
mitochondrial transition pore and acid sphingomyelinase-dependent ceramide production that leads
to lysosomal permeabilization.
In exploring the events downstream of lysosomal permeabilization, we found that ceramide acts in a
Cathepsin D-dependent manner to induce cell death. In the cytosol Cathepsin D initiates the
activation of pro-apoptotic factors including Bid and Bax. We engineered Bax mutants that lack its
different functional domains and studied the ability of each mutant protein to regulate apoptosis and
necrosis. Then we infected Bax-deficient zebrafish larvae expressing the Bax mutants that were
unable to induce apoptosis. We found the BH3 domain, which is required for Bax oligomerization
during apoptosis, not to be required for necrosis under excess TNF conditions. Rather, an Nterminal transmembrane helix suggested to be the first portion of Bax in contact with the
mitochondrial membrane was found to be required for necrosis. Current studies are focused on
understanding post-translational modifications of Bax that determine whether activation of the same
protein promotes apoptosis or necrosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
329
303 Host-Pathogen interaction and beyond
22/09/2016
A secreted Mycobacterium tuberculosis protein promotes disseminated disease: insights
from an outbreak
J. Saelens1, D. Sisk1, S. Lee3, J. Stout2, D. Tobin1
1
Molecular Genetics and Microbiology, Tobin lab, Duke University 2Department of Medicine,
Division of Infectious Diseases and International Health 3Human Vaccine Institute and Department
of Medicine, Duke University Medical Center, Durham, United States
Extrapulmonary tuberculosis causes significant morbidity and mortality worldwide. Here, we report
an unusual outbreak of tuberculosis with an exceptionally high rate of disseminated disease in
immunocompetent adults. We developed a zebrafish model of mycobacterial dissemination to
examine specific genetic variants from the outbreak strain. We identify one variant impacting a
secreted protein, and demonstrate a role for this variant in altering macrophage motility and the rate
of disseminated infections.
330
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
304 Host-Pathogen interaction and beyond
22/09/2016
CD4+ T Cells Recognizing PE/PPE Antigens Directly or via Cross Reactivity are Protective
against Pulmonary Mycobacterium tuberculosis Infection
F. Sayes2, A. Pawlik2, W. Frigui2, M.I. Gröschel2, S. Crommelynck2, C. Fayolle3-1, F. Cia6,
G.J. Bancroft6, D. Bottai5-4, C. Leclerc3-1, R. Brosch2, L. Majlessi2
1
INSERM U1041 2Unité de Pathogénomique Mycobactérienne Intégrée 3Unité de Régulation
Immunitaire et Vaccinologie, Institut Pasteur, Paris, France 4Department of Biology 5Ricerca
Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, University of Pisa, Pisa,
Italy 6London School of Hygiene and Tropical Medicine, London, United Kingdom
Mycobacterium tuberculosis (Mtb), possesses at least three type VII secretion systems, ESX-1, -3
and -5 that are actively involved in pathogenesis and host-pathogen interaction. We recently
showed that an attenuated Mtb vaccine candidate (Mtb Δppe25-pe19), which lacks the
characteristic ESX-5-associated pe/ppe genes, but harbors all other components of the ESX-5
system, induces CD4+ T-cell immune responses against non-esx-5-associated PE/PPE protein
homologs. These T cells strongly cross-recognize the missing esx-5-associated PE/PPE proteins.
Here, we characterized the fine composition of the functional cross-reactive Th1 effector subsets
specific to the shared PE/PPE epitopes in mice immunized with the Mtb Δppe25-pe19 vaccine
candidate. We provide evidence that the Mtb Δppe25-pe19 strain, despite its significant attenuation,
is comparable to the WT Mtb strain with regard to: (i) its antigenic repertoire related to the different
ESX systems, (ii) the induced Th1 effector subset composition, (iii) the differentiation status of the
Th1 cells induced, and (iv) its particular features at stimulating the innate immune response. Indeed,
we found significant contribution of PE/PPE-specific Th1 effector cells in the protective immunity
against pulmonary Mtb infection. These results offer detailed insights into the immune mechanisms
underlying the remarkable protective efficacy of the live attenuated Mtb Δppe25-pe19 vaccine
candidate, as well as the specific potential of PE/PPE proteins as protective immunogens.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
331
305 Host-Pathogen interaction and beyond
22/09/2016
Single-cell FRET cytometry assay improves knowledge on the cytosolic access of
Mycobacterium tuberculosis
R. Simeone2, F. Sayes2, O. Song1, M. Gröschel2, P. Brodin1, R. Brosch2, L. Majlessi2
1
INSERM U1019, CNRS UMR 8204, Lille 2Pathogénomique Mycobactérienne Intégrée, Institut
Pasteur, Paris, France
The capacity of Mtb to block phagosome-lysosome fusion and allow intraphagosomal replication
has long been considered as a characteristic feature of Mtb which is mainly an intraphagosomal
pathogen. However, the pathogenic potential of Mtb is intimately linked to the interplay between the
host immune response and the persistence of the bacteria and notably the access of Mtb to the host
cytosol has numerous consequences on the activation of different axes of innate immunity.
To get novel insights into this important cell biological process, we had previously adapted to Mtb
studies, a single-cell Fluorescence Resonance Energy Transfer (FRET)-based assay, used in an
automated fluorescent microscopy. This approach allowed us to confirm the implicative role of the
ESX-1/type VII secretion system of Mtb and the secretion/export of two key virulent protein factors,
the 6 kDa Early Secreted Antigenic Target (ESAT-6/EsxA) and the 10 kDa Culture Filtrate Protein10 (CFP-10/EsxB), in the induction of phagosomal rupture. This event, initially evidenced at 3 days
post-infection, is then followed by host cell death (Simeone et al., PloS Pathogens 2012). We
recently adapted this FRET-based single cell method to multi-color cytoometry. Using this more
sensitive and quantitative approach, we studied the ESX-1-dependent phagosomal rupture in
several phagocyte types and under different physiological conditions. One of our findings is that the
phagosomal rupture occurs as early as 3h post-infection and evolves in a proportional manner with
the activation of inflammasome and IFN-b production. By tracking rare infected phagocytes within
the lungs and spleens of infected mice, we provided the first and unique evidence that Mtb-induced
phagosomal rupture occurs in vivo in different subsets of phagocytes Furthermore, by use of mouse
macrophages with a functional or non-functional allele of the Natural Resistance-Associated
Macrophage Protein-1 (Nramp-1), we established a link between the inhibition of phagosomal
acidification and phagosomal membrane rupture (Simeone et al., PloS Pathogens 2015).
Our results propose that Mtb is more than a passive pathogen and that its life cycle in host cells of
Mtb is highly complex. Our work open up new research pathways and contribute to a better
understanding of the host-pathogen interaction of Mtb.
332
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
306 Host-Pathogen interaction and beyond
22/09/2016
Deciphering the mechanisms of HIV-1 exacerbation induced by Mycobacterium
tuberculosis in monocytes/macrophages
S. Souriant3, L. Balboa2, K. Pingris3, D. Kviatcowsky2, B. Raynaud-Messina3, C. Cougoule3,
I. Mercier3, M. Kuroda4, P. González-Montaner1, S. Inwentarz1, E.J. Moraña1, M.C. Sasiain2,
O. Neyrolles3, I. Maridonneau-Parini3, G. Lugo-Villarino3, C. Vérollet3
1
Instituto Prof. Dr. Raúl Vaccarezza, Hospital de Infecciosas Dr. F.J. Muñiz 2Inmunologia de
Enfermedades Respiratorias, Instituto de Medicina Experimental (IMEX)-CONICET, Buenos Aires,
Argentina 3IPBS, CNRS UMR 5089, Toulouse, France 4Division of Immunology, Tulane National
Primate Research Center, Covington, United States
Tuberculosis (TB) and the Acquired ImmunoDeficiency Syndrome (AIDS) are two of the deadliest
diseases due to a single infectious agent, namely Mycobacterium tuberculosis (Mtb) and the Human
Immunodeficiency Virus (HIV-1), respectively. Worsening these public health issues is the fact that
HIV-1 is frequently associated with Mtb. The well-known synergy between the two pathogens places
an immense burden in the healthcare systems, particularly in resource-limited countries where coinfection with these pathogens are highly prevalent. Therefore, the development of new therapeutic
strategies requires a better understanding of the synergistic relationship between Mtb and HIV-1.
Both pathogens are able to impair the host immune response and share monocytes/macrophages
as common host target cells. Our work aims at deciphering the mechanisms by which Mtb
exacerbates HIV-1 infection in monocytes/macrophages.
Here, we demonstrate that treatment of human primary monocytes with pleural effusions from TB
patients (PE-TB) exacerbates HIV-1 infection (i.e. number of infected cells, virus entry and
replication, and formation of multinucleated giant cells), in comparison to treatment with PE
obtained from patients with other pulmonary infections (PE-nonTB). This increased HIV-1 infection
is associated with a deregulator (M2c) activation program of monocytes/macrophages,
characterized by the CD16+CD163+MerTK+CD169+ cell-surface marker signature, also observed on
monocytes from the pleural cavity of tuberculosis patients. The establishment of an in vitro culture
system, based on the utilization of supernatants from Mtb-infected primary human macrophages,
allowed us to differentiate freshly isolated monocytes from healthy donor towards both the M2c
activation program and the exacerbation of the HIV-1 infection of these cells, thus mimicking the
effects enacted by PE-TB. Using this approach, we identified specific molecular mechanisms
responsible for this HIV-1 infection amplification induced by Mtb in monocytes/macrophages,
including the increased expression of HIV-1 entry co-receptors CCR5 and CXCR4, and the IL10/STAT3 signaling pathway.
Collectively, this study improves our understanding of how Mtb modulates the differentiation
process of human monocytes towards a macrophage activation program that increases
susceptibility to viral infection and formation of potential cell reservoirs for HIV-1. Finally, this work
will provide novel target candidates with diagnostic and therapeutic potential against the comorbidity established between AIDS and TB.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
333
307 Host-Pathogen interaction and beyond
22/09/2016
Modulating activity of the inflammasome during infection with Mycobacterium tuberculosis
- a therapeutic adjunct?
S. Subbarao, N. Krishnan, B. Robertson
MRC Centre for Molecular Bacteriology, Imperial College, London, United Kingdom
Introduction: Mycobacterium tuberculosis (Mtb) remains a considerable cause of morbidity and
mortality.Whilst our host immunity is pivotal in containing Mtb infection, it has long been recognised
as also contributing to tissue damage, cavitatory breakdown, paradoxically facilitating pathogen
spread.There is an urgent need for alternative treatments such as immunomodulating agents, as an
adjunct to anti-microbial chemotherapy. This would reduce TB-associated tissue destruction,
increase anti-microbial access and reduce treatment length.
The inflammasome is a cytosolic complex that facilitates pro-inflammatory and microbicidal
responses through induction of IL-1b. This is highly regulated at the translational level, but
uncontrolled activation is associated with hyper-inflammatory diseases.The induction of the
inflammasome by H37Rv varies markedly within the literature.
Method: We are examining how clinical isolates interact with the inflammasome and promote
activation of IL-1b. We have also investigated whether other immunomodulatory drugs show
potential as adjuncts to anti-tuberculosis chemotherapy.
Results: Infection of immortalised BMDMs (wild-type, caspase 1/11-/-, NLRP3-/-, AIM2/NLRP3-/-) with
Mtb clinical isolates elicits differential inflammasome induction and variable caspase-1 activation.
Strains varied in induction of p20 IL-1b, which regulates conversion of pro-IL-1b into IL-1b, and is
activated under acidic conditions by cathepsin D. In vitro, therapeutic blockade of the inflammasome
(glibenclamide, CRID3, anakinra) demonstrates a dose dependent reduction in IL-1b release and
synergism with rifampicin.
Conclusion: We demonstrate the importance of using clinical isolates in vitro; the strain dependent
inflammasome induction suggests a mycobacterial protease may cleave pro-IL-1b. Differential
induction of p20 IL-1b, suggests varied regulation of IL-1b activation through cathepsin D activation.
Blockade of the inflammasome in ‘high-risk’ individuals offers a tailored approach to TB therapy. In
summary, Mtb has evolved to differentially manipulate the host innate response; compilation of how
multiple strains manipulate the host together with the clinical and radiological data could identify
individuals who would benefit from immunomodulatory therapy.
334
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
308
Host-Pathogen interaction and beyond
22/09/2016
Mortality of adult tuberculous meningitis is associated with Increased tryptophan
metabolism and genetic variation in IDO1 in Indonesia
R. Van Crevel3, A. Van Laarhoven3, C. Clish, Clary4, S. Dian1, R. Notebaart3, C. Ruesen3,
J. Annisa1, L. Chaidir1, M. Netea3, B. Alisjahbana1, V. Kumar2, A. Ganiem1
1
Health Research Unit, TB HIV working group, Universitas Padjadjaran, Bandung, Indonesia
2
Department of Genetics, University of Groningen, University Medical Centre Groningen,
Groningen 3Department of Internal Medicine and Radboud Center for Infectious Diseases,
Radboudumc, Nijmegen, The Netherlands 4The Broad Institute of MIT and Harvard, Boston, United
States
Background: Damaging or ineffective host immune responses may contribute to the high mortality
of tuberculous meningitis (TBM), but the underlying mechanisms remain largely unknown. We
examined cerebrospinal fluid (CSF) inflammatory markers, metabolomics and host genetics in a
cohort of adult TBM patients in Indonesia.
Material/methods: 307 HIV-negative TBM patients were included based on typical CSF leukocyte
count and CSF:blood glucose ratio, and positive M. tuberculosis culture. Metabolomics analysis
using liquid chromatography mass spectrometry was performed on CSF and serum of 33 culture
positive TBM patients and 22 controls with CSF cell count < 5 cells/μl and negative mycobacterial
culture. 358 metabolites were identified in CSF and 397 in serum of cases and controls. Data were
visualized using principal component analysis (PCA), classified with partial least square (PLS)
analysis, and correlating to diagnosis and survival status through pathway enrichment analysis.
Polymorphisms in genes coding for enzymes in identified pathways were genotyped using an
Illumina HumanCoreExome-24 BeadChip, and correlated to survival.
Results: PCA showed clear visual separation of TBM patients and controls based on metabolite
concentrations in CSF but not in serum. In CSF this corresponded to a 94% accuracy for separating
patients from controls and 75% for TBM survivors versus non-survivors. Pathway analysis showed
thiamine metabolism, cyanoamino acid metabolism, aminoacyl-tRNA-biosynthesis and tryptophan
metabolism as top upregulated pathways in TBM (false discovery rates p< 10-20). CSF tryptophan
was 9.8-fold lower in patients versus controls, while its metabolites (kynurenine, quinolinate) were
all higher in patients, reflecting strongly increased Indoleamine 2,3-dioxygenase 1 (IDO1) activity.
Interestingly, CSF tryptophan levels were lower especially in patients who survived (31-fold
compared to controls) and much less affected (4-fold compared to controls) in those who died.
Polmorphisms in IDO1, which codes for the rate-limiting enzyme in conversion of tryptophan to
kynunrenine were associated with survival.
Conclusions: This first metabolomics study of TBM shows strongly increased tryptophan
metabolism in TBM. Tryptophan is essential for growth of M. tuberculosis, and tryptophan
metabolism skews the balance between regulatory and effector T-cells. Our results suggest
that induction of tryptophan metabolism could limit M. tuberculosis growth and damaging
immunopathology in TBM. these findings may help development of new immunomodulatory
strategies for TBM and could also aid in its diagnosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
335
POSTER SESSION 4
Friday, September 23
SUMMARY
Drug design-new developments .................................................. 339
From animal models to man ........................................................ 380
309 Drug design-new developments
23/09/2016
Bacteriocin AS-48 is active against Mycobacterium tuberculosis
C. Aguilar Pérez5-6, B. Gracia5-6, A. Vitoria5-4-6, R. Cebrián1, M. Maqueda1, J.A. Ainsa2-3-5-6
1
Departamento de Microbiología, Facultad de Ciencias, Universidad de Granada, Granada 2CIBER
de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid 3Instituto de
Biocomputacion y Fisica de Sistemas Complejos, BIFI, Universidad de Zaragoza 4Servicio de
Microbiología, Hospital Clínico Universitario Lozano Blesa 5Instituto de Investigacion Sanitaria de
Aragon (IIS-Aragon) 6Genética de Micobacterias, University of Zaragoza, Zaragoza, Spain
The increasing incidence of multi-drug resistant strains of Mycobacterium tuberculosis and the very
few drugs available for treatment is promoting the development of a new line of drugs that try to
solve these problems. Following this idea, we explored the antibacterial peptide AS-48, produced by
E. faecalis, which is targeting the bacterial membrane, and it is active against several gram-positive
bacteria. Targeting the membrane is an advantage considering that there are a lot biosynthetic
pathways involved, so it is less likely to select a resistance mutation.
This study has assessed the bactericidal effect of AS-48 in M. tuberculosis strains, including H37Rv
and other clinical and reference strains, and also against some non-tuberculous clinical
mycobacterial species. We demonstrated that AS-48 has a bactericidal action against M.
tuberculosis and other mycobacterial species. The combination of AS-48 with either lysozyme or
first line drugs (commonly used in the treatment of tuberculosis) increases bactericidal action of AS48, showing a synergic interaction. Under these conditions, AS-48 kills M. tuberculosis at a lower
dose, and exhibits a MIC (Minimal Inhibitory Concentration) close to some of the first line anti-TB
agents.
In addition, we assayed cytotoxicity of AS-48 against THP-1 and MHS macrophage cell lines, and
found that at concentrations close to the MIC of AS-48 we could not detect any cytotoxic effect.
In summary, we consider that bacteriocin AS-48 has an interesting potential in antituberculosis
therapy because of its activity against mycobacterial species and its low cytotoxicity against cell
lines.
KeyWords: AS-48, antimicrobial peptide, antituberculosis activity, synergy.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
339
310 Drug design-new developments
23/09/2016
Pathway-based whole cell screening of compounds against mycobacteria: targeting
genome integrity
W. Aragaw2, U. Lakshmanan1, C. Ling1, A. Poulsen1, J. Hill1, Y. Yamada2, T. Dick2
1
Experimental Therapeutics Center, Agency for Science, Technology and Research
(A*STAR) 2Department of Microbiology and Immunology, Yong Loo Lin School of Medicine,
National University of Singapore, Singapore, Singapore
Tuberculosis (TB) continues to pose heavy toll on global health. The increasing trends of drug
resistance along with other etiologic factors made the TB disease more difficult to control. A most
serious aspect of the problem of drug resistance is the emergence of strains resistant to many of
the standard anti-TB drugs. To counter the spread of drug resistance and to effectively control the
disease epidemic as a whole, new medicines are demanded. A major challenge to developing new
TB drugs is identifying whole cell active compounds with their putative targets. In this study, we
employ a cell-based high throughput screening approach coupled to pathway reporter system to
find novel target-lead couples. We broadly target genome integrity with a premise that inhibition of
the indispensable protein components involved in mycobacterial genome stability would result in
desirable therapeutics. To identify chemotypes that interfere with genome integrity, we use a
recombinant Mycobacterium bovis BCG harbouring an integrative plasmid with mCherry reporter
gene transcriptionally fused to a DNA damage inducible promoter (BCG::pMV306-PrecA-mCherry). In
the primary screening, compounds with growth inhibition activity against BCG are identified from a
commercial library (Enamine) of 70,000 synthetic compounds. The screen yielded 18 hit compounds
which upon re-test showed dose dependent activity. All confirmed hits were then subjected to the
subsequent pathway specific screening. Two whole cell actives (compounds E04 and E08) elicited
a dose dependent induction of mCherry expression in the DNA damage reporter assay signifying
their interference with mycobacterial genome integrity. In vitro potency studies in BCG showed a
pattern of concentration dependent growth inhibition (MIC50 = 6.25 µM). With respect to cidal
activity, both compounds exhibited similar kill pattern with a 3 log reduction in CFU/ml at 25 µM.
Cytotoxicity profiling revealed no apparent in vitro toxicity and haemolytic activity up to 10X the MIC
concentration. To provide further evidence for the possible target(s) involved, spontaneous resistant
mutants are being isolated and characterized.
340
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
311 Drug design-new developments
23/09/2016
Identification of novel drug target pathways and scaffolds to combat tuberculosis
G. Arora
VIDRC, TB LAB, Translational health science and technology institute, Delhi-Ncr, India
Due to emergence of drug resistant TB strains, there is an urgent need to validate newer drug target
pathways and scaffolds with novel mechanism of action. In my lab we are validating amino acid and
carbon metabolism pathways as drug targets to combat M. tuberculosis. L-serine biosynthetic
pathway represents one such pathway, in which phosphoserine phosphatase (PSP), a key essential
enzyme involved in conversion of O-phospho-L-serine to L-serine, acts at the final step. We have
done detailed biochemical characterization of PSP homolog in M. tuberculosis, SerB2 and
developed a malachite green based high throughput assay system to identify novel SerB2 specific
inhibitors. We have identified 10 novel scaffolds that were structurally different from known PSP
inhibitors and few of these were highly specific in their ability to inhibit SerB2 enzyme in comparison
to human PSP enzyme, non-cytotoxic against mammalian cell lines and inhibited M. tuberculosis
growth in vitro. Surface plasmon resonance experiments demonstrated the relative binding for these
inhibitors. The two best hits identified in our screen, clorobiocin and rosaniline were bactericidal in
activity and killed intracellular bacteria in a dose dependent manner. We are also focusing on
carbon metabolic pathways such as Tricarboxylic acid pathway, which is a converging point of many
catabolic pathways of central carbon metabolism, to understand how they play a key role in
physiology and virulence of M. tuberculosis and to identify novel scaffolds that target these
pathways. In addition, using whole-cell screening approach, we identified 28 compounds out of
5000 screened as novel inhibitors of M. tuberculosis from small molecule library. Future
experiments would include cytotoxicity evaluation of the hits against mammalian cell lines and their
mechanistic studies.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
341
312 Drug design-new developments
23/09/2016
Boromycin kills mycobacterial persisters without detectable resistance
W. Moreira, D. Aziz, T. Dick
Department of Microbiology and Immunology, National University of Singapore, Singapore,
Singapore
Boromycin is a boron-containing polyether macrolide antibiotic isolated from Streptomyces
antibioticus. It was shown to be active against Gram positive bacteria and to act as an ionophore for
potassium ions. The antibiotic is ineffective against Gram negative bacteria where the outer
membrane appears to block access of the molecule to the cytoplasmic membrane. Here we asked
whether Boromycin is active against Mycobacterium tuberculosis which, similar to Gram negative
bacteria, possesses an outer membrane. The results show that Boromycin is a potent inhibitor of
mycobacterial growth (MIC50 = 80 nM) with strong bactericidal activity against growing and nongrowing drug tolerant persister bacilli. Exposure to Boromycin resulted in a rapid loss of membrane
potential, reduction of the intracellular ATP level and leakage of cytoplasmic protein. Consistent with
Boromycin acting as a potassium ionophore, addition of KCl to the medium blocked its
antimycobacterial activity. In contrast to the potent antimycobacterial activities of the polyether
macrolide, its cytotoxicity and haemolytic activity were low (CC50 = 30 uM, HC50 = 40 uM) with a
selectivity index of more than 300. Spontaneous resistant mutants could not be isolated suggesting
a mutation frequency of less than 10-9 / CFU. Taken together, the results suggests that targeting the
mycobacterial membrane / ion gradient may be an attractive chemotherapeutic intervention to kill
otherwise drug tolerant persister bacilli, and to slow down the development of genetic antibiotic
resistance.
342
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
313 Drug design-new developments
23/09/2016
Development and validation of low-cost dna amplification-based assays for rapid diagnosis
of tuberculosis and detection of mutations associated with rifampicin resistance in
moroccan patients
E. Bentaleb3-2, M. Abid4, M.D. El Messaoudi1, S. Amzazi2, H. Sefrioui3, H. Ait Benhassou3
1
Laboratoire de Tuberculose, Institut Pasteur du Maroc, Casablanca 2Department of Biology,
Laboratory of Biochemistry and Immunology, Mohammed V University of Rabat 3Medical
Biotechnology Center, Moroccan Foundation for Advanced Science, Innovation and Research
( MAScIR), Rabat 4Laboratoire de Génétique Mycobactérienne, Institut Pasteur du Maroc, Tangier,
Morocco
Tuberculosis (TB) is one of the deadliest infectious diseases worldwide. Each year, about 9 million
people develop the disease and approximately 1.5 million of them die. In Morocco as in most
developing countries where the timely and accurate diagnosis of TB remains a great challenge, TB
constitutes a major public health threat with 30,000 new cases each year. Furthermore, control of
TB has been complicated by the emergence of multidrug-resistant (MDR) M. tuberculosis strains.
Currently, the conventional methods such as light microscopy and classic culture used to detect
Mycobacterium tuberculosis in Moroccan patients, especially in less equipped regions are complex,
unreliable, labor-intensive, technically challenging and time-consuming.
In that context, we are actually developing at MAScIR (Moroccan foundation for Advanced Science,
Innovation and Research) and for the first time in Morocco a new TB detection test based on loopmediated isothermal amplification technology (LAMP). Loop-mediated isothermal amplification
(LAMP) has emerged as a powerful tool for rapid diagnosis of Tuberculosis in endemic settings of
resource-poor countries, due to its simplicity, rapidity and cost-effectiveness. Our TB-LAMP assay is
a single step LAMP (SS-LAMP) based on the amplification of IS6110 specific to Mycobacterium
tuberculosis complex (MTBC). It has been successfully tested on DNA extracted from culture and
also directly on liquefied sputum samples without any prior DNA extraction or denaturation,and
also without the final enzymatic inactivation step.
On the other hand, to encounter the alarming increase in the global incidence of MDR-TB infection,
a need for methods that can rapidly and effectively identify drug-resistant cases becomes an
emergency. Thus, we are currently developing a highly sensitive, accurate, and rapid and an
affordable assay, based on qPCR-HRM technology, able to detect and identifiy mutations in rpoB
gene responsible of rifampicin resistance to Mycobacterium tuberculosis, by using five recombinant
plasmids as reference.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
343
314 Drug design-new developments
23/09/2016
In vitro and in vivo profiling of a new anti-Tuberculosis serie discovered by phenotypic
screening
I. Blanc, L. Goullieux, C. Lair, L. Somody, C. Briot, C. Cantalloube, X. Boulenc, E. Fontaine,
M. Magnon, M. Doubovetzky, A. Milla, A. Oualim, A. Pellet, E. Bacqué, S. Lagrange, L. Fraisse
R&D, SANOFI, Marcy L'etoile, France
Tuberculosis (TB) is caused by Mycobacterium tuberculosis (Mtb). It is the first leading infectious
cause of death worldwide. In order to address this unmet medical need, Sanofi has entered into
collaboration with TB Alliance, a key player in the TB drug discovery field, with the objective to
develop new combinations with the potential to shorten treatment of TB.
This poster will report the discovery and properties of a novel anti-TB series that stems from a
growth inhibition phenotypic screening. We will present, in particular, the in vitro and in vivo profile
of RA01, an optimized analogue of the series. in vitro, RA01 is very potent against replicating Mtb,
extra- or intra-macrophages as well as against representative clinical and mono-resistant Mtb
strains. RA01 also exhibits a significant serum-shift though retaining submicromolar potency and is
weakly active on non-replicating Mtb. In vivo, RA01 showed good exposure and satisfactory PK
parameters in mice, at pharmacologically active doses. It is efficacious in an Highly Acute TB mouse
model (MED at 100 mg/kg). It has also shown similar efficacy in an Acute TB model along with a
weak effect in a Chronic TB model that features Non Replicating Mtb.
Overall, RA01 is a potent inhibitor of Mtb growth. Combined to other antibiotics, it has the potential
to improve current TB treatments.
344
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
315 Drug design-new developments
23/09/2016
iniBAC induction is vitamin B12 and MutAB-dependent in Mycobacterium marinum
M. Boot, M. Sparrius, K. Jim, S. Commandeur, R. Van De Weerd, A. Speer, W. Bitter
Medical Microbiology and Infection control, VU medical center, Amsterdam, The Netherlands
Although the drug targets of most first-line antibiotics for Mycobacterium tuberculosis have been
identified, less research has focused on the intra-bacterial stress response that follows upon
treatment with antibiotics. Studying the roles of drug-induced stress genes may lead to the
identification of crucial stress-coping mechanisms that can provide additional drug targets to
increase treatment efficacy.
The three-gene operon iniBAC has no known function and is strongly up-regulated upon treatment
with isoniazid and ethambutol. We have confirmed upregulation of the iniBAC genes in
Mycobacterium marinum and found that the operon is also induced during cell infection
experiments, albeit that the response is more heterogenic as compared to bacterial culture. Next,
we set out to elucidate the genetic network that results in iniBAC induction in M. marinum. By
means of transposon mutagenesis, we identified that the operon is highly induced upon mutations in
genes encoding enzymes of the vitamin B12 biosynthesis pathway and the vitamin B12-dependent
methylmalonyl–CoA-mutase (MutAB). Subsequent lipid analysis showed that a mutA::tn mutant has
decreased PDIM levels, suggesting a link between iniBAC induction and the production of methylbranched lipids. Moreover, a similar screen in M. bovis BCG identified PDIM mutations to cause the
up-regulation of iniBAC genes. We have attempted to determine the metabolic cue that causes
iniBAC upregulation and conclude that there is a strong correlation between the utilization of fatty
acids and the induction of the iniBAC operon. Based on these data, we propose that iniBAC is
induced in response to mutations that cause defects in the biosynthesis of methyl-branched lipids.
The resulting metabolic stress caused by these mutations, or caused by ethambutol or isoniazid
treatment, may be relieved by iniBAC to increase of bacterial survival. We are currently dissecting
the role of IniBAC proteins in this process.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
345
316 Drug design-new developments
23/09/2016
Developement of new synthetic inhibitors of Mycobacterium tuberculosis DprE2
G. Chiodarelli1-3, G. Besra2, L.R. Cox3, J. Lelievre1, M. Cacho-Izquierdo1
1
GlaxoSmithKline, Tres Cantos, Spain 2School of Biosciences 3School of Chemistry, University of
Birmingham, Birmingham, United Kingdom
Mycobacterium tuberculosis is the pathogen responsible for human tuberculosis (TB). TB is still one
of the leading killers accounting about 9 million cases and 1.7 million deaths every year. The current
TB treatment is a six-month regimen with the forty years old drugs rifampicin, isoniazid, ethambutol
and pyrazinamide. The length of the treatment makes compliance difficult thereby favouring the
emergence of the resistant strains multi-drug resistant TB (MDR-TB) and extensively-drug resistant
TB (XDR-TB), for this reason is important to find more potent drugs and shorter treatment. DprE1
and DprE2 are the two subunits of the epimerase enzyme involved in the biosynthesis of decaprenyl
phosphor-D-arabinofuranose (DPA). DPA is the only furan sugar donor used by the bacterium to
synthesise the arabinogalctan (AG) and lipoarabinomannan (LAM), two of the major constituents of
the bacteria cell wall. Because of the essentiality and broad range of inhibitors targeting DprE1, this
enzyme is considered one of the most interesting targets for the development of new anti-tubercular
agents. In contrast with DprE1, DprE2 has received far less attention even if it has been
demonstrated that DprE2 is essential as well. The goal of this project is to develop a lead compound
from the DprE2 hit GSK426032A discovered by high throughtput screening by GSK in Tres Cantos
(Spain). At the moment the SAR investigation around our hit is limited by the lack of isolated and
purified DprE2, this does not allow us to have access to crystal structures of the protein and
enzymatic assays. Our approach to investigate the SAR of the hit is to synthesise and test against
M. tuberculosis whole cells several series of derivatives generated by modifications in different sites
in the original structure of the hit. The information derived from the SAR studies can help us to
obtain derivatives with improved physical chemical properties and safety profile.
346
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
317 Drug design-new developments
23/09/2016
Relational Sequencing TB Data Platform (ReSeqTB): a global collaborative effort to build
and deploy a centralized relational data warehouse for investigating the correlations
between M. tuberculosis genotypes and phenotypic drug resistance
P. Miotto2, E. Tagliani2, B. Tessema6, I. Comas4-3, N. Hillery8, L. Chindelevitch1, M. Ezewudo7,
J. Posey7, T. Rodwell8, D. Dolinger6, M. Schito9, C. Reseqtb Consortium5
1
Simon Fraser University, Vancouver, Canada 2Emerging Bacterial Pathogens Unit, San Raffaele
Scientific Institute, Milan, Italy 3CIBER on Epidemiology and Public Health, Madrid 4Tuberculosis
Genomics Unit, Institute of Biomedicine of Valencia, Valencia, Spain 5ReSeqTB,
Consortium 6Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland 7Centers for
Disease Control and Prevention, Atlanta 8University of California, San Diego 9Critical Path institute,
Tucson, United States
A global consortium has been established to develop the Relational Sequencing TB Data Platform
(ReSeqTB, https://platform.reseqtb.org/), aimed at understanding the genetic basis of drug
resistance (DR) in Mycobacterium tuberculosis (MTB) by correlating genotypic data with phenotypic
drug susceptibility testing (DST) and patient metadata (e.g. treatment outcomes).
ReSeqTB is a standardized, validated analysis pipeline, relational database and curated archive,
designed to be a plug-and-play and cloud-based bioinformatics solution for identifying canonical and
novel drug-resistance related genetic variants from raw whole genome sequencing files. The
platform has been developed to process and share global genotypic, phenotypic and related
metadata on MTB with transparent, regulatory compliant methodology and clear documentation,
while ensuring contributor-specific intellectual property and data privacy is preserved. The variant
analysis pipeline works with raw Illumina® fastq data and implements a series of quality control
thresholds. In parallel we have developed a statistical framework based on a combination of
likelihood ratios (LR) and p-values. Mutations are categorized as having “high”, “moderate” or
“minimal” confidence for association with DR, “not associated” or “indeterminate”. Both the variant
pipeline and the statistical drug resistance association framework have been validated using 90
MTB strains drawn from a well-characterized drug resistant strain collection. The results were
compared with other well-documented analysis and drug resistance databases like the PhyResSE
web tool and the Knowledge Synthesis TBDR DB (www.TBDR.org).
Once our statistical approach was validated we aimed at expanding the range of mutations
significantly associated to resistance. We applied our approach to data obtained from a
comprehensive systematic literature review for mutations and associated liquid/solid phenotypic
DST for rifampicin, isoniazid, fluoroquinolones second-line injectable drugs, and pyrazinamide. The
total number of MTB isolates used for evaluation ranged from 571 to 9537. Forty-four mutations with
moderate to high confidence associations with DR were identified for the testing data set. The data
set is being utilized by ReSeqTB as a baseline for drug resistance prediction and will be dynamically
validated and updated as retrospective and prospective NGS data fills the ReSeqTB data platform.
The final aim is to provide a tool for diagnostics development and for clinical drug resistance
prediction.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
347
318 Drug design-new developments
23/09/2016
Pulmonary co-administration of the synergic drugs Ethionamide and Booster using
aerosolized nanoparticles: a promising strategy to treat tuberculosis
J. Costa-Gouveia1, E. Pancani3, S. Jouny1, V. Delorme1, G. Salzano3, N. Willand2, A. Baulard1,
R. Gref3, P. Brodin1
1
U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Univ. Lille, CNRS, Inserm,
CHU Lille, Pasteur Institute of Lille 2U1177 - Drugs and Molecules for living Systems, Univ. Lille,
Inserm, Pasteur Institute of Lille, Lille 3UMR 8214 - ISMO, Univ. Paris Sud, Univ. Paris-Saclay,
CNRS, Orsay, France
Tuberculosis (TB) is a leading cause of death worldwide. The use of ethionamide (ETH), an
important second line anti-TB drug, is hampered by low oral bioavailability and the onset of severe
side effects. Recently discovered “booster”, molecules strongly increasing the efficacy of ETH, could
restore the use of this drug and thus improve the current clinical outcome of drug-resistant TB. To
investigate the simultaneously delivery of ETH and its booster BDM41906 in the lungs, we coencapsulated the synergic compounds in biodegradable polymeric nanoparticles (NPs), overcoming
the bottlenecks inherent to the strong tendency of ETH to crystallize and the limited water solubility
of the Booster.
The efficacy of the designed formulations was evaluated in TB infected macrophages using an
automated confocal high-content screening platform, showing that the drugs maintained their
activity after incorporation in NPs. Among tested formulations, “green” β-cyclodextrin (pCD) NPs
displayed the best physiochemical characteristics and were selected for in vivo studies. The NPs
suspension, administered directly into mice lungs using a Microsprayer®, led to a significant
decrease of the pulmonary bacterial load as compared to untreated mice. This study paves the way
for a future use of pCD NPs for the pulmonary delivery of the [ETH:Booster] pair.
Acknowledgments: Financial support for this work was provided by the European Community
(CycloN Hit Grant n° 608407), the Feder (12001407 (D-AL) Equipex Imaginex BioMed) and the
Region Nord Pas de Calais (convention n° 12000080).
348
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
319 Drug design-new developments
23/09/2016
Identification of novel mutations that confer resistance to pyrazinamide in virulent
Mycobacterium tuberculosis
N. Dillon, Y. Minato, A. Baughn
Microbiology, University of Minnesota, Minneapoils, United States
Pyrazinamide is often characterized as an irreplaceable component of the current short course
therapy, yet its efficacy is threatened by the emergence of drug resistance. One third of
pyrazinamide resistant clinical isolates have no identified mechanism of drug resistance. The
elucidation of novel pyrazinamide resistance mutations from clinical isolates is complicated by the
absence of parental strains for sequence verification. We therefore sought to generate
pyrazinamide resistant mutants in the virulent laboratory strain H37Rv through transposon
mutagenesis. Anticipating a preponderance of pncA mutations during pyrazinamide exposure due to
its characterized role in resistance, we instead selected isolates on pyrazinoic acid, the bioactivated
form of pyrazinamide, negating the selective advantage of pncA mutations. Prior to this work, only a
single mutation, in panD, had been found to lead to pyrazinoic acid resistance. Using our
mutagenesis screen we selected sixteen random colonies from the transposon library for further
phenotypic characterization. Phenotypic characterization of these strains demonstrated that each
isolate was resistant to both pyrazinoic acid and pyrazinamide under various culture conditions.
These isolates did not have increased resistance to isoniazid, indicating resistance is specific to
pyrazinamide. Of the sixteen colonies we identified nine unique insertions not previously linked to
pyrazinamide resistance. We therefore employed a tn-seq based approach to analyze the
remainder of the library. In all, we identified a little over one hundred different genes whose
mutagenesis led to pyrazinoic acid resistance. The identified insertions clustered to genes
responsible for central metabolism, proteosome function, and cellular stress response. Comparison
of our data with published genomes for clinical isolates with no mutations linked to pyrazinamide
resistance now allows us to identify the polymorphisms responsible for the loss of drug activity in
vivo. Through isolation and identification of the mutations responsible for one third of resistant
clinical isolates we can begin understanding and combating the emergence of pyrazinamide
resistance in Mycobacterium tuberculosis infections.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
349
320 Drug design-new developments
23/09/2016
Does the side-chain of delamanid and Q203 convey special affinity for Mycobacteria?
tested on proven and possible anti-Mtb alkaloid derivatives
R. Feilcke, P. Imming
Institute of Pharmacy, Pharmaceutical Chemistry, Martin-Luther-University Halle-Wittenberg,
Germany, Halle (saale), Germany
In 2014 (EMA, 2014), delamanid met the need for new antitubercular drugs. This was an immense
progress against the rising number of cases with XDR and MDR-tuberculosis, but the possibilities of
therapy still lack highly active substances that will lead to a short treatment regimen. The side chain
of this molecule attracted our attention. It is almost similar to the side chain of Q203 that is currently
in phase I clinical study (WGND, 2016). We investigated if this side chain would confer
antimycobacterial activity to other scaffolds. We therefore attached 4-(4-(trifluoromethoxy)phenoxy)piperidine to different heterocycles and submitted the derivatives to whole cell
antimycobacterial assays.
Figure 1: Delamanid and Q203 with a similar side chain.
Core structures were chosen among established antibiotics e.g. metronidazol as well as among
alkaloids e.g hippadine. This pyrrolophenanthrinone is derived from Amaryllidaceae alkaloids and
shows some antibacterial activity. Not much attention was paid to this class of compounds as
antibacterials yet, even though the extract of the leaves of Crinum purpurascens showed good
activity in some tests (Nkanwen et al., 2009). In order to facilitate access to this alkaloid class, a
short synthetic route was investigated that led to one of the core structures of the tested molecules.
Figure 2: Core structure derived from pyrrolophenanthrinone alkaloids.
Results of the whole cell assays as well as insights into synthetic details will be presented on the
poster in September 2016.
References:
• European Medicines Agency (EMA), Summary of the European Public Assessment Report
(EPAR) for Deltyba,
[http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/002552/h
uman_med_001699.jsp&mid=WC0b01ac058001d124], Accessed on 25th of May 2016
• Working Group on New TB Drugs (WGND), Drug Pipeline,
[http://www.newtbdrugs.org/pipeline.php], Accessed on 25th of May 2016
• Nkanwen, E., et al. (2009), Antibacterial agents from the leaves of Crinum purpurascens herb
(Amaryllidaceae), African health sciences 9, (264-269)
350
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
321 Drug design-new developments
23/09/2016
Covalent Modification of the Mycobacterium tuberculosis FAS-II Dehydratase by Isoxyl and
Thiacetazone
A. Grzegorzewicz4, N. Eynard1-2, A. Quémard1-2, E.J. North5, A. Margolis4, J.J. Lindenberger6,
J. Korduláková3, V. Jones4, P.J. Brennan4, R.E. Lee5, D.R. Ronning6, M.R. Mcneil4, M. Jackson4
1
Département Tuberculose et Biologie des Infections, CNRS; IPBS (Institut de Pharmacologie et de
Biologie Structurale) 2Université de Toulouse, Toulouse, France 3Department of Biochemistry,
Comenius University, Bratislava, Slovakia 4Microbiology Immunology and Pathology, Colorado
State University, Fort Collins 5Department of Chemical Biology and Therapeutics, St. Jude
Children’s Research Hospital, Memphis 6Department of Chemistry and Biochemistry, University of
Toledo, Toledo, United States
Isoxyl (ISO) and thiacetazone (TAC) are two antitubercular prodrugs formerly used in the clinical
treatment of tuberculosis. Although both prodrugs have recently been shown to kill Mycobacterium
tuberculosis through the inhibition of the dehydration step of the type II fatty acid synthase pathway,
their detailed mechanism of inhibition, the precise number of enzymes involved in their activation,
and the nature of their activated forms remained unknown. Our data demonstrates that both ISO
and TAC specifically and covalently react with a cysteine residue (Cys61) of the HadA subunit of the
dehydratase, thereby inhibiting HadAB activity. The results unveil for the first time the nature of the
active forms of ISO and TAC and explain the basis for the structure−activity relationship of and
resistance to these thiourea prodrugs. The results further indicate that the flavin-containing
monooxygenase EthA is most likely the only enzyme required for the activation of ISO and TAC in
mycobacteria.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
351
322 Drug design-new developments
23/09/2016
Whole blood Bactericidal Activity of faropenem with and without rifampicin against
Mycobacterium tuberculosis
M. Gurumurthy1, R. Verma1, C. Naftalin1, D. Hee1, Q. Lu3, K.Y. Seng2, L. Lee1, N. Paton1
1
Department of Medicine 2Department of Pharmacology, National University of
Singapore 3Singapore Clinical Research Institute, Singapore, Singapore
Background: In vitro studies indicate that faropenem (an oral penem) has activity against TB and
synergy with rifampicin and may have potential for repurposing as a TB drug. We investigated this
further in a whole-blood bactericidal activity (WBA) trial.
Methods: Healthy volunteers were randomised to receive a single dose of faropenem (600mg; with
amoxicillin/clavulanic acid 500mg/125mg; FAR-AMC group, n=8); rifampicin (10mg/kg; RIF group;
n=14); or the combination (RIF-FAR-AMC group; n=14). Blood was drawn pre-dose and at 0.5, 1, 2,
3, 4, 5, 6 and 8 hours post-dose. Drug levels were measured using liquid chromatography-tandem
mass spectrometry. WBA was measured by inoculating blood samples with Mycobacterium
tuberculosis (Mtb) H37Rv and estimating the change in bacterial colony forming units (CFU) at the
end of 72 hours’ incubation. The primary outcome measure was cumulative WBA at 8 hours
following drug ingestion.
Results: We found no evidence of activity in the FAR-AMC group (cumulative WBA 0.06ΔlogCFU;
p=0.99 vs zero change). Cumulative WBA showed a trend in favour of the RIF-FAR-AMC group at 8
hours (-0.57±0.09 and -0.60±0.10ΔlogCFU in RIF and RIF-FAR-AMC groups respectively; p=0.113;
p=0.023 for comparison over the first 1 hour post-dose). The Cmax and AUC of faropenem were
5.4mg/L and 16.2h*mg/L respectively, and MIC against Mtb H37Rv was 5-10 mg/L.
Conclusions: Faropenem does not appear to be useful as TB drug when used alone, possibly due
to low plasma levels relative to the MIC. However, we found limited evidence of modest synergy
with rifampicin that may merit further testing in clinical trials.
This study is registered in Clintrials.gov (NCT02393586).
352
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
323 Drug design-new developments
23/09/2016
3,5-Dinitrobenzylsulfanyl heterocycles as novel antitubercular agents: structure – activity
relationship study
G. Karabanovich1, L. Valášková1, J. Němeček1, J. Roh1, J. Stolaříková2, K. Mikušová3, R. Székely4,
P. Pávek1, V. Klimešová1, A. Hrabalek1
1
Department of Inorganic and Organic Chemistry, Charles University, Faculty of Pharmacy in
Hradec Kralove, Heyrovského, Hradec Králové 2Department of Bacteriology and Mycology,
Regional Institute of Public Health, Partyzánské Náměstí, Ostrava, Czech Republic 3Department of
Biochemistry, Comenius University, Faculty of Natural Sciences, Mlynská Dolina, Bratislava,
Slovakia 4Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne,
Switzerland
In this work we deal with the synthesis and structure-activity relationships of 1-alkyl/aryl-5-[(3,5dinitrobenzyl)sulfanyl]-1H-tetrazoles, 5-substituted-2-[(3,5-dinitrobenzyl)sulfanyl]-1,3,4-oxadiazoles,
1,3,4-thiadiazoles and 4,5-disubstituted-3-[(3,5-dinitrobenzyl)sulfanyl]-4H-1,2,4-triazoles as novel
antitubercular agents.[1, 2]
The influence of the position of nitro groups, introduction of additional group to 3,5dinitrobenzylsulfanyl moiety, reduction of one nitro group or its replacement with other electronwithdrawing group on antimycobacterial activity were intensively studied. The role of heterocycle
and the linker between heterocycle and 3,5-dinitrophenyl moiety was also widely explored.
Compounds with the most promising activities were further evaluated against multi-drug resistant
tuberculosis strains, other bacteria or fungi. The effects on viability of mammalian cell lines, e.g.
isolated human hepatocytes, were also studied.
All derivatives bearing 3,5-dinitrobenzyl moiety exhibited high antimycobacterial activity against
drug-susceptible and drug-resistant Mycobacterium tuberculosis strains and showed no activity
against other bacteria or fungi. Moreover, these compounds exhibited low in vitro toxicity in
mammalian cell lines.
The highest efficacy was displayed by 5-substituted-2-[(3,5-dinitrobenzyl)sulfanyl]-1,3,4-oxadiazoles
and 1,3,4-thiadiazoles with minimum inhibitory concentrations as low as 0.03 µM (0.011-0.026
µg/mL).
Metabolic radiolabeling experiments indicated that 3,5-dinitrobenzylsulfanyl-1,3,4-oxadiazoles inhibit
the synthesis of mycobacterial nucleic acids. However, the determination of specific target in
mycobacterium is under evaluation.
The study was supported by the Czech Science Foundation project (14-08423S) and Charles
University in Prague (SVV 260 291).
1. Karabanovich G. Eur. J. Med. Chem. 2014, 82, 324-340.
2. Karabanovich G. J. Med. Chem. 2016, 59 (6), 2362–2380.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
353
324 Drug design-new developments
23/09/2016
rpoB gene mutations among Mycobacterium tuberculosis isolates from extrapulmonary
tuberculosis
A. Khosravi Boroujeni2, A. Ghadiri1, H. Meghdadi2, A.H. Sina3, M. Mirsaeidi4
1
Immunology and Cell & Molecular Research Center 2Microbiology and Infectious & Tropical
Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences 3Danesh Medical
Laboratory, Ahvaz, Iran 4Department of Medicine, University of Miami, Miller School of Medicine,
Miami, United States
Background: The current study was aimed to analyze the mutations occurring in the rpoB gene of
Mycobacterium tuberculosis (MTB) isolates from clinical samples of extrapulmonary tuberculosis
(EPTB) using the nested rpoB-PCR technique combined with cloning.
Methods: Seventy formalin-fixed, paraffin-embedded samples from confirmed EPTB cases were
analyzed. Nested PCR based on the rpoB gene was performed on the extracted DNAs, combined
with cloning and subsequent sequencing.
Results: In total, 67 (95.7%) were positive for the presence of MTB. Sequence analysis of the 81bp
region of rpoB gene, demonstrated mutations in 41 (61.2%) out of 67 sequenced samples. Several
point mutations including deletion mutations at codons 510, 512, 513 and 515, with 45% and 51% of
the mutations in codons 512 and 513 respectively were seen, along with 26% replacement
mutations at codons 509, 513, 514, 518, 520, 524 and 531. The most common alteration was Gln à
His, at codon 513, presented in 30 (75.6%) isolates.
Conclusions: This study confirmed that 75.6% of RIF-resistant strains have sequence alterations in
codon 513 of the 81bp region of rpoB gene. In addition, the simultaneous mutation at the codons
512 and 513, were demonstrated in 34.3% of the isolates.
354
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
325 Drug design-new developments
23/09/2016
Genetic diversity of multidrug resistant Mycobacterium tuberculosis strains isolated from
tuberculosis patients in Iran, using MIRU-VNTR technique
A. Khosravi Boroujeni, S. Khandan Dezfuli1, A. Hashemi Shahraki2, M. Hashemzadeh1
1
Infectious & tropical Diseases Research Center, Ahvaz Jundishapur University of Medical
Sciences, Ahvaz 2Epidemiology, Institute Pasteur of Iran, Tehran, Iran
Introduction: Tuberculosis (TB) is considered as one of the most important infectious diseases in
the world, and recent rise and spread of multi-drug resistant (MDR) Mycobacterium tuberculosis
(MTB) strains, have made the matter worsened. Therefore, there is an urgent need to understand
and estimate the incidence of tuberculosis in large and small biographical ranges by aid of
molecular biology methods. Due to the importance of TB prevalence in Iran, this study was
designed to investigate the genetic diversity among MDR strains of MTB by MIRU-VNTR typing
scheme.
Materials and Methods: A total of 88 drug resistant M. tuberculosis isolates belong to pulmonary
TB cases were collected from several TB reference centers of Iran. Drug susceptibility testing for
Isoniazid and Rifampin was performed using the agar proportion method and MDR isolates were
underwent genotyping by using 12-locus- based MIRU-VNTR typing.
Results: On performing proportion method, 22 isolates were identified as MDR. By typing of MDR
isolates using 12-loci MIRU-VNTR technique, high diversity were demonstrated in MDR strains and
these were classified into 20 distinct MIRU-VNTR genotypes, including 2 clusters (2 strains/cluster)
and 18 unique patterns. MIRU loci 10 and 26, were the most discriminatory loci with 8 and 7 alleles
respectively.; while MIRU loci 2, 20, 24 and 39 were found to be the least discriminatory with 1-2
alleles each. We noticed a mixed infection in isolate 53, as this isolate comprised simultaneous 2
alleles in MIRU loci 40, 10, 16 and 39. The discriminatory power of MIRU-VNTR was high
(HGDI=0.991%) in present study with clustering rate of 0.086%.
Conclusions: The results show that MIRU-VNTR typing is a useful tool for studying genetic
diversity of MTB in regional settings. Besides, this genotypic analysis will help the health sectors to
construct an effective control program for the disease. Additionally, it can detect mixed infection
which can facilitate management of treatment.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
355
326 Drug design-new developments
23/09/2016
Oligotuftsin-based Peptide Carriers for Novel Antimycobacterial Active Agents: Synthesis
and In vitro Evaluation
M. Krátký1, J. Vinšová1, Z. Baranyai3, S. Bösze3, N. Szabó2
1
Department of Inorganic and Organic Chemistry, Charles University in Prague, Faculty of
Pharmacy in Hradec Králové, Hradec Králové, Czech Republic 2Laboratory of Bacteriology,
Korányi National Institute for Tuberculosis and Respiratory Medicine 3MTA-ELTE Research Group
of Peptide Chemistry, Budapest, Hungary
The global tuberculosis epidemic and increasing emergence of drug-resistant Mycobacterium
tuberculosis strains as well as non-tuberculous mycobacteria (NTM) call for intensive research on
new therapeutic interventions including novel drugs and drug delivery systems (DDS) for them.1
DDS have been used largely for the modification of inconvenient properties of bioactive molecules.
Illustratively, they are able to increase solubility, bioavailability or decrease toxicity. Additionally,
they are useful in targeted drug delivery.2
We selected tuftsin-based oligopeptides as potential carriers for our small antimycobacterial active
molecules. Tuftsin derivatives are non-toxic, non-immunogenic and biodegradable. They also
stimulate immune response and target macrophages specifically, thus enhancing cellular uptake
and selectivity index. A pilot study with isoniazid indicates that this concept seems to be viable.3
Salicylanilides (2-hydroxy-N-phenylbenzamides) have exhibited a significant in vitro activity against
both tuberculous and NTM including drug-resistant strains at low micromolar concentrations.
However, they share a significant cytotoxicity and poor solubility.4 These obstacles can be
overcome, i.a., by employment of DDS.
Peptide carriers based on repeated oligotuftsin sequence [TKPKG]n were obtained using solidphase synthesis (Fmoc/tBu strategy, rink amide MBHA resin, diisopropylcarbodiimide/HOBt in Nmethylpyrrolidone). N-Terminus and/or lysine side chain amino group(s) were modified by various
substituents (carboxylic acids, fluorescent labels, short peptides spacers, aminooxyacetic acid etc.).
Carriers were cleaved from resin, purified and then coupled with salicylanilide derivatives bearing a
carbonyl group via oxime bond. Obtained conjugates were purified and characterized (MS, RPHPLC).
The conjugates were evaluated for their in vitro extracellular antimycobacterial activity (two strains
of M. tuberculosis, M. abscessus), intracellular activity in infected macrophages, cytotoxic and
cytostatic properties and cellular uptake. In general, salicylanilide-oligotuftsin conjugates showed
improved activity against both extracellular and intracellular mycobacteria, enhanced cellular uptake
together with decreased toxicity. They were effective also against multidrug-resistant M.
tuberculosis and “chemoresistant” M. abscessus. These promising in vitro results could stimulate
further research on this field.
References
1. Vinšová J., Krátký M.: Nova Science Publishers 2010, ISBN 978-1-61668-233-0.
2. Tiwari G. et al.: Int. J. Pharm. Invest. 2, 2 (2012).
3. Horvati K. et al.: J. Pept. Sci. 15, 385 (2009).
4. Krátký M., Vinšová J.: Curr. Pharm. Des. 17, 3494 (2011).
356
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
327 Drug design-new developments
23/09/2016
Pharmacomodulation on the piperidinol skeleton: Design, synthesis, and biological
evaluation of novel PIPD1 derivatives as Mycobacterium abscessus agents
F. Dubar4, C. Dupont2-3, V. Le Moigne3, J.L. Herrmann3, Y. Guerardel4, L. Kremer2-1, C. Biot4
1
CPBS, INSERM 2Centre National de la Recherche Scientifique FRE 3689, Centre d’études
d’agents Pathogènes et Biotechnologies pour la Sante, Université de Montpellier, Montpellier
3
3UMR1173, INSERM, Université de Versailles St Quentin, Versailles St Quentin 4UGSF - France,
Université Lille 1, Villeneuve D'ascq, France
Mycobacterium abscessus (M. abscessus) is a nontuberculous mycobacteria (NTM) that is
responsible for a wide range of disease, particularly pulmonary diseases. M. abscessus complex is
the most drug resistant of the mycobacterial pathogens.1 Even classical antitubercular drugs
(Clarithromycn, amikacin and Cefoxitin) are used in the treatment to M. abscessus, the long
treatment period are often unsuccessful. Moreover, no vaccine is currently avaiblable against this
pathogen.2
In this context, increased effort to develop new drugs for NTM diseases are really needed.
Recently, we identified a new piperidinol-based molecule, PIPD1.3 This promising compound exhibit
potent activity against clinical M. abscesssus strains in vitro and in infected macrophages.
In this study, novel derivatives of PIPD1 were designed, synthesized and tested for their in vitro
antimycobacterial effects on M. abscessus strains. The range of CMI is from 2 µg/ml to 0.25 µg/ml.
In vitro cytotoxicity effects of these new compounds were also evaluated in vitro against murine
J774 cells line in order to determine the therapeutic index of these series. The structure-activity
relationships (SARs) were discussed.
1. Henkle E., Hedberg K., Schafer S., Novosad S., Winthrop K.L. (2015) Population-based
incidence of Pulmonary Nontuberculous Mycobacterial Disease in Oregon 2007 to 2012. Ann
Am Thorac Soc 12 : 642-647.
2. Le Moigne V., Gaillard J.L., Herrmann J.L. (2016) Vaccine strategies against bacterial
pathogens in cystic fibrosis patients. Médecine et Maladies Infectieuses 46: 1, 4–9.
3. Dupont C., Viljoen A., Dubar F., Blaise M., Bernut A., Pawlik A., Bouchier C., Brosch R.,
Guérardel Y., Lelièvre J., Ballell L., Herrmann J.L., Biot C., Kremer L. (2016) A new piperidinol
derivative targeting mycolic acid transport in Mycobacterium abscessus. Mol Microbiol. doi:
10.1111/mmi.13406.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
357
328 Drug design-new developments
23/09/2016
GTPase-Obg mRNA expression analysis in Mycobacterium under stress conditions
V. Kumar2, H.N. Singh1, A.K. Tomar2, S. Yadav2
1
School of Sciences, Noida International University, Gautam Budh Nagar, UP, India, Gautam Budh
Nagar 2Department of Biophysics, All India Institute Of Medical Sciences, New Delhi, India
To counter Multi Drug Resistance (MDR) and Extensively Drug Resistance (XDR) strains of
Mycobacterium tuberculosis, there is an urgent need of identification and characterization of new
drug targets. GTPase-Obg is a GTP binding protein that belongs to a highly conserved protein
family in both prokaryotes and eukaryotes. It plays many roles in bacteria, including ribosome
biogenesis, protein translation regulation, stress response, cell growth regulation, etc. Past studies
have reported that GTPase-Obg from Mycobacterium tuberculosis interacts with ribosome, but its
role in the bacterial defence mechanism is unclear to date. Here, our study reports expression,
purification, biophysical characterization and expression analysis under stress conditions of
GTPase-Obg. Circular dichroism experiments revealed that GTPase-Obg underwent conformational
changes in presence of GTP. mRNA expression analysis was performed using real time PCR in
various stress conditions, including heat and cold shock and antibiotics. Significant increase in
GTPase-Obg mRNA was observed during different stress conditions, highlighting a possible role of
GTPase-Obg in Mycobacterium stress response. Further, three-dimensional structure of GTPaseObg was modelled and its interaction with some known GTPase inhibitors was studies in silico. This
study concludes that GTPase-Obg acquires functional stability in presence of GTP through helix
destabilization, may have a crucial role in Mycobacterium stress response and might be explored as
a new target to counter drug resistance.
358
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
329 Drug design-new developments
23/09/2016
Genetic evidence for a new binding site of Bedaquiline (TMC207) in the ε subunit of the
mycobacterial F-ATP synthase
S. Kundu2, G. Biukovic2, G. Grüber1, T. Dick2
1
School of Biological Science, Nanyang Technological University 2Department of Microbiology &
Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,
Singapore
The F-ATP synthase binding and inhibition mechanism of Bedaquiline (BDQ; former name
TMC207), a newly approved TB drug, is the focus of this project. Previous studies ([1][2][3]) suggest
that the c-subunit of the mycobacterial F-ATP synthase contains the binding site of the drug.
However, we showed recently - employing biochemical, biophysical and modeling methods - that
TMC207 may in addition bind to the interface of subunits ε, γ and c [4,5]. Here we confirm the
involvement of subunit ε in this new binding mode in intact bacteria using genetics. Recombineering
[6] was employed to introduce targeted missense mutations into the ε subunit, encoding gene at
sites of ε close to the postulated BDQ-binding site. Exchanging these amino acids was expected to
result in shifts of the minimal inhibitory concentration (MIC50) indication increased or decreased
binding of the drug to its ε target. The amino acid changes introduced were W16A and R37G [4]. To
determine whether the amino acid exchange mutations affect MIC50s of M. smegmatis atpCW16A and
M. smegmatis atpCR37G, growth inhibition dose response curves were generated with the drug. The
results showed no significant difference between wildtype bacteria and M. smegmatis atpCR37G
suggesting that the interaction of residue R37 of ε with BDQ may not be essential for BDQ-binding.
In contrast, M. smegmatis atpCW16A showed a 5-fold decrease in its MIC50 form 10 +/- 2 nM in the
wildtype strain to 2 +/- 1 nM in the mutant strain. This result suggests that W16 of subunit ε is close
to the BDQ-binding site as predicted previously by in vitro tryptophan fluorescence- and NMR
spectroscopy [4]. It furthermore suggests that the substitution of the bulky sidechain of W16 with a
smaller side chain of alanine allows for a tighter binding of BDQ to ε. In conclusion, we provide
genetic in vivo evidence for a new binding and inhibition mechanism of BDQ involving subunit ε of
the mycobacterial F-ATP synthase.
This work is supported by the Ministry of Health (MOH), Singapore (NMRC, CBRG12nov049; G.
Grüber and T. Dick). Subhashri Kundu receives NUS Research Scholarship from Yong Loo Lin
School of Medicine.
References:
1. de Jonge, M.R., Koymans, L.H., Guillemont, J.E., Koul, A. and Andries, K., 2007. A computational model of
the inhibition of Mycobacterium tuberculosis ATPase by a new drug candidate R207910. PROTEINS:
Structure, Function, and Bioinformatics, 67(4), pp.971-980.
2. Segala, E., Sougakoff, W., Nevejans-Chauffour, A., Jarlier, V. and Petrella, S., 2012. New mutations in the
mycobacterial ATP synthase: new insights into the binding of the diarylquinoline TMC207 to the ATP synthase
C-ring structure. Antimicrobial agents and chemotherapy, 56(5), pp.2326-2334.
3. Huitric, E., Verhasselt, P., Koul, A., Andries, K., Hoffner, S. and Andersson, D.I., 2010. Rates and mechanisms
of resistance development in Mycobacterium tuberculosis to a novel diarylquinoline ATP synthase inhibitor.
Antimicrobial agents and chemotherapy, 54(3), pp.1022-1028.
4. Biuković, G., Basak, S., Manimekalai, M.S.S., Rishikesan, S., Roessle, M., Dick, T., Rao, S.P., Hunke, C. and
Grüber, G., 2013. Variations of Subunit ε of the Mycobacterium tuberculosis F1Fo ATP Synthase and a Novel
Model for Mechanism of Action of the Tuberculosis Drug TMC207. Antimicrobial agents and chemotherapy,
57(1), pp.168-176.
5. Hotra, A., Suter, M., Biuković, G., Ragunathan, P., Kundu, S., Dick, T. and Grüber, G. (2016) Deletion of a
unique loop in the mycobacterial F-ATP synthase γ subunit sheds light in its inhibitory role in ATP hydrolysis
driven H+-pumping. FEBS J. in press (doi: 10.1111/febs.13715).
6. Van Kessel, Julia C., and Graham F. Hatfull. "Efficient point mutagenesis in mycobacteria using
single‐stranded DNA recombineering: characterization of antimycobacterial drug targets." Molecular
microbiology 67.5 (2008): 1094-1107.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
359
330 Drug design-new developments
23/09/2016
Efficient nanoencapsulation of Bedaquiline
L. De Matteis6, D. Jary1-2, A. Lucía8-4-3, I. Serrano7, S. García6, F. Navarro1-2, J. M. De La Fuente7,
J.A. Ainsa8-4-3-5
1
Technologies for Healthcare and Biology division, CEA, LETI, MINATEC Campus 2Univ. Grenoble
Alpes, Grenoble, France 3Instituto de Investigacion Sanitaria de Aragon (IIS-Aragon) 4CIBER de
Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III 5Instituto de Biocomputacion
y Fisica de Sistemas Complejos, BIFI 6Instituto de Nanociencia de Aragon (INA), Universidad de
Zaragoza 7Instituto de Ciencia de Materiales de Aragon (ICMA), Universidad de ZaragozaCSIC 8Dpto. Microbiología, medicina preventiva y salud publica. Grupo Genética de
Micobacterias., University of Zaragoza, Zaragoza, Spain
The problem of antibiotic resistance in Mycobacterium tuberculosis has been declared a global
health emergence by the WHO. Rising cases of MDR-TB, and XDR-TB are making increasingly
difficult to treat tuberculosis nowadays (1).
After 50 years without any new drug for TB, in 2012 the FDA approved the use of bedaquiline, the
first drug designed specifically to treat MDR-TB. It is very effective but shows serious side effects.
Consequently, this antibiotic should be prescribed only when no other treatment options are
available (2). Besides classical drug discovery strategies, new approaches are urgently needed to
get a faster, more efficient and less harmful treatment.
Novel drug delivery systems based on nanocarriers are a promising strategy to overcome current
therapeutic challenges due to their unique physicochemical properties (3, 4), which include small
size or high surface to volume ratio. Nanocarriers improve the aqueous solubility of poorly soluble
drugs, protects the drugs, and allow a controlled release of the medication to decrease the
frequency of administration. Additionally they can be modified to control their biodistribution allowing
selective transport to the sites of infection. The development of effective and safe nanotherapy
methods is particularly relevant in the treatment of MDR-TB, as very long treatments with highly
toxic second-line drugs are required for those cases. In this sense the encapsulation of bedaquiline
in novel nanocarriers is of special interest (5).
In this work, chitosan based nanocapsules and Lipid NanoParticles, based on the Lipidots®
technology, have been synthesized for the encapsulation of bedaquiline. The encapsulation of this
antibiotic has been optimized for both nanocarriers, upon quantification of drug loading efficiency,
improvement of the drug payload, nanoparticles stability amelioration in storage condition. For the
best candidates, drug release has been determined in biological media for in vitro assays. The
antimycobacterial activity has finally been evaluated to confirm that the drug is still active after
encapsulation. Also, their cytotoxicity has been assayed in different cell lines.
1. WHO, Drug resistant TB: surveillance and response. Supplement to global tuberculosis report
2014. Available in
http://www.who.int/tb/publications/global_report/gtbr14_supplement_web.pdf
2. Mingote LR, Namutamba D, Apina F, Barnabas N, Contreras C, Elnour T, Frick MW, Lee C,
Seaworth B, Shelly D, Skipper N, Tavora dos Santos Filho E. Lancet. 2015, 385: 477-479.
3. Mishra B, Patel BB, Tiwari S. Nanomedicine: NBM. 2010, 6: 9-24.
4. Onoue S, Yamada S, Chan HK. Int J Nanomed. 2014; 9: 1025-1037.
5. Mustafa S, Pai RS, Singh G, Devi K. J Drug Targeting. 2015, 23: 287-304.
360
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
331 Drug design-new developments
23/09/2016
An optimized background regimen with the next generation benzothiazinone PBTZ169 for
active tuberculosis
A. Lupien3, A. Vocat3, B. Lechartier2, V. Makarov1, S. Cole3
1
Bakh Institute Of Biochemistry, Russian Academy of Science, Moscow, Russia 2Service de
pneumologie, CHUV 3Global Health Institute, École polytechnique fédérale de Lausanne (EPFL),
Lausanne, Switzerland
Tuberculosis (TB) mainstay treatment relies on a four-drug regimen including isoniazid, rifampicin,
pyrazinamide (PZA) and ethambutol for six months. The emergence of multidrug (MDR) and
extensively drug-resistant (XDR) TB has now challenged the efficiency of this treatment to cure TB.
Intensive efforts have been made to develop new anti-TB drugs. Several of these novel or
repurposed drugs are in clinical trials to evaluate their efficacy at curing TB. We are focusing on the
promising piperazine-containing benzothiazinone PBTZ169 that inhibits the essential flavo‐protein
enzyme DprE1. PBTZ169 is active against MDR‐ and XDR‐clinical isolates of Mycobacterium
tuberculosis. When combined with other TB drugs, PBTZ169 acts synergistically with bedaquiline
(BDQ) and clofazimine (CFM). A new regimen comprising PBTZ169, BDQ and PZA was found to be
more efficacious than the standard three-drug treatment in a murine model of chronic disease.
PBTZ169 is therefore an attractive drug candidate to treat TB in humans.
The aim of this project is to determine the activity in vitro of PBTZ169 in combination with anti-TB
drugs, to elaborate a potential OBR (optimized background regimen) against active TB. To do so,
PBTZ169 was tested against active M. tuberculosis using two-drug checkerboard and viability
assays. PBTZ169 has additive activity when it is combined with first- and second-line anti-TB drugs.
At the MIC, PBTZ169 combined with BDQ, CFM and delamanid produce a two log reduction in the
bacterial burden (synergism) compared to the drugs alone. For all the other drugs tested in
combination with PBTZ169 (meropenem, clarithromycin, sutezolid, linezolid and lansoprazole
sulfide), we observed additive activity. Neither antagonism nor increased cytotoxicity were found
suggesting that PBTZ169 could be added to most drug combinations without any adverse effects.
Interestingly, several two-drug combinations that do not include PBTZ169 showed concentrationdependent synergism. Further studies on these two-drug combinations using ex vivo models and
three-drug regimens are ongoing.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
361
332 Drug design-new developments
23/09/2016
Killing and sensitizing of Mycobacterium tuberculosis to antibiotics and TB drugs using a
protein complex from human milk
V. Meikle2, A.P. Håkansson1, M. Niederweis2
1
Experimental Infection Medicine, Malmö, Lund University, Lund, Sweden 2Microbiology, University
of Alabama at Birmingham, Birmingham, United States
Mycobacterium tuberculosis (Mtb) is intrinsically resistant to many antibiotics which are effective
against other bacteria. Increasingly infections with Mtb occur with drug resistant strains that cause
TB with very poor prognosis. The development of new antibiotics is hugely expensive and return of
investment is low. An alternative strategy is to sensitize bacteria to current antibiotics e.g. by drug
efflux pump inhibitors. We used HAMLET (human alpha-lactalbumin made lethal to tumor cells), a
complex of an altered form of human milk alpha-lactalbumin with fatty acids, that uses a novel death
pathway to kill bacterial pathogens such as Streptococcus pneumoniae and examined whether
HAMLET is active against Mtb. Using the microplate Alamar blue assay we showed that HAMLET
completely inhibited growth of Mtb H37Rv at a concentration of 200 µg/mL. Non-toxic amounts of
HAMLET potentiated the efficacy of antibiotics and TB drugs by 12 to 130-fold. HAMLET also killed
Mtb in infected THP-1 macrophages with a minimal inhibitory concentration of 200 µg/ml, while
concentrations up to 400 µg/ml did not affect macrophage viability. This result demonstrates that
HAMLET is active against intracellular Mtb. The inhibitory activity of HAMLET against Mtb and its
large potentiating effect on antibiotics and TB drugs, open an exciting avenue for developing safe,
alternative and more efficient treatment regimens which do not require the development of new
drugs, may overcome the resistance of current MDR-, XDR- and TDR-Mtb strains and may include
antibiotics which, by themselves, are not effective against Mtb.
362
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
333 Drug design-new developments
23/09/2016
Design, Syntheses and Studies of New Potent and Selective Anti-tuberculosis Agents
Based on Imidazopyridine and Imidazothiazole Cores
G. Moraski
Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, United States
Multidrug (MDR) and extensive drug resistant (XDR) forms of TB are becoming common. New
treatments are desperately needed. Our early studies focused on syntheses of mycobactins
(essential virulence factors for Mtb, that facilitate iron uptake), analogs and drug conjugates.
Simultaneous fragment based screening of all synthetic intermediates led to the discovery of new
small molecule anti-TB agents including appropriately functionalized imidazopyridines and
imidazothiazoles that have remarkably potent anti-TB activity.
In order to fully explore the potential of this new class of anti-TB agents, collaborations were formed
with the Lilly TB initiative, the Infectious Disease Research Institute (IDRI) and the NIH, including
the laboratories of Dr. Clif Barry, and Prof. Jeffrey Schorey’s group at Notre Dame. The early
chemistry developed by Garrett Moraski at Notre Dame (now Montana State University) was readily
adapted to the Automated Synthesis and Purification Laboratory (ASPL) at Eli Lilly and Co and
allowed further expansion of SAR. Additional collaborative efforts, including high throughput
screening (University of Illinois, Chicago), development and utilization of separate sensitive assays
(IDRI) and determination of in vivo pharmacokinetics at Lilly and the NIH and elucidation of the
mode of action (inhibition of QcrB) have accelerated studies that have advanced the studies to
determine in vivo efficacy in rodents and primates. The combined results that will be described
indicate that imidazo[1,2-a]pyridine-3-carboxamides and the corresponding imidazothiazoles are
exciting, potent, selective anti-TB agents that merit additional development opportunities. While
chemistry is the driver, substantial navigation of the project has been due to productive
interdisciplinary collaborations.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
363
334 Drug design-new developments
23/09/2016
A tail tale: what does it take to get under TB skin
W. Moreira1, D. Aziz1, C. Huan2, T. Yang2, S. Nyantakyi2, M.L. Go2, T. Dick1
1
Microbiology 2Pharmacy, National University of Singapore, Singapore, Singapore
Resistance and persistence represent two major challenges faced by current anti-TB regimens. TB
drugs, like most antibiotics, target specific macromolecules. Genetically acquired antibiotic
resistance arises mainly from mutations in target genes and occurs readily in patients due to noncompliance to lengthy multi-drug regimens and spatio-temporal pockets of monotherapy.
Persistence of infection is thought to be due to various types of metabolically quiescent non-growing
drug tolerant bacteria. Ideally new TB drugs should kill persister bacilli and show a low propensity
for the development of genetic drug resistance. The bacterial membrane integrity and its associated
homeostatic functions (e.g. selective permeability, energy metabolism) remains an essential and
vulnerable target regardless of the metabolic status of the cell. Additionally, resistance development
to membrane-targeting antibiotics is expected to be slow as the target is the product of complex
biosynthetic pathways, as opposed to a single macromolecule. The large number of antimicrobial
peptides made by the host validates the membrane as an antibacterial target site. Furthermore, the
successful clinical use of a few membrane-targeting antibiotics (e.g. colistin, polymixin, daptomycin)
supports this notion. Remarkably, these molecules are inactive against mycobacteria. These large
compounds (MW >3000) are possibly blocked by the outer membrane and cannot reach the
cytoplasmic membrane of mycobacteria. It is interesting to note that most of these large molecules
possess a lipophilic side chain that favours their anchoring in the membrane. We hypothesized that
a lipophilic side chain could suffice to target a small molecule to the cytoplasmic membrane. Such
small molecule would not be blocked by the outer membrane. We made use of the widely use
indole scaffold to design a small molecule that interferes and disrupt mycobacterial membrane
integrity and functions. We provide chemical and biochemical evidences suggesting that these
compounds compromise the membrane integrity causing depolarization and disruption of energy
metabolism leading to cell death. Induction of promoter-reporter associated with envelope stresssensing pathway further support these findings. These compounds were active against non-growing
persister bacilli and spontaneous resistant mutants could not be isolated (mutation frequency <109).
Taken together these results suggest that targeting mycobacterial membrane integrity may be an
attractive chemotherapeutic intervention level to kill otherwise drug tolerant persister bacilli, and to
slow down the development of genetic antibiotic resistance.
364
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
335 Drug design-new developments
23/09/2016
Membrane targeting drugs against mycobacteria - a road less travelled
D. Mukherjee2, H. Zou4, S. Liu1-4, R. Beuerman1-3-4, T. Dick2
1
SRP Neuroscience and Behavioural Disorders, Duke-NUS Graduate Medical School 2Department
of Microbiology & Immunology 3Department of Ophthalmology, National University of
Singapore 4Singapore Eye Research Institute, Singapore, Singapore
Aims: It is well known that the major issues with TB treatment are resistance and persistence.
Drugs with novel mechanisms of action are needed to help address these issues. We test the
hypothesis that targeting the cytoplasmic membrane may be an effective way to kill persister
mycobacteria as well as delay the emergence of resistance.
Methods: In vitro activity of AM-0016, a novel semi-synthetic xanthone-based antibacterial, was
assessed against growing and persister mycobacteria. Resistance mutation frequencies were also
determined. Membrane potential was assessed biochemically and electron microscopic analyses
were carried out.
Results: AM-0016 rapidly sterilized growing tubercle bacillus cultures and displayed strong
bactericidal activity against hypoxic and drug induced persister bacteria. Spontaneous resistance
mutation frequency was lower than 10-8. Exposure to AM-0016 resulted in rapid collapse of the
membrane potential. Imaging revealed deformation of the cell envelope.
Conclusions: Targeting the cytoplasmic membrane may be an attractive approach to eliminate
persister mycobacteria and slow down the emergence of genetic drug resistance.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
365
336 Drug design-new developments
23/09/2016
Fragment-based Mycobacterium tuberculosis whole cell screen delivers hits for drug
discovery
D. Negatu2-3, J. Liu1, C. Aldrich4, M. Gengenbacher1-2, T. Dick1-3
1
Yong Loo Lin School of Medicine, Department of Medicine, BSL-3 Core Facility 2Yong Loo Lin
School of Medicine, Department of Medicine, Tuberculosis Research Laboratory 3Yong Loo Lin
School of Medicine, Department of Microbiology and Immunology, Antibacterial Drug Discovery
Laboratory, National University of Singapore, Singapore, Singapore 4Department of Medicinal
Chemistry, University of Minnesota, Minneapolis, Minnesota, United States
Mycobacterium tuberculosis (Mtb) is now the leading cause of infectious disease mortality by a
single pathogen. New drugs, ideally with novel mechanism (s) of action are needed to treat drugresistant strains and simplify the current lengthy treatment regimen. Many TB drugs (isoniazid,
pyrazinamide, para-aminosalicylic acid, ethionamide) are small molecular weight compounds that
are bioactivated to reactive metabolites that inhibit multiple metabolic pathways. By todays
standards, these drugs would be considered fragments owing to their low molecular weight
(MW<170), a property which would generally exclude them from most compound collections used in
high-throughput screening (HTS). We hypothesize that fragment libraries represent a rich source of
untapped chemical matter for the identification of new antitubercular agents, which may act through
multi-targeting and potentially by bioactivation. We screened a library of 1000 chemically diverse
fragment compounds. At a concentration of 1 mM, the growth inhibition of M. tuberculosis H37Rv
measured by turbidity delivered 84 (8.4%) primary hits at 70% cut off. Of the hits, 33 compounds
showed at least 50% growth inhibition at 500 µM in dose response assays, which is comparable
with the MIC50 of pyrazinamide, a first line TB drug. Fifteen compounds also displayed activity
against the clinically relevant non-tuberculosis mycobacteria species, Mycobacterium avium and
Mycobacterium abscessus, exhibiting at least 50% growth inhibition at 500 µM. Further profiling of
the hits for membrane toxicity (hemolysis assay) and cytotoxicity (HepG2, HEK, A549) identified 19
compounds with acceptable hemolytic activity and selectivity (Selectivity Index > 10) in tested cell
lines. Our findings suggest that fragment libraries may be valuable resources for Mtb drug discovery
delivering low molecular weight hits with attractive physicochemical properties.
366
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
337 Drug design-new developments
23/09/2016
Evaluating indazole sulfonamides targeting IMPDH as potential leads against
Mycobacterium tuberculosis
Y. Park4, K. Rhee6, T. Ioerger5, J. Sacchettini5, V. Dartois7, T. Blundell2, M. Rizzi1, S. Donini1,
K. Arora4, L. Via4, P. Ray3, S. Green3, C. Barry4, H. Boshoff4
1
Dipartimento di Scienze del Farmaco, University of Piemonte Orientale, Via Bovio,
Italy 2Department of Biochemistry, University of Cambridge, Cambridge 3Drug Discovery Unit,
University of Dundee, Dundee, United Kingdom 4Tuberculosis Research Section, National Institute
of Allergy and Infectious Diseases, Bethesda 5Department of Computer Science and Engineering,
Texas A&M University, College Station 6Division of Infectious Diseases, Department of Medicine,
Weill Cornell Medical College, New York 7Public Health Research Institute, New Jersey Medical
School, Newark, United States
A potent indazole sulfonamide was identified through high-throughput screening of Mycobacterium
tuberculosis (Mtb). This non-cytotoxic compound did not directly inhibit cell wall biogenesis but
triggered a slow lysis of Mtb cells as measured by release of intracellular GFP. Isolation of resistant
mutants followed by whole-genome sequencing showed in two cases an unusual gene amplification
of a 40 gene region spanning Rv3371 to Rv3411c and in one case a potential promoter mutation
upstream of guaB2 (Rv3411c) encoding inosine monophosphate dehydrogenase (IMPDH).
Subsequent biochemical validation confirmed direct inhibition of IMPDH by an uncompetitive mode
of inhibition and growth inhibition could be potentiated by supplementation with guanine, a bypass
mechanism for the IMPDH pathway. Further development of SAR allowed good correlation of
IMPDH IC50 values and minimum inhibitory concentration (MIC) against Mtb and allowed active
derivatives to be synthesized with a chemical linkage permitting bead attachment. Beads containing
these immobilized derivatives specifically interacted with IMPDH in cell lysates. X-ray
crystallography of the IMPDH-IMP-inhibitor complex revealed that the primary interactions of these
compounds with IMPDH were direct pi-pi interactions with the IMP substrate. Advanced lead
compounds in this series with acceptable pharmacokinetic properties failed to show efficacy in
acute or chronic murine models of tuberculosis (TB). Time-kill experiments in vitro suggest that
sustained exposure to drug concentrations above MIC for 24 hours were required for a cidal effect,
levels that have been difficult to achieve in either mice or marmosets. It also requires rapid growth of
Mtb for cidal activity in vitro growth rate dependent killing assay. In addition direct measurement of
guanine levels in resected lung tissue from tuberculosis patients revealed 0.5-2 mM concentrations
in caseum and normal lung tissue. The high lesional levels of guanine and the slow lytic effect of
IMPDH inhibition pose challenges to developing drugs against this target for use in treating TB.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
367
338 Drug design-new developments
23/09/2016
Structure-activity relationships and biological profile of novel diarylthiazoles as antitubercular drugs
G. Prosser2, N. Kalia2, S. Green1, P. Ray1, H. Boshoff2, C. Barry2
1
Drug Discovery Unit, University of Dundee, Dundee, United Kingdom 2Tuberculosis Research
Section, NIH, Bethesda, United States
Novel anti-tubercular drugs that can reduce treatment duration and are active against drug resistant
strains are urgently needed to combat disease spread and progression. Using high-throughput
screening of synthetic compound libraries we have identified a novel diarylthiazole scaffold as
having potent anti-tubercular activity, under both aerobic and anaerobic conditions and within
infected J774 macrophages. Structure-Activity Relationship studies have identified key aspects of
the core scaffold that can be modified to increase potency and solubility, and a lead candidate has
demonstrated promising oral exposure in a murine model. Spontaneous resistant mutants raised
against a series lead compound contained mutations within the prrB gene, coding for the sensorhistidine kinase partner of the PrrAB two-component system. Over-expression of the operon in
H37Rv via an integrative vector failed to induce significant alterations in drug MICs. Lack of
knowledge on the physiological role(s) of this TCS stimulated us to perform genetic and biochemical
studies on the operon and its individual components. We present data on drug-protein interactions
in vitro between select diarylthiazoles and purified recombinant PrrB and each of its distinct
domains, and also identify other potential native mycobacterial protein binding partners using a
drug-immobilized affinity-based pull-down assay. The physiological role of the operon has been
investigated through elucidation of the consensus DNA binding motif of the PrrA response regulator
and subsequent genome-wide in silico analysis of putative PrrA-promoter binding sites. Genetic
essentiality of the operon and each of its individual components in both M. tuberculosis and M.
smegmatis was demonstrated through an inability to delete the corresponding gene(s) in the
absence of a second copy integrated elsewhere in the genome. We are currently generating
regulated knock-down strains for each gene and plan to identify regulon members using microarray
technology. Together, our results identify a promising novel anti-tubercular drug-target pair and
provide insight into the physiological role(s) of the enigmatic and essential PrrAB two-component
system in Mycobacteria.
368
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
339 Drug design-new developments
23/09/2016
Repurposing clinically approved cephalosporins for tuberculosis therapy
S. Ramon Garcia1-2, R. González Del Río2, A. Santos Villarejo2, G. Sweet1, F. Cunningham2,
D. Barros2, L. Ballell2, A. Mendoza-Losana2, S. Ferrer-Bazaga2, C. Thompson1
1
Microbiology & Immunology, The University of British Columbia, Vancouver, Canada 2Diseases of
the Developing World, GlaxoSmithKline, Tres Cantos, Spain
While modern cephalosporins developed for broad spectrum antibacterial activities have never been
pursued for tuberculosis (TB) therapy, we explored their potential repositioning as new anti-TB
drugs and identified first generation cephalosporins having clinically relevant inhibitory
concentrations, both alone and in synergistic drug combinations.
We previously demonstrated that combinatorial drug therapy, traditionally designed to avoid
emergence of drug resistant Mycobacterium tuberculosis (Mtb) strains, might also be employed to
increase the efficacies of available antibiotics, allowing them to be repurposed for TB therapy within
synergistic combinations1. Rifampicin, the cornerstone drug for TB therapy, is not administered at its
optimal clinical dose due to long-established toxicity concerns. If the anti-mycobacterial activity of
rifampicin could be increased, TB therapy could be shortened, thus reducing the rate of
transmission and the emergence of drug resistance2.
In pursuit of this vision, we screened an in-house library of ca. 600 commercially available
antibiotics, and found that the cephalosporins had strong synergies with rifampicin and ethambutol,
a first-line anti-TB drug. In addition, common chemical patterns required for single drug activity
against Mtb were identified using structure-activity relationships (SAR) studies. Synergy was also
observed even under intracellular growth conditions where beta-lactams typically have limited
activities. Cephalosporins and rifampicin were 4- to 64-fold more active in combination than either
drug alone; however, limited synergy was observed with rifapentine or rifabutin. Clavulanate was a
key synergistic partner in triple combinations. Cephalosporins (cefadroxil; or other beta-lactams)
and clavulanate rescued the activity of rifampicin against a rifampicin resistant strain. Uptake
experiments demonstrated that synergy was not due exclusively to increased rifampicin
accumulation within the mycobacterial cells. Cephalosporins were also synergistic with new anti-TB
drugs such as bedaquiline and delamanid.
The fact that these cephalosporins are orally bioavailable with good safety profiles, together with
their anti-mycobacterial activities reported here, suggest that they could be repurposed within new
combinatorial TB therapies.
References:
1. Ramon-Garcia, S. et al. Synergistic drug combinations for tuberculosis therapy identified by a
novel high-throughput screen. Antimicrob Agents Chemother 55, 3861-3869 (2011).
2. van Ingen, J. et al. Why Do We Use 600 mg of Rifampicin in Tuberculosis Treatment? Clin
Infect Dis 52, e194-199 (2011).
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
369
340 Drug design-new developments
23/09/2016
Identification and prioritisation of hits from whole cell phenotypic screening of
mycobacterium tuberculosis in the context of treatment shortening, lessons and future
directions
P. Ray2, H. Boshoff3, K. Arora3, P. Tsang3, T. Bayliss2, J. Harrison2, D. Murugesan2, K. Buchanan2,
S. Green2, F. Zuccotto2, K. Read2, P. Scullion2, R. Epemolu2, C. Mackenzie2, V. Mizrahi1,
D. Warner1, C. Barry3, P. Wyatt2
1
Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town,
South Africa 2Drug Discovery Unit, University of Dundee, Dundee, United Kingdom 3Laboratory of
Clinical Infectious Diseases, National Institute of Allergy and Infectious Disease, National Institutes
of Health, Bethesda, United States
Results of a phenotypic M. tuberculosis H37Rv screening campaign against two independently
compiled commercial libraries of over five hundred thousand compounds and a proprietary
agrochemical library of over fifty five thousand compounds is disclosed. In addition to conventional
HTS triage processes, which include selecting starting points based on physicochemical properties
and cytotoxicity, we introduce early H37Rv biology, ADME-Tox and physicochemical filters to aid
prioritisation of novel starting points which are envisaged more likely to lead to treatment shortening
drug discovery projects. A robust process for evaluating and prioritising >100 confirmed hit starting
points for synthetic lead-generation and mechanism of action activities is described. Future
directions include the need for screening untapped areas of chemical space beneficial for lesion and
TB pentration.
370
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
341 Drug design-new developments
23/09/2016
Synthesis and evaluation of new benzothiazinones with piperazinylamide side chain
A. Richter1, T. Wetzlar1, K. Voigt2, U. Möllmann2, S. Franzblau3, P. Imming1
1
Martin Luther Universität Halle Wittenberg, Halle 2Leibniz Institute for Natural Product Research
and Infection Biology Hans-Knöll-Institute, Jena, Germany 3Institute for Tuberculosis Research,
University of Illinois, Chicago, United States
Benzothiazinones (BTZs) display very strong antimycobacterial activity in vitro [1]. Some derivatives
of this compound class, for example BTZ043 [2] and PBTZ169 [3], are very active against
Mycobacterium tuberculosis with MICs < 1 nM. The outstanding in vitro activity has not yet been
fully translated into in vivo effectivity, thus requiring relatively high doses of 50 – 200 mg/kg daily in
order to reduce CFUs in mice considerably [1, 4].
BTZs inhibit the mycobacterial enzyme decaprenylphosphoryl-β-D-ribose-2-epimerase (DprE1),
interfering with the construction of a functioning cell wall. Important for the activity is a nitro group in
the molecule, which is reduced by FADH2/DprE1 to a nitroso BTZ that binds covalently to the thiol
group of Cys-387 near the catalytic center [5,6]. In Fig. 1 we depict the complex process of this twostep mechanism-based inhibition:
Figure 1. Steps of DprE1 inhibition by BTZs
We synthesized a panel of new benzothiazinones based on the scaffold shown in Fig. 2 with the
purpose of improving aqueous solubility while maintaining high activity. The synthesis was possible
by an efficient one step pathway using thioureas.
Figure 2. Scaffold of BTZs synthesised and studied (X = c or S, n = 1 or 2)
The set contains different amide as well as sulfonamide side chains at the R1 position. Activities
covered a wide range from 1000 nM to 0.2 nM (MICs against M. vaccae). Some of the new BTZs
reached or even surpassed the activity of the lead substance BTZ043 in this assay.
Solubility data of these compounds were generated by CLND (chemiluminescence nitrogen
detection) measurements. Although we focused on hydrophilic side chains, the solubility was equal
or even slightly lower compared to BTZ043.
Furthermore, the nitro group was replaced with other groups that have less potential of toxicity while
maintaining the covalent mode of inhibition.
1. Makarov, V., et al.: Science 2009, 324, 801-804
2. Möllmann, U., Makarov, V., Cole, S. T.: WO2009010163A1 2009
3. Makarov, V., Cole, S. T.: WO2012/066518A1 2012
4. Makarov, V., et al.: EMBO Mol. Med. 2014, 6 (3), 372-383
5. K. Mikusová et al., Curr. Pharm. Design 2014, 20, 4249-4273
6. Trefzer, C., et al.: J. Am. Chem. Soc. 2012, 134 (2), 912-915
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
371
342 Drug design-new developments
23/09/2016
Structure-activity relationships of 3,5-dinitrophenyltetrazoles, oxadiazoles and triazoles as
selective antitubercular agents
J. Roh1, J. Nemecek1, G. Karabanovich1, L. Valaskova1, J. Stolaříková2, V. Klimešová1,
A. Hrabalek1
1
Charles University in Prague, Faculty of Pharmacy, Hradec Králové 2Regional Institute of Public
Health, Ostrava, Czech Republic
In
our
previous
work
we
found
that
1-substituted
and
2-substituted-5-(3,5dinitrobenzylsulfanyl)tetrazoles possessed highly selective antitubercular activity with minimal
inhibitory concentration (MIC) values as low as 1 µM against drug susceptible strains of
M. tuberculosis and 0.25 - 1 µM against six clinically isolated MDR M. tuberculosis strains. The
further studies of these compounds and their analogues led to the development of 5-substituted 2(3,5-dinitrobenzylsulfanyl)oxadiazoles and thiadiazoles as novel antitubercular agents with
outstanding activities against replicating and also nonreplicating strains of M. tuberculosis.
Furthermore, this class of compounds possessed highly selective antimycobacterial effect, because
they showed no antibacterial or antifungal activity, low cytotoxicity in mammalian cell lines and low
genotoxicity. Despite the presence of 3,5-dinitrophenyl fragment, as in the potent inhibitors of
mycobacterial decaprenylphosphoryl-β-D-ribofuranose 2´-oxidase (DprE1), these compounds did
not inhibit DprE1 and more likely affect the synthesis of mycobacterial nucleic acids.[1]
In the continuation of our work we further studied the structure-activity relationships of these lead
compounds. In particular, we decided to remove the methylsulfanyl linker between the 3,5dinitrophenyl moiety and the heterocycle. Hence, the series of 3,5-dinitrophenyl-substituted
tetrazoles, oxadiazoles and triazoles, with 3,5-dinitrophenyl group attached directly to heterocycle,
were prepared. The change of the position of 3,5-dinitrophenyl fragment had no effect on the level
of antimycobacterial activity - the majority of prepared compounds possessed excellent in vitro
activities against M. tuberculosis H37Rv and several MDR stains of M. tuberculosis, with MIC values
as low as 0.03 µM. These compounds again showed highly selective antimycobacterial effect,
because they were not active against other bacteria or fungi and possessed low in vitro cytotoxicity.
The study was supported by the Czech Science Foundation project (14-08423S) and Charles
University in Prague (SVV 260 291).
1. G. Karabanovich et al. J. Med. Chem. 2016, 59, 2362−2380.
372
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
343 Drug design-new developments
23/09/2016
Combining the shotgun and sniper approach to target the mycobacterial cell wall
A. Shetty2, U. Lakshmanan1, C. Ling1, A. Poulsen1, J. Hill1, Y. Yamada2, T. Dick2
1
Experimental and Therapeutics Centre, A* STAR 2Microbiology and Immunology, National
University of Singapore, Singapore, Singapore
Discovering new drugs for tuberculosis (TB) is like playing darts with spaghetti: promising lead
molecules that are potent enzyme inhibitors but cannot penetrate the mycobacterial cell. Our goal is
to identify attractive targets with their associated whole-cell active lead compounds for development
of new clinical candidates. To achieve this, we have employed a pathway-based whole-cell screen
approach, which allows screening against a defined pathway inside the bacterial cell using the
double-membrane barrier as a filter. Of particular interest is the cell wall biosynthesis pathway
because it is well validated as a drug target.
Our study design employs a two-tier screening strategy for hit identification. First, a high-throughput
screen of over 70,000 chemically diverse scaffolds for growth inhibitors of Mycobacterium bovis
BCG (M. bovis BCG), which led to the selection of 18 compounds. Second, a pathway-based
screen of the primary hits in a M. bovis BCG reporter strain carrying a cell wall stress inducible
promoter, iniBAC fused to the mCherry reporter gene. Differential expression of the reporter gene
allowed us to identify a new class of cell wall targeting compounds, E10 and E11. Both compounds
are non-cytotoxic and have a minimum inhibitory concentration (MIC50) of about 10µM against M.
bovis BCG and M. tuberculosis. Currently, we are carrying out efficacy studies against persisters
and target deconvolution by chemical genetics (resistance generation coupled with whole genome
sequencing).
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
373
344 Drug design-new developments
23/09/2016
Discovery and development of inhibitors of Mycobacterium tuberculosis primase and
inorganic pyrophosphatase
O. Tsodikov
Department of Pharmaceutical Sciences, University of Kentucky, Lexington, United States
Innovative strategies are needed to discover novel anti-tuberculosis therapeutics, as alternatives
are needed to existing antibiotics that suffer from resistance, toxicity and ineffective mechanisms of
action. Recently we developed a novel coupled primase-pyrophosphatase assay for discovery of
inhibitors of two essential bacterial enzymes, primase (DnaG) and inorganic pyrophosphatase
(PPiase). DnaG is responsible for synthesis of RNA primers for chromosomal DNA replication, and
PPiase breaks down inorganic pyrophosphate (PPi) to drive all DNA and RNA synthesis reactions in
the cell. We applied this assay to discovery of inhibitors of DnaG and PPiase from Mycobacterium
tuberculosis. The high-throughput assay of a library of small molecules yielded low-microM
inhibitors of primase and inorganic pyrophpsphatase. We obtained crystal structures of the RNA
polymerase domain of M. tuberculosis DnaG and inorganic pyrophosphatase alone and in complex
with inhibitors, which clarify the mechanisms of catalysis and inhibition and drive medicinal
chemistry efforts to develop these compounds into therapeutic candidates.
References
1. Biswas T, Resto-Roldan E, Sawyer SK, Artsimovitch I and Tsodikov OV. (2013) A novel nonradioactive primase-pyrophosphatase activity assay and its application to the discovery of
inhibitors of Mycobacterium tuberculosis primase DnaG. Nucleic Acids Res., 41, e56.
2. Gajadeera C#, Willby M#, Green KD, Shaul P, Fridman M, Garneau-Tsodikova S*, Posey JE*
and Tsodikov OV*. (2015) Antimycobacterial activity of DNA intercalator inhibitors of
Mycobacterium tuberculosis primase DnaG. J. Antibiot., 68, 153-157. (# first co-authors, * cocorresponding authors)
3. Pratt AC, Dewage SW, Pang AH, Biswas T, Barnard-Britson S, Cisneros GA* and Tsodikov
OV*. (2015) Structural and computational dissection of the catalytic mechanism of the
inorganic pyrophosphatase from Mycobacterium tuberculosis. J. Struct Biol., 192, 76-87. (*
co-corresponding authors)
374
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
345 Drug design-new developments
23/09/2016
Pyrrolo[3,4-c]pyridine-1,3(2H)-diones: A novel anti-mycobacterial class targeting
mycobacterial respiration
R. Van Der Westhuyzen1, A. Moosa3, D. Warner3-2, L. Street1, K. Chibale1-2
1
Drug Discovery and Development Centre (H3-D) 2Institute of Infectious Disease and Molecular
Medicine 3MRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology,
University of Cape Town, Cape Town, South Africa
High throughput screening of a library of small polar molecules against Mycobacterium tuberculosis
led to the identification of a phthalimide-containing ester hit compound, which was optimized for
metabolic stability by replacing the ester moiety with a methyl oxadiazole bioisostere. A route
utilizing polymer-supported reagents was designed and executed to explore structure activity
relationships with respect to the N-benzyl substituent, leading to compounds with nanomolar
activity. The frontrunner compound from these studies was well tolerated in mice. A M. tuberculosis
cytochrome bd oxidase deletion mutant (∆cydKO) was hyper-susceptible to compounds from this
series, and a strain carrying a single point mutation in qcrB, the gene encoding a subunit of the
menaquinol cytochrome c oxidoreductase, was resistant to compounds in this series. In
combination, these observations indicate that this novel class of anti-mycobacterial compounds
inhibits the cytochrome bc1 complex, a validated drug target in M. tuberculosis.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
375
346 Drug design-new developments
23/09/2016
Unexpected high prevalence of resistance-associated Rv0678 variants in MDR TB patients
without documented prior use of clofazimine or bedaquiline
C. Villellas2, N. Coeck1, C. Meehan1, N. Lounis2, S. Niemann3, B. De Jong1, L. Rigouts1, K. Andries2
1
Institute of Tropical Medicine, Antwerp 2Janssen Research and Development, Beerse, Belgium
3
National Reference Centre for Mycobacteria, Borstel, Germany
Objectives: Resistance-associated variants (RAVs) in Rv0678, a gene regulating the expression of
the MmpS5-MmpL5 efflux pump, have been shown to lead to 2- to 8- fold increases in bedaquiline
(BDQ) MIC, and 2- to 4-fold increases in clofazimine (CFZ) MIC. The prevalence of these Rv0678
RAVs in clinical isolates and their impact on treatment outcomes are important factors to take into
account in BDQ treatment guidelines. After discovering an Rv0678 RAV in a baseline isolate from a
patient without documented prior use of CFZ or BDQ, the prevalence of these mutations in clinical
isolates of TB patients was assessed. The potential effect of rifampicin in selecting Rv0678 mutants
was investigated.
Methods: Baseline isolates from two BDQ multidrug-resistant (MDR)-TB clinical trials were
assessed for Rv0678 RAVs by Sanger sequencing and corresponding BDQ MICs were determined
on 7H11 agar. Rv0678 RAVs were also investigated in non-MDR-TB sequences of the populationbased Hamburg cohort, using the SNP-calling pipeline Snippy. The comparative fitness of 2
fluorescent-marked strains (rpoB-Rv0678 double mutant and rpoB single mutant) was tested by flow
cytometry in presence of a range of rifampicin concentrations.
Results: Rv0678 RAVs were identified in 21/345 (6.1%) of MDR-TB baseline isolates. BDQ MICs of
these isolates were either normal (0.03 to 0.24 µg/ml, n=10), high (0.48 to 1, n=8), or low (0.004 to
0.015, n=4). A variant at position -11 in the intergenic region between Rv0678 and Rv0677c in 11%
of the baseline isolates appeared to increase the sensitivity for BDQ (median MIC = 0.015). In nonMDR-TB isolates, the frequency of Rv0678 RAVs was lower (4/817 or 0.5%). No difference in
fitness between the double rpoB-Rv0678 mutant and the single rpoB mutant was found upon
exposure to rifampicin.
Conclusions: RAVs in Rv0678 mutations occur more frequently in MDR-TB patients than
previously anticipated, and are not associated with prior use of BDQ or CFZ. Rifampicin is unlikely
the selective pressure leading to this phenomenon. In the majority of cases, Rv0678 RAVs do not
lead to BDQ MICs above the breakpoint. Given this unpredictable impact on MICs, phenotypic drugsusceptibility methods are preferred over molecular methods to assess BDQ susceptibility.
376
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
347 Drug design-new developments
23/09/2016
Derivatives of Oxyphosphorus Acids with Antimycobacterial Activity
J. Vinsova1, M. Krátký1, J. Stolaříková2
1
Department of Inorganic and Organic Chemistry, Faculty of Pharmacy, Charles University, Hradec
Kralove 2Regional Institute of Public Health in Ostrava, Laboratory for Mycobacterial Diagnostics
and Tuberculosis, Ostrava, Czech Republic
The rise of multidrug-resistant tuberculosis (MDR-TB) and extensively drug-resistant (XDR-TB)
cases and co-infection with HIV is alarming. These facts stimulate the development of new effective
anti-tuberculosis drugs acting by new or different mechanism of action, attacking on the latent
(dormant) forms which may become active and when are mutated, cause incurable pandemic.
Based on our previous results of salicylanilide diethyl (thio)phosphates showing an increased
antimycobacterial activity against both tuberculous and non-tuberculous mycobacteria, including
drug-resistant strains, a decreased cytotoxicity and thus improved selectivity in comparison with the
parent salicylanilides1,2, we have synthesized and evaluated other new substituted 2(phenylcarbamoyl)phenyl phosphenite derivatives.
Parent compounds, i.e., the most active salicylanilides from previous studies1,2,3, prepared by
microwave assistance from substituted salicylic acids and anilines in the presence of PCl33, were
esterified via chlorides of appropriate oxyphosphorus acids in the presence of triethylamine2. Using
dichlorides
of
phosphorus-based
acids,
substituted
3-phenyl-3-hydrobenzo[e]
[1,3,2]oxazaphosphinin-4-one 2-oxides were synthesized. Some derivatives were obtained by a
reaction of substituted salicylanilides with phosphite in the presence of tertiary base, 4(dimethylamino)pyridine
and
tetrachloromethane
(Atherton-Todd
reaction).
All the prepared esters and heterocycles were investigated for their in vitro activity in Šula’s
semisynthetic medium to determine their minimum inhibitory concentrations (MICs). The
antimycobacterial activity was evaluated against Mycobacterium tuberculosis CNCTC (Czech
National Collection of Type Cultures) 331/88 (H37Rv), CNCTC nontuberculous mycobacteria:
Mycobacterium avium 330/88 and Mycobacterium kansasii 235/80 and a clinically isolated strain of
M. kansasii 6509/96. The most active derivatives having MIC ≤ 1 µmol.L-1 underwent additional
evaluation against MDR-TB strains clinically isolated from patients. Both synthesis and biological
evaluation results have been patented.4 Their mechanism of action is currently under investigation.
References:
1. Vinšová J. et al.: Molecules 19, 7152 (2014).
2. Vinšová J. et al.: Bioorg. Med. Chem. 22, 728 (2014).
3. Krátký M. et al.: Eur. J. Med. Chem. 45, 6106 (2010).
4. PCT/CZ2015/000129, Czech patent PV-2014-915.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
377
348 Drug design-new developments
23/09/2016
Copper-related innate immunity inspired drug discovery targeting Mycobacterium
tuberculosis
S. Shah, A. Dalecki, C. Crawford, F. Wolschendorf
Medicine, University of Alabama at Birmingham, Birmingham, Al, United States
Mycobacterium tuberculosis (Mtb) is becoming a drug-resistant superbug. In an effort to identify
much needed new classes of compounds that act by novel mechanisms, we took inspiration from
the innate immune system’s use of copper ions to defend against bacterial intrusion. A pilot screen
uniquely tailored to identify compounds with copper-related anti-mycobacterial activities yielded 89
hit molecules with copper-dependent activity. For comparison, the same library gave only 16
molecules that inhibited growth of Mtb in a copper independent fashion, indicating that compounds
with copper-related activities comprise a major, yet unrecognized, reservoir of potentially exploitable
antibacterial activities. Chemoinformatic analysis revealed 7 distinguished hit clusters with the
cluster of oxines comprising one of the largest. The minimal inhibitory concentration (MIC) of oxine
itself was 0.16 μM against replicating and non-replicating Mtb. The presence of extracellular copper
ions was absolutely essential for its activity on whole cells, suggesting a mode of action unrelated to
its ability to extract catalytic metal ions from crucial metalloenzymes. Instead, oxine’s activity
appears to be related to an increase in cell-associated copper, consistent with action as copper
ionophore. Furthermore, we found that only copper ions confer antibacterial properties to the
resulting oxine/metal-complex. The addition of Zn, Fe, or Mn, which also coordinate with oxine,
neither enhanced nor ablated copper-specific effects, which is remarkable considering that the
respective metal complexes are of nearly identical structure and geometry. Finally, in a macrophage
infection model, oxine treatment significantly reduced intracellular bacterial burden by more than
50% within 48h. Given the stark copper-dependent nature of its activity, this has provided evidence
for the first time that copper ions within Mtb-infected host cells directly enable the antibacterial
properties of a copper-dependent inhibitor of Mtb. In summation, our findings highlight metal binding
abilities of drugs as a potential focus for future medicinal chemistry and demonstrate the potential of
innate immunity-inspired screening platforms to uncover molecules with novel modes of action
against Mtb.
378
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
349 Drug design-new developments
23/09/2016
Evaluation of Genotype MTBDRsl version 2 for rapid detection of second-line antituberculosis drug resistance in Ghana identifies the first pre-XDR TB case
D. Yeboah-Manu2, S. Osei-Wusu2-3, A. Asante-Poku2, I.D. Otchere2, S. Omari1, A. Forson1
1
Department of Chest Diseases, Korle-Bu Teaching Hospital 2Bacteriology, Noguchi Memorial
Institute for Medical Research, University of Ghana 3West Africa Centre for Cell Biology of
Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
Background: Early profiling of clinical MTBC strains for drug resistance is essential for proper
control of TB. Whereas the genotype MTBDRplus for first line drugs is globally accepted for its
specificity and sensitivity, the MTBDRsl for second line v1 received conflicting responses leading to
the development of v2. We evaluated the genotype MTBDRsl v2 for rapid screening of MTBC
strains against second-line drugs in Ghana.
Method: We screened 117 (76 MDR, 14 RIF mono-resistant and 27 INH mono-resistant) resistant
TB isolates against moxifloxacin (MOX), streptomycin (STR) and amikacin (AMK) using the
Epsilometer test (Etest). In addition, all the 76 MDR, 14 RIFr and 3 INHr were screened with the
MTBDRsl. Genes (gyrA, gyrB, eis, rrs, tap, whiB7 and tlyA) of strains resistant to MOX or AMK were
PCR amplified and sequenced for mutation analysis.
Results: The isolates were obtained from 81.2% males and 28.8% females with an average age of
37.1 years. New cases, relapsed and treatment failures were respectively 69(59.0%), 47(40.2%)
and 1(0.8%). Isolates phenotypically resistant to STR, AMK and MOX were 38 (32.5%), 13 (11.1%)
and 1 (0.9%) respectively. Among the isolates, only 1 MDR isolate was diagnosed resistant to both
MOX (MIC=3µg/mL) and AMK (MIC =16µg/mL) by both the phenotypic assay and MTBDRsl LPA.
Gene sequencing analysis led to the identification of gyrA (G87C) and rrs (A514C and A1401G) in
this isolate. Whereas none of the remaining isolates were resistant to MOX (MIC<0.125µg/mL), 9/76
(11.8%) MDR, 1/14 (7.1%) RIFr and 3/27 (11.1%) INHr were resistant to AMK with MICs ranging
from 1 to 8µg/mL. However, none of these were identified as resistant to AMK by the MTBDRsl
LPA. Except one MDR isolate harbouring a tlyA mutation (N236K), no other mutation was detected.
The agreement between the phenotypic and the LPA was 87.1% and 100% whereas the
sensitivity/specificity was 7.7%/100% and 100%/100% for AMK and MOX respectively.
Conclusion: We report the first instance of a pre-XDR-TB case in Ghana. Our findings support the
use of the MTBDRsl v2 in Ghana. However, its inability to detect low level resistance to AMK is a
major concern.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
379
350 From animal models to man
23/09/2016
Discovery of potent and specific M. tuberculosis leucyl-tRNA synthetase inhibitors
(GSK070)
X. Li3, V. Hernandez3, F.L. Rock3, Y.S.L. Mak3, M. Mohan3, W. Mao3, Y. Zhou3, E.E. Easom3,
J.J. Plattner3, W. Zou1, C. Alemparte2, E. Perez-Herran2, I. Giordano2, A. Mendoza2, D. Barros2,
M.R.K. Alley3
1
BioDuro, Beijing, China 2Diseases of the Developing World, GlaxoSmithKline, Tres Cantos,
Spain 3Anacor Pharmaceuticals, Palo Alto, United States
Background: Leucyl-tRNA synthetase (LeuRS) is an essential enzyme for protein synthesis.
LeuRS has two catalytic sites, an aminoacylation site, which charges tRNALeu with leucine and an
editing or proof-reading site that hydrolyses incorrectly charged tRNALeu. Application of the
oxaborole tRNA trapping (OBORT) mechanism (Rock & Mao et al. Science, 2007, 316: 1759) to M.
tuberculosis (Mtb) LeuRS lead to the discovery of GSK070, which is in development for tuberculosis
treatment.
Material/methods: Enzyme and cell-based structure-activity relationships (SAR) were used to
optimize potency. The LeuRS IC50 values were measured using the aminoacylation assay. The
nosocomial pathogen MIC values were determined by CLSI M7-A10 and the M. tuberculosis H37Rv
MIC values were determined using resazurin. Inhibition of human cell protein synthesis was
determined in HepG2 cells.
Results: We designed and synthesized a series of LeuRS inhibitors by introducing various
substituents onto the aromatic ring of the 3-aminomethyl benzoxaborole scaffold. A number of
compounds were identified, which showed good antitubercular activity with high selectivity over
human cytoplasmic protein synthesis inhibition. Further evaluation of these Mtb LeuRS inhibitors led
to the discovery of GSK070, which exhibits potent inhibition of Mtb LeuRS (IC50 = 0.216 µM) and
antitubercular activity (Mtb H37Rv MIC = 0.08 µM). Additionally, it is highly specific for the Mtb
LeuRS enzyme, as the IC50 for human cytoplasmic LeuRS is 140 µM.
Conclusions: GSK070, selected as preclinical candidate, is a new antitubercular agent that targets
Mtb LeuRS, a novel protein synthesis target for tuberculosis.
380
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
351 From animal models to man
23/09/2016
Early safety profile of gsk070 a new antitubercular
M. Cacho1, E. Jiménez-Navarro1, A. Mendoza1, S. Gresham2, S. Turner2, R. Lincoln2, P. Bushdid4,
L. Xi3, M.D. Allen3, D. Barros-Aguirre1
1
TB DPU, Tres Cantos, Spain 2R&D PTS Safety Assessment, GlaxoSmithKline, Ware, United
Kingdom 3Anacor Pharmaceuticals, Inc, Palo Alto 4R&D PTS Safety Assessment Reproductive
toxicology, GlaxoSmithKline, Upper Merion, United States
Background: Oxaborols series are inhibitors of Leucyl t-RNA synthetase and selective antitubercular agents with a remarkable activity against a selection of DS-TB, MDR-TB and XDR-TB
clinical isolates. This series exhibits excellent physicochemical properties and has demonstrated in
vivo antitubercuular activity.
Material/methods: GSK070 has been evaluated to discharge the following toxicology risks:
general, genetic, reproductive, cardiovascular, phototoxicicty, and safety pharmacology. It has been
tested through eXP cross functional panel of human receptors, enzymes and transporters. Ames
test and MLA in the presence and absence of S9 fraction was performed to evaluate genetic
toxicology. Inhibition of cardiac ion channels, hERG, Cav1.2 and Nav1.2 were tested by Patch
clamp platforms and ex vivo rabbit ventricular wedge was assessed to evaluate cardiovascular
liability and a 7 day repeat dose toxicity study in male rats.
Results: GSK070 inhibited hERG and CaV1.2 receptors with IC50 of 10uM for both channels. In a
subsequent rabbit ventricular wedge study there were no changes in any of the parameters
measured, and from the cardiovascular rat study there is a >60-fold cover to the estimated free
Cmax at a clinical dose of 33mg/day from the 10mg/kg/day dose at which no effects were observed.
GSK070 was tested in a bacterial mutation screening assay (Ames test) +/- S9 fraction and was not
mutagenic in any of the strains investigated and it was also negative in the MLA in the presence and
absence of S9. GSK070 was tested in the rat whole embryo culture (WEC) in vitro test system and
there is a minimum of an 18-fold margin from the NOEL at 10uM in the WEC and a 151-fold margin
at 100mg/kg/day in a rat 7 day study.
Conclusions: Based on an integrated assessment of all the safety findings (general tox, safety
pharmacology, genotoxicity, reproductive toxicity) and taking into consideration that there are
biomarkers and/or monitoring available for the toxicities observed in vivo. The preliminary
toxicological profile (in vitro and in vivo) indicates an acceptable therapeutic window and the riskbenefit profile is such that there is no data that would preclude the progresion of this molecule.
All animal studies were ethically reviewed and carried out in accordance with European Directive
86/609/EEC and the GSK Policy on the Care, Welfare and Treatment of Animals
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
381
352 From animal models to man
23/09/2016
In vitro grown human lung organoids as a model of tuberculosis
N. Iakobachvili1, C. Lopez-Iglesias1, N. Sachs2, H. Clevers2, P. Peters1
1
M4I Nanoscopy Division, Maastricht University, Maastricht 2Hubrecht Institute, Utrecht, The
Netherlands
Tuberculosis remains a major global health threat; as a result, a huge number and variety of
animals are being used for fundamental research into mycobacterial pathogenesis, virulence
mechanisms and the development of novel treatment regimens and vaccines. Animal suffering is
thought to be extensive and the validity of these models remains highly questionable as the
causative agent of the disease, Mycobacterium tuberculosis, is an obligate human pathogen.
Models of tuberculosis based on human cells currently do not extend beyond infection of
immortalized cell lines, isolated and differentiated monocytes from human blood, and simple cocultures of the above.
We are developing an in vitro model of tuberculosis using stem cells from healthy human lung tissue
that have been cultured to expand, differentiate and naturally self-organize into organoids closely
resembling human bronchi. The lab-grown “mini lungs” display important features of native lung
epithelia and are composed of naturally differentiated cells of respiratory lineage (including ciliated,
basal, secretory and epithelial cells) and their products. Together, the microenvironment of the lung
is mimicked although some immune components are missing which we aim to separately introduce
into organoid cultures. Mycobacteria are introduced to cultures by intraluminal injection and are
monitored by 2-photon excitation microscopy and confocal microscopy when using live fluorescent
stains; and by electron microscopy following high pressure freezing and freeze substitution.
Whilst still in its preliminary stage, we believe that the use of a more physiologically relevant model
such as the mini-lung organoids can produce more predictive data for human tuberculosis than
animal models. The lung organoids can be further developed as a novel platform for screening of
anti-mycobacterial compounds or adapted to model other respiratory pathogens.
382
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
353 From animal models to man
23/09/2016
Tuberculosis Preclinical and Clinical Resources at the National Institute of Allergy and
Infectious Diseases.
A. Kraigsley, J.P. Boyce, K. Lacourcière, B. Laughon, F.J. Leyva, M. Makhene, A. Mcbride,
C.F. Sizemore
NIH/NIAID/DMID/RDB, United States
The Division of Microbiology and Infectious Diseases (DMID), part of the National Institute of Allergy
and Infectious Disease (NIAID), part of the U.S. National Institutes of Health (NIH) is committed to
finding new ways to better understand, diagnose, treat, and ultimately prevent tuberculosis (TB).
The continued high burden of TB in many countries and the emergence of MDR TB and XDR TB
has intensified the need for new interventions. In addition to supporting a comprehensive research
and product development grants portfolio, NIAID/DMID also offers preclinical and clinical services to
help researchers identify and develop products, technologies and targets relevant to treatment of
tuberculosis. This presentation focuses on a summary of resources for preclinical and clinical
biomedical research in TB that are issued through NIAID’s Biodefense and Emerging Infectious
Diseases Programs. These services include in vitro evaluation of new chemical entities, in vivo
animal model testing as well as early stage evaluation of new diagnostic candidates, vaccines,
research reagents, genome sequencing and bioinformatics support. Many of these services are
available at very low cost to U.S. and international investigators in academia, not-for-profit
organizations, industry and governments through an application process. Requests are evaluated
based on standard criteria including preliminary data and development plans. Institutional
agreements will be executed to ensure confidentiality and protect intellectual property rights.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
383
354 From animal models to man
23/09/2016
Predicting the Human Dose for a novel LeuRS inhibitor, GSK070, for the Treatment of
Tuberculosis
M. Remuinan, F. Ortega, S. Crouch, A. Mendoza, R. Gomez, L. Alameda, S. Ferrer, D. Barros,
X. Li1, M.R.K. Alley2
1
GSK, Tres Cantos, Spain 2Anacor Pharmaceuticals, Palo Alto, United States
Background: GSK070 is a selective inhibitor of mycobacterial leucyl t-RNA synthetase (LeuRS)
with a novel MoA for the treatment of tuberculosis (TB). After achieving positive results in the murine
chronic assay for TB, preliminary human dose projections were performed to aid in clinical
progression.
Material/methods: Following in-vivo intravenous dosing in the preclincal species (mouse, rat and
dog), the human pharmacokientic parameters of clearance and volume of distribution were derived
using in-vitro/in-vivo extrapolation (IVIVE), allometry and simple PBPK modeling (CloePK Software,
Cyprotex). A target efficacious exposure in human was determined using the dose (1.3mg/kg) at the
maximum effect (EDmax) level observed in the therapeutic efficacy murine chronic assay. Human
dose estimations were calculated using a target efficacious exposure of 3481ng*hr/ml which
corresponds to the efficacious AUC0-24h at steady state measured in whole blood.
Results: With the PK parameters from allometry and PBPK multiple scenarios were designed for
the human dose projection in order to achieve the efficacious target exposure (whole blood and
unbound). Predicted Vdss was moderate in all cases, while clearance was variable (low to
moderate) depending on the modelling strategy used. All methodologies projected a low human
dose (<200mg) to achieve the therapeutic target in whole blood (3481ng*h/mL).
Conclusions: GSK070, a novel preclinical candidate for the treatment of tuberculosis, provides
favorable predicted human pharmacokinetic parameters with a low projected human dose.
All animal studies were ethically reviewed and carried out in accordance with European Directive
2010/63/EU and the GSK Policy on the Care, Welfare and Treatment of Animals.
384
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
355 From animal models to man
23/09/2016
In vivo antitubercular activity of GSK070, a Mycobacterium tuberculosis leucyl-tRNA
synthetase inhibitor
J. Rullas1, L. Guijarro-Lopez1, A. Mendoza1, X. Li2, M.R.K. Alley2, D. Barros1
1
Biology TB, GlaxoSmithKline, Tres Cantos, Spain 2Anacor Pharmaceuticals Inc, Palo Alto, United
States
Background: Protein synthesis inhibitors play an important role in the treatment of tuberculosis.
Parenteral aminoglycosides form a key part of the treatment of multidrug-resistant (MDR) TB and
currently the oxazolidinones are the only class of protein synthesis inhibitors that are orally
bioavailable and are effective against TB. The rise of resistance including extensively (XR) and
totally resistant (TR) TB demands the need for new oral frontline drugs. Here, we show the excellent
in vivo profile of GSK070, a new Mycobacterium tuberculosis leucyl-tRNA synthetase inhibitor
(LeuRS).
Material/methods: The experimental designs for the acute and combination studies have been
previously described by Rullas et al. (AAC 2010; 54:2262) for the acute assay and Nuermberger et
al. (AAC 2008; 52:1522) for the model used for the combinations, respectively. For the chronic
assay, mice were infected intratracheally with 100 CFU, the treatment was administered once a day
7/7 for 8 weeks starting on week 6 after infection. Mice were sacrificed 24 hours after the last
administrations, and lung CFU were counted by plating in OADC-7H11 media supplemented with
0.4% activated charcoal. All animal studies were ethically reviewed and carried out in accordance
with European Directive 86/609/CEE and the GSK Policy on the Care, Welfare and Treatment of
Animals.
Results: For the acute and chronic assays, GSK070 administered orally once a day to infected
C57BL/6 mice induced the killing in vivo of M. tuberculosis H37Rv. The maximum reduction of lung
M. tuberculosis burden in the acute model (about 4 log10 units compared to untreated mice) was
obtained upon daily oral administration of a dose of 1.1 mg/kg for 8 days. The maximum killing in
the chronic model (about 2 log10 units) was obtained upon daily oral administration of a dose of 1.3
mg/kg for two months. In both models there was quantifiable remaining bacterial burden after the
end of treatment, although when the treatment was prolonged, we could observe a further decline in
the lung CFU counts in the chronic model. When administered in combination, the combo Pa824+Bedaquiline+Linezolid+GSK070 was more active than the standard of care (Rif+Iso+Pyr+Eta)
or Pa-824+Bedaquiline+Linezolid.
Conclusions: GSK070 has excellent in vivo antitubercular activity with demonstrated efficacy in
both the standard acute and chronic murine assays, and when it was administered combination with
new antitubercular drugs is more active than standard of care.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
385
356 From animal models to man
23/09/2016
Exploring the role of the RskA-SigK regulon in the virulence of Mycobacterium bovis
A. Smyth4, F. Veyrier1, M. Behr2, E. Lavelle3, S. Gordon4
1
Institut Armand Frappier, Laval 2Department of Microbiology and Immunology, McGill University,
Montreal, Canada 3Adjuvant Research Group, School of Biochemistry and Immunology, Trinity
College Dublin 4School of Veterinary Medicine, University College Dublin, Dublin, Ireland
Mycobacterium bovis and Mycobacterium tuberculosis (M. tb) share 99.95% nucleotide sequence
identity, but while M. tb is almost exclusively a human pathogen M. bovis affects a wide range of
domesticated and wild animal species and can cause zoonotic infections in humans. The precise
molecular mechanisms that allow M. bovis to infect, cause disease and transmit across a wide
variety of animal hosts has yet to be explained.
A potential virulence mechanism that may play a role is the SigK regulon that is differentially
regulated across the MTBC. The SigK regulon is under the control of the regulator RskA; two SNPs
in rskA alter the gene in M. bovis, and as a result the SigK regulon is constitutively expressed in M.
bovis. In comparison, M. tb shows minimal expression of genes in the SigK regulon under in vitro
growth, but induces expression of the regulon during intracellular infection. No specific role in
infection has been discovered for any of the SigK regulated proteins.
A previous experimental infection of bovine alveolar macrophages with M. bovis 2122/97 and M. tb
H37Rv has indicated differential cellular responses to the two species, most significantly an
increase in the Type I interferon response to M. bovis. We aim to investigate what role the SigK
regulon may have in causing this differential cellular response. The effects of constitutive SigK
activation on mycobacterial interaction with innate immune cells will be analysed using a
recombinant strain of M. tb containing rskA and sigK alleles of M. bovis and hence showing
upregulation of the SigK regulon as observed in M. bovis. The cellular response to the recombinant
M. tb strain is being explored using primary bovine macrophages and the murine cell line RAW
264.7, and analysed via transcriptomics and complementary targeted approaches.
386
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
357 From animal models to man
23/09/2016
Mycobacterial imaging with fluorescent benzothiazinone derivatives for in vitro and in vivo
applications
R. Sommer, S. Cole
Global Health Institute, EPFL, Lausanne, Switzerland
Tuberculosis (TB), a bacterial disease caused by Mycobacterium tuberculosis, remains a major
global health problem and new antibacterial agents are required to fight the increasing number of
drug-resistant strains. Benzothiazinones (BTZs), a new class of anti-TB molecules, are highly potent
covalent inhibitors of DprE1, an essential enzyme involved in the synthesis of the mycobacterial cell
wall. The drug candidate PBTZ169 is currently in phase I trials. We converted BTZ molecules into
fluorescent probes by coupling the benzothiazinone core with different fluorophores, such as
carboxytetramethylrhodamine (TAMRA) or the near-infrared fluorophore Dy750. These modified
drugs still retained modest antimicrobial activity despite the addition of a bulky fluorescent moiety
and were used to show by microscopic examination that DprE1 is situated at the poles of the bacilli
in the periplasmic space of several mycobacterial species (M. marinum, M. smegmatis and M.
tuberculosis). These probes are useful to track the target of BTZs and to investigate the
mechanisms of drug action and the organization of the cell wall synthesis machinery. Given the high
specificity of BTZs for mycobacteria, we also seek to use near-infrared fluorescent BTZ derivatives
to evaluate the bacterial load in a mouse model of chronic TB by non-invasive in vivo optical
imaging, thus allowing longitudinal studies, reduction of the number of animals and gain of time
compared to the standard viability assays (CFU count) currently used to evaluate the efficacy of
new drug regimens or vaccine candidates. M. tuberculosis reporter strains expressing near-infrared
fluorescent proteins and/or luciferase will be used in parallel to optimize the sensitivity, reduce the
limit of detection and increase the precision of the localization of the optical signal.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
387
358 From animal models to man
23/09/2016
The mycobacterial ESX-1 secretion system substrate ESAT-6 is not sufficient for
membranolytic activity
W. Conrad, M. Osman, J. Shanahan, F. Chu, K. Takaki, J. Cameron, R. Brosch, L. Ramakrishnan
University of Cambridge, Cambridge, United Kingdom
The mycobacterial ESX-1 secretion system is required for virulence. The ESX-1 substrate ESAT-6
is an immunodominant antigen and has been broadly implicated in ESX-1 virulence attributes. ESX1 deficient mycobacteria are defective for macrophage phagosome permeabilization, activation of
cytosolic DNA sensing pathways, intramacrophage growth and macrophage death. These
phenotypes are thought to be mediated via host membrane disruption which has been attributed to
ESAT-6 because multiple studies have shown recombinant ESAT-6 to lyse eukaryotic membranes.
We show that the membranolytic activity previously attributed to recombinant ESAT-6 is due to
residual nonionic detergent ASB-14 detergent in the preparations. Our findings suggest
reconsideration of all prior findings attributing ESX-1 lytic function to ESAT-6. Rather they
demonstrate that ESX-1 mediated membranolytic activity is either mediated by ESAT-6 acting in
conjunction with other ESX-1 substrates. Alternatively, it may simply be a marker of ESX-1
secretion, and cytolytic activity may be mediated by different ESX-1 substrates. These findings also
warrant reconsideration of the role ESAT-6 plays in virulence.
* Equal contributors
388
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
359 From animal models to man
23/09/2016
The cytochrome bc1-aa3: an absurd target for tuberculosis?
Kevin Pethe
Nanyang Technological University, 59 Nanyang Drive, 636921 Singapore
The clinical approval of bedaquiline (Sirturo) validated oxidative phosphorylation as a novel drug
target space for tuberculosis. More recently, the discovery of small-molecules targeting the
cytochrome bc1-aa3 (a terminal oxidase of the electron transport chain) has triggered interest on
the respiratory cytochrome c pathway. A series of imidazopyridine amide compounds interfering
with the function of the cytochrome bc1 were identified independently by several teams. Q203, the
most advanced IPA derivative, is in clinical development phase I under a US FDA investigational
new drug application. The metabolic redundancy of the electron transport chain, especially at the
level of the terminal electron acceptor, raises some questions about the chemical vulnerability of the
respiratory cytochrome c pathway. Our data demonstrate that the potency of Q203 is greatly
influenced by many external factors, including nutrient availability and growth conditions. In addition,
functional redundancy with alternate terminal electron acceptors modulates the essentiality of the
cytochrome bc1-aa3. The possible consequences for clinical development of Q203 and for
treatment of other mycobacteria infections will be discussed.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
389
360 From animal models to man
23/09/2016
Methionine Aminopeptidase B (mapB), but not mapA, is essential for M. Tuberculosis viability
Daniel. Barkan
Koret School of Veterinary Medicine, Hebrew University, Rehovot, Israel
Introduction: In all living cells, the initiating methionine in most newly synthesized proteins is
removed by a Methionine Aminopeptidase enzyme (MAP). For reasons not completely clear, the
genes coding for these enzymes are essential in all living cells. In cells that have two copies, there
is either one essential and one redundant, or they are synthetically lethal. Mycobacterium
tuberculosis has two genes – mapA and mapB. Their respective essentiality is currently unknown. A
study using chemical inhibitors concluded mapA was essential, and mapB was not. In contrast,
analysis of a transposon library concluded that mapB was the essential one. We decided to
individually delete each of these genes to resolve which one is essential, and what is the phenotype
of deletion of the non-essential one.
Methods: For each gene, we pre-complemented M. bovis bacteria with an extra copy of the gene at
the attB site, performed a deletion of the native gene with a transducing phage, and then attempted
to remove the complementing genes using another integrating cassette. In one case, we also use
conditional expression.
Results: We successfully deleted each gene, when the bacteria was pre-complemented with that
same gene. When we attempted to remove the complementing gene of mapA, we could easily do it,
getting a full mapA deletion mutant. This mutant had a very mild growth retardation phenotype invitro. In contrast, removal of the complementing mapB from a ΔmapB mutant was not possible, thus
proving mapB was essential for the survival of the bacteria in-vitro.
Conclusions: Contrary to result previously obtained by chemical inhibition and by ASRNA, mapB,
and not mapA, is the essential MAP gene in M. bovis (and Mtb). Future attempts at an inhibitor
development should therefore focus on it, rather than mapA. The reason why this pathway is
essential remains unclear, for the mare function of methionine removal appears, to us, not sufficient
for rendering the genes essential.
390
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
AUTHORS
&
CO-AUTHORS INDEX
A
Abdallah A.
Abdelaal H.
Abdin M.Z.
Abdulwhhab M.
Abel L.
Abid M.
Abrahams K.
Accard G.
Adam D.
Adamson J.
Addicott K.
Aebersold R.
Afanasyev P.
Afzali Behbahani M.
Agarwal N.
Agarwal S.
Aguilar Pérez C.
Aguilo N.
Ahmed W.
Ahuja N.
Ainsa J.A.
Ait Benhassou H.
Ajuh P.
Akabayov B.
Akopian T.
Al Saati T.
Alameda L.
Alavi S.M.
Alcaîs A.
Alderwick L.J.
Aldrich C.
Alemparte C.
Alisjahbana B.
Allen M.D.
Alley M.R.K.
Alonso M.
Al-Saati T.
Alvarez M.
Alvarez S.
Alzari P.
Amallraja A.
Amorim Franco T.
Amzazi S.
Andersen P.L.
Andreas N.
Andreu N.
Andrew P.
Andries K.
Angala S.
Angulo N.
Anil Kumar V.
Annisa J.
Antonio M.
Anton-Leberre V.
Anwar S.
Appelberg R.
Apriani L.
Author/Co-Author
13
90P
162P
280P
258P
95P, 313P
109P, 133P
234P
245P
131P
248P
67P
172P
65P
162P
152P, 249P
309P
30
138P
242P
309P, 330P
95P, 313P
136P
203P
126P
298P
354P
65P
258P
122P, 204P, 240P
336P
133P, 350P
89P, 278P, 308P
351P
55, 350P, 354P, 355P
206P
46
215P
30
215P
91P
205P
313P
26
177P
117P, 238P
116P
346P
10
266P
148P
308P
141P
46
239P
270P
278P
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
Apt A.
Aragaw W.
Arnold F.
Arnvig K.
Arora D.
Arora G.
Arora K.
Arora N.
Arries J.
Arya R.
Asante-Poku A.
Åsberg S.
Ascher D.
Astolfi Rosado L.
Ates L.
Aubry A.
Aung T.
Avdienko V.
Av-Gay Y.
Ayorinde A.
Aziz D.
263P
310P
167P
96P
150P, 281P
311P
337P, 340P
164P
97P
210P
66P, 86P, 142P, 349P
292P
19
206P
13, 163P
72P
232P
263P
98P
267P
312P, 334P
B
Author/Co-Author
Bach H.
98P
Bachy V.S.
181P
Bacon J.
240P
Bacqué E.
129P, 147P, 314P
Baek S.
144P
Baer C.
107P
Bah A.
235P
Bahrmand A.
183P
Baidoo A.
231P
Bailo Vergara R.
99P
Bajpai U.
211P
Balakrishna A.
219P
Balboa L.
298P, 306P
Balhana R.
100P, 102P, 134P
Baliga N.
99P
Ballell L.
109P, 233P, 339P
Ballell-Pages L.
54
Ballinger E.
147P
Banaei-Esfahani A.
67P
Bancroft G.J.
304P
Bandyopadhyay S.
170P
Baneres Roquet F.
209P
Banerjee N.
114P, 118P
Bansal R.
148P
Baranyai Z.
326P
Barbouche R.
283P
Barer M.
280P
Barisch C.
282P
Barros D.
109P, 339P, 350P, 354P, 355P
Barros-Aguirre D.
55, 351P
Barry C.
31, 60, 337P, 338P, 340P
Barton A.
20
Barzon L.
291P
Bassano L.
132P
393
Basu P.S.
100P, 102P
Basu Roy R.
177P
Batra S.
151P
Battaglia S.
68P
Batthyány C.
215P
Bauer A.
129P
Baughn A.
319P
Baulard A.
53, 318P
Bayliss T.
340P
Beckert P.
84P
Beckham K.
15, 165P, 207P
Behar S.
47
Behr M.
41, 78P, 79P, 251P, 356P
Bellinzoni M.
54, 215P
Belon C.
234P
Benabdessalem C.
283P
Benard A.
46, 125P, 298P
Bendt A.
269P
Benedetti Y.
130P
Bentaleb E.
313P
Berger C.
172P, 173P
Berná L.
4
Bernard C.
72P
Berney M.
101P
Berney-Meyer L.
101P
Bernut A.
233P, 235P, 241P
Bertozzi C.
218P, 226P
Besra G.
109P, 122P, 133P, 204P, 316P
Best K.
196P
Beste D.
100P, 102P
Bestebroer J.
163P
Beuerman R.
232P, 335P
Bhagyaraj E.
242P
Bhaskar S.
198P
Bhatnagar R.
114P, 118P
Bhatt A.
99P, 123P, 204P
Bhatt S.
294P
Bhuwan M.
164P
Biot C.
233P, 327P
Birhanu A.
103P
Bishwal S.
210P
Biswas P.
104P
Biswas R.
186P
Bitter W. 13, 15, 163P, 165P, 168P, 286P, 315P
Biukovic G.
208P, 329P
Blaise M.
209P, 229P, 233P, 237P, 252P
Blanc I.
129P, 130P, 147P, 314P
Blanc L.
221P
Blanchard J.
205P, 212P, 230P
Blanco-Ruano D.
133P
Blanc-Potard A.B.
234P
Blazquez J.
69P
Blomgran R.
266P
Blundell T.
19, 337P
Boisson-Dupuis S.
62, 258P
Boom H.
71P
Boot M.
315P
Borel S.
296P
394
Borgers K.
Borges M.
Boritsch E.
Borrás R.
Borrell S.
Borroni E.
Boshoff H.
Bosmans R.
Bösze S.
Botella H.
Bottai D.
Boucher N.
Bouchier C.
Boudinot P.
Boulenc X.
Boulle M.
Boutte C.
Boyce J.P.
Boy-Röttger S.
Braian C.
Brandenburg J.
Bräuchle C.
Braverman J.
Brennan P.J.
Bresciani N.
Brighenti S.
Briot C.
Brites D.
Brodin P.
Bronda K.
Brosch R.
Broset E.
Brossier F.
Brown K.
Brown M.
Browne J.A.
Bryant J.
Buchanan K.
Buechler J.
Bull T.
Bunduc C.
Bunjun R.
Burgers W.
Burke M.
Burmeister M.
Burns-Huang K.
Bushdid P.
Busso S.L.
Bustamante J.
Butcher P.
Buttari N.
C
Cabibbe A.M.
Caccamo N.
Cacho M.
105P, 180P
270P
106P
182P
67P, 70P
68P
60, 147P, 337P, 338P, 340P
172P, 173P
326P
9, 17, 154P
304P
14
106P, 233P
46
129P, 130P, 314P
299P
107P
353P
213P
259P
225P, 243P, 301P
225P
44
321P
108P
300P
130P, 314P
71P, 81P
1, 221P, 305P, 318P
288P
3, 29, 106P, 175P, 215P,
233P, 235P, 304P, 305P
30, 178P
72P
39, 288P
239P
247P, 264P
39
340P
19
137P
15, 165P
244P, 271P
244P, 271P
51
245P
147P
351P
66P, 86P
258P
23
54
Author/Co-Author
68P
22
351P
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
Cacho-Izquierdo M.
Cadieux N.
Callewaert N.
Camassa S.
Cambier C.
Cameron S.
Cantalloube C.
Carata E.
Cardoso F.
Cardoso M.
Carlsson F.
Carrat C.
Carroll P.
Carvalho T.
Casanova J.L.
Cascioferro A.
Castañeda-García A.
Castro R.
Castro-Ramos M.
Catanzaro A.
Catanzaro D.
Catinas O.
Cebrián R.
Cha H.
Cha S.B.
Chae H.
Chaidir L.
Chain B.
Chan C.
Chandra V.
Chase M.
Chaudhari T.K.
Chaudhuri T.K.
Chavez A.
Chawla Y.
Chee C.
Chen C.
Chen D.
Chen S.L.
Chernyaeva E.
Chiarelli L.R.
Chibale K.
Chindelevitch L.
Chiner-Oms Á.
Chiodarelli G.
Chng S.
Cho S.N.
Choi H.
Chor J.
Christensen D.
Chung J.C.S.
Church G.
Cia F.
Cianci M.
Ciccarelli L.
Cichonska A.
Cirillo D.M.
Clark R.
316P
202P
105P, 180P
253P
284P
205P
129P, 314P
253P
276P
270P
297P
265P
194P
276P
258P
215P
69P
70P
4
91P
91P
299P
309P
184P
185P, 199P, 200P, 261P
179P
89P, 308P
196P
93P, 157P
242P
7
164P
111P, 198P
7
281P
269P
60
172P, 173P
157P
73P, 74P
54
345P
317P
75P
316P
32, 176P
200P, 261P
246P, 261P
232P
194P
157P
7
304P
113P
15, 165P
23
68P, 108P
14
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
Clark T.G.
238P
Clevers H.
352P
Clish, Clary C.
308P
Cobat A.
258P
Coeck N.
346P
Cohen-Gonsaud M.
285P
Coker E.
267P
Cole S.
19, 56, 64, 85P, 138P, 174P,
213P, 331P, 357P
Coler R.N.
200P
Collière L.
129P
Collins B.
67P
Collinson L.
296P
Comas I.
75P, 182P, 317P
Commandeur S.
286P, 315P
Conlon K.
264P
Cook G.
155P
Cooper A.M.
45
Correia C.
247P
Cortes T.
51, 117P
Cortesía C.
175P
Coscolla M.
67P, 141P
Costa-Gouveia J.
318P
Cougoule C.
298P, 306P
Couturier C.
147P
Cox J.
48, 109P
Cox L.R.
316P
Crawford C.
348P
Crommelynck S.
304P
Crouch S.
354P
Cruciani M.
291P
Cumming B.
131P, 248P
Cunningham F.
339P
Cusack S.
55
D
Author/Co-Author
Dahl J.
110P
Dalecki A.
348P
Dallenga T.
287P
Danilchanka O.
166P
Dartois V.
60, 216P, 227P, 337P
Daryan A.K.
181P
Das A.
186P, 210P
Das G.
275P
Das M.
210P
Datta P.
20
Davis L.
141P
De Carvalho L.
132P
De Chastellier C.
235P, 237P
De Jong B.
141P, 346P
De Libero G.
269P
De Matteis L.
330P
De Proft F.
206P
De Sessions P.F.
238P
De Souza Carvalho-Wodarz C.
296P
Debnath R.
210P
Debnath S.
210P
395
Degiacomi G.
54
Del Carmen Sasiain M.
46
Delacourt C.
258P
Delogu G.
253P
Delorme V.
318P
Demina G.
228P
Den Hertog A.
25
Deramaudt T.
235P
Derbyshire K.
14
Dhar N.
19, 85P, 156P
Dhiman R.
249P
Dhingra S.
250P
Di Camillo B.
291P
Diallo M.
7
Dian S.
89P, 308P
Diandini E.
278P
Dick T.
140P, 157P, 159P, 160P, 208P,
216P, 227P, 310P, 312P, 329P,
334P, 335P, 336P, 343P
Diedrich C.
296P
Dieli F.
22
Diels M.
141P
Diez-Tascón C.
272P
Dillon N.
319P
Dini L.
253P
Dionne M.
288P
Divangahi M.
41
Dockrell H.M.
239P
Dodd C.E.
49
Doherty A.
69P
Doherty T.M.
193P
Dolganov G.
141P
Dolinger D.
317P
Domenech P.
79P
Donini S.
19, 337P
Doornbos K.
166P
Doubovetzky M.
129P, 314P
Dougan G.
125P
Drancourt M.
94P, 209P, 229P
Drewes G.
109P
Driessen M.
141P
Drijkoningen J.
39
D. Robertson B.
143P
Drocourt D.
221P
Dubar F.
233P, 327P
Dubé J.
251P
Dubey N.
289P
Dubois V.
169P, 252P
Dufe V.
206P
Dumas E.
252P
Dupont C.
169P, 233P, 327P
Durán R.
215P
Dutilh B.
89P
Dutta A.
186P
Duval C.
298P
Dwivedi V.P.
275P
Dyatloff A.
263P
396
E
E. Schaible U.
Easom E.E.
Egere U.
Eggers L.
Ehrt S.
Ehtesham N.Z.
Eich J.
Eisenach K.
Ekins S.
El Messaoudi M.D.
Elayati K.
Elghraoui A.
Elharar Y.
Elliott S.
Engel R.
Eniyan K.
Epemolu R.
Equbal M.J.
Erdogan H.
Eskandarian H.A.
Esposito M.
Etna M.P.
Evangelopoulos D.
Evans J.
Everall I.
Eyangoh S.
Eynard N.
Ezewudo M.
F
Fabre E.
Farrow-Johnson J.
Fateh A.
Favrot L.
Fayolle C.
Feilcke R.
Feinberg J.
Feldmann J.
Fernandez-Reyes D.
Ferrer S.
Ferrer-Bazaga S.
Festjens N.
Fillatreau S.
Fimia G.
Finin P.
Fink L.
Fissette K.
Fletcher H.
Flo T.
Floto A.
Floto R.A.
Flynn J.
Fohtung J.
Fontaine E.
Foo C.
Forson A.
Author/Co-Author
194P
350P
267P
273P
9, 17, 154P
111P, 164P, 198P, 255P
287P
71P
54
313P
112P
76P, 77P, 83P
119P
290P
225P
211P
340P
198P
206P
18
54
291P
23
81P
39
40
321P
317P
Author/Co-Author
265P
145P
183P
205P, 212P, 230P
304P
320P
258P
70P
23
354P
339P
105P, 180P
46
253P, 291P
131P
91P, 92P
141P
239P
170P, 260P, 292P
288P
39
60
202P
129P, 314P
213P
142P, 231P, 349P
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
Forti F.
Fortune S.
Fraisse L.
Frank M.
Franzblau S.
Freire D.
Frigui W.
Fuerst J.
Furusho H.
Futterer K.
136P
7, 33, 115P, 126P, 202P
129P, 130P, 147P, 314P
84P
341P
113P
215P, 304P
3
172P, 173P
109P
G
Author/Co-Author
273P
214P
5, 66P, 67P, 70P, 71P,
81P, 86P, 141P
Gaibelet G.
265P
Gaifem J.
276P
Gaillard J.L.
234P, 235P
Ganiem A.
308P
Gao Q.
6
Garcia B.
141P
Garcia J.
137P
García S.
330P
García-García S.
272P
Garcia-Pelayo C.
181P
Garcia-Vallejo J.
163P
Garg R.
114P
Garton N.
280P
Garza-Garcia A.
51
Geerlof A.
207P
Gehre F.
141P
Genevaux P.
113P
Gengenbacher M.
157P, 160P, 216P, 336P
Gennaro M.
20
Gerrick E.
7, 115P
Geslin M.
129P
Ghadiri A.
324P
Ghanei M.
183P
Ghanem M.
78P, 79P
Ghisotti D.
136P
Ghoma Linguissi L.S.
84P
Giacomini E.
291P
Gicquel B.
30, 46, 193P
Gideon H.P.
195P
Gidon A.
292P
Gijsbers A.
173P
Gil E.
196P
Gil M.
215P
Gil Castro A.
270P, 276P
Gil-Brusola A.
182P
Gilleron M.
10, 127P, 221P, 265P
Gillespie S.
23, 158P
Gilman R.
266P
Ginbot Z.
271P
Ginda K.
121P
Giordano I.
350P
Gabriel G.
Gaevaya L.
Gagneux S.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
Girardi E.
253P
Girard-Misguich F.
169P, 252P
Gisbers A.
172P
Glenn S.
116P
Glickman M.S.
9
Go M.L.
334P
Gobe I.
100P, 102P
Goig Serrano G.
182P
Gold B.
37, 146P, 147P
Goldberg A.
126P
Goldmann T.
243P
Goletti D.
253P
Goliath R.
195P, 271P
Gomez R.
354P
Gomez-Romero M.
177P
González Del Río R.
339P
González-Candelas F.
75P
González-Montaner P.
306P
Gonzalo-Asensio J.
30, 178P, 190P
Goodarzi H.
65P
Goodmanson A.
91P
Gopal P.
160P, 216P, 227P
Gordon S.
59, 247P, 264P, 356P
Gormley E.
264P
Gosetti F.
54
Gouilleux L.
147P
Goulard C.
234P
Goullieux L.
314P
Goyal R.
148P
Grabowska A.
113P, 117P
Gracia B.
309P
Grassi A.
291P
Gray T.
14
Green S.
337P, 338P, 340P
Gref R.
318P
Greif G.
4
Gresham S.
351P
Griffiths G.
287P, 296P
Grode L.
27
Grogono D.
39
Gröschel M.
304P, 305P
Grüber G.
208P, 219P, 329P
Grzegorzewicz A.
321P
Guerardel Y.
209P, 220P, 233P, 327P
Guerin M.
34
Guermonprez P.
288P
Guerra-Laso J.
272P
Guijarro-Lopez L.
133P, 355P
Guilhot C.
8
Gupta A.
158P
Gupta M.
118P, 289P
Gupta P.
242P
Gupta V.
114P
Gur E.
119P
Gurumurthy M.
322P
Gutierrez C.
113P
Gutierrez M.
132P, 296P
Gygli S.
67P
397
H
Author/Co-Author
Hadizadeh Tasbiti A.
183P
Hafneh N.
254P
Hagens K.
194P
Haj Aissa F.
283P
Håkansson A.P.
332P
Haks M.C.
22
Haldar K.
280P
Haldar P.
280P
Hammond R.
158P
Han S.J.
179P, 185P
Hanna N.
207P
Harris S.
39, 66P, 86P
Harrison J.
204P, 340P
Hartkoorn R.
19
Harvey N.
177P
Hasenoehrl E.
101P
Hashemi Shahraki A.
325P
Hashemzadeh M.
325P
Hasnain S.E.
111P, 164P, 198P, 255P
Hattendorff J.
71P
Haug M.
260P
Hauptmann M.
256P
Haworth C.
39
Hayova L.
214P
Hee D.
322P
Hegde S.
138P, 212P
Henrissat B.
94P
Hernandez J.E.
120P
Hernandez V.
350P
Hernández-Pando R.
13
Herrmann J.L.
38, 169P, 233P, 234P,
235P, 241P, 252P, 327P
Herrmann T.
85P
Hibberd M.L.
238P
Hill E.
39
Hill J.
202P, 310P, 343P
Hill P.
278P
Hiller M.
301P
Hillery N.
317P
Hilpert K.
137P
Hingley-Wilson S.
120P
Hmama Z.
187P
Hoal E.G.
258P
Hoffmann H.
68P
Hoffmann-Thiel S.
68P
Hoffner S.
83P, 93P
Hofmann S.
137P
Hogarth P.J.
181P
Hohl Michael.
167P
Holmes E.
177P
Holst O.
225P
Honeyborne I.
23
Honoré N.
106P
Hopkins F.
7
Hotra A.
208P
Houben E.
13, 15, 165P, 168P
Houle D.
41
398
Houthuys E.
Hrabalek A.
Huan C.
Hudrisier D.
Huijnen M.
Hunt D.
Hunter J.
Hunter W.
Hurwitz R.
Hwa S.
Hwang M.H.
I
Iakobachvili N.
Imming P.
Indrati A.
Inwentarz S.
Ioerger T.
Iqbal I.
Isabel Veiga M.
Izumi H.
180P
323P, 342P
334P
43, 46
89P
132P
299P
217P
128P
257P
80P
Author/Co-Author
172P, 173P, 286P, 352P
320P, 341P
278P
306P
107P, 337P
294P
276P
3
J
Author/Co-Author
J Rubin E.
252P
Jabot-Hanin F.
258P
Jackson L.
91P
Jackson M.
10, 127P, 321P
Jacobs W.R.
2, 101P, 125P, 155P, 230P
Jäger F.
217P
Jakimowicz D.
121P
Jang M.S.
221P
Jang Y.H.
80P
Jankute M.
122P
Janmeja A.
242P
Janvier G.
222P
Jarlier V.
72P
Jary D.
330P
Javid A.
123P
Javid B.
124P, 161P
Jensen K.
229P
Jeong H.
184P
Jeronimo Frias Rose J.
300P
Ji S.
269P
Jiang X.
146P
Jim K.
315P
Jiménez B.
177P
Jiménez C.
13
Jiménez-Navarro E.
351P
Joe M.
204P
Joloba M.
71P
Jones V.
321P
Jonnala S.
131P
Joosten S.A.
22
Jouneau L.
46
Jouny S.
318P
Juambeltz A.
4
Juan-García J.
272P
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
K
Author/Co-Author
Kaiser P.
128P
Kalia N.
338P
Kalra R.
242P
Kalscheuer R.
123P
Kalsum S.
259P
Kamariza M.
218P
Kaminski L.
125P
Kampmann B.
177P, 267P
Kana B.
116P
Kandror O.
126P
Kaneko T.
130P
Kannan N.
260P
Kanno A.I.
178P
Kant S.
114P
Kaprelyants A.
228P
Kapuria A.
249P
Karabanovich G.
323P, 342P
Kaufmann S.H.
46, 128P, 157P
Kaya F.
216P, 227P
Keane J.
112P
Keating T.
240P
Keep N.
223P
Kendall S.
87P, 223P
Keri G.
19
Kester J.
126P
Khan R.
20
Khandan Dezfuli S.
325P
Khanna V.
106P
Khare G.
293P
Khosravi A.D.
65P
Khosravi Boroujeni A.
324P, 325P
Khubaib M.
164P, 255P
Kidwai S.
152P, 249P
Killick K.
247P
Kim H.
185P, 199P, 200P, 246P, 274P
Kim H.M.
261P
Kim J.M.
80P
Kim J.S.
185P, 199P, 246P, 261P, 274P
Kim N.R.
80P
Kim S.Y.
236P
Kim W.S. 185P, 199P, 200P, 246P, 261P, 274P
Kim Y.
77P
Kiravu A.
271P
Kispert A.
243P
Kissane K.
147P
Kitdorlang Dkhar H.
242P
Klieber S.
129P
Klimešová V.
323P, 342P
Kling A.
129P
Kneuper H.
217P
Koch A.
81P, 149P
Koeken V.
308P
Koh W.J.
236P
Kohl T.A.
84P
Kolbe K.
225P
Kolly G.
213P
Kondova I.
298P
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
Kondratieva E.
263P
Kondratieva T.
263P
Koram K.
66P, 86P
Korduláková J.
127P, 321P
Korman M.
119P
Korotkov K.
168P
Korotkova N.
168P
Kraigsley A.
353P
Krátký M.
326P, 347P
Kreitmann L.
41
Kremer L.
42, 94P, 169P, 209P, 220P, 229P,
233P, 234P, 235P, 237P, 241P,
252P, 327P
Kreutzfeldt K.
17
Krishnamoorthy G.
128P
Krishnan N.
307P
Krügel A.
167P
Kudryavtsev I.
201P
Kuijl C.
163P
Kuittinen I.
23
Kumar A.
148P, 211P, 219P, 294P
Kumar V.
308P, 328P
Kundu P.
186P
Kundu S.
208P, 329P
Kuroda M.
306P
Küssau T.
229P
Kutsch O.
277P
Kviatcowsky D.
306P
Kwedi S.
84P
Kwon K.W.
185P, 199P, 200P, 261P, 274P
L
Author/Co-Author
L. Andersen P.
194P
Laboudie P.
129P
Lacourcière K.
353P
Laencina L.
169P, 252P
Lagrange S.
19, 57, 129P, 130P, 147P, 314P
Lai R.
51
Lair C.
129P, 130P, 314P
Lakshmanan U.
310P, 343P
Lakssir B.
95P
Lalande J.D.
41
Lamers M.
33
Lamprecht D.
131P
Lang D.
156P
Lang R.
46
Lang U.
33
Lapidus A.
73P
Lapierre P.
14
Lardizabal A.
197P
Larrouy-Maumus G.
10, 104P, 132P
Lasserre M.
4
Lastrucci C.
298P
Lau A.
187P
Laughon B.
353P
Lavelle E.
356P
Layre E.
265P
399
Le Chevalier F.
215P
Le Moigne V.
169P, 234P, 252P, 327P
Lechartier B.
213P, 331P
Leclerc C.
304P
Lee H.S.
80P
Lee L.
322P
Lee M.H.
9
Lee R.E.
321P
Lee S.
184P, 303P
Lefrançois L.
295P
Leite L.C.
178P
Leitges M.
243P
Lelievre J.
233P, 316P
Lelli M.
85P
Leopold D.
267P
Lerm M.
171P, 259P, 262P, 266P, 300P
Lerner T.
296P
Leung L.
155P
Leyva F.J.
353P
Li X.
55, 350P, 354P, 355P
Liang Y.
232P
Lienard J.
297P
Lima A.
215P
Lin G.
37
Lincoln R.
351P
Lindenberger J.J.
321P
Lindhorst T.
225P
Lindroth M.
259P
Ling C.
310P, 343P
Ling Y.
147P
Linge I.
263P
Linke D.
301P
Lisa N.
215P
Liu J.
336P
Liu S.
335P
Lloyd G.
204P
Locht C.
283P
Lodh A.
210P
Loitto V.
171P
López G.
175P
Lopez Quezada L.
147P
López-Fidalgo E.
272P
Lopez-Iglesias C.
172P, 173P, 352P
López-Medrano R.
272P
Lopez-Perez P.M.
137P
Lotterman A.
202P
Louet C.
292P
Lounis N.
346P
Lovering A.
204P
Low J.L.
216P
Lowary T.L.
204P
Loyens M.
283P
Lu Q.
322P
Lucía A.
330P
Lugo-Villarino G.
298P, 306P
Lupien A.
19, 213P, 331P
Lupoli T.
37
Lustig G.
299P
400
Lutfalla G.
Luukinen H.
M
Ma L.
Ma N.
Małecki T.
Machado H.
Machugh D.
Mackenzie C.
Madduri A.
Madzou-Laboum I.K.
Maerk M.
Magee D.
Magnon M.
Mahajan S.
Mahamed D.
Mahmoudi S.
Majlessi L.
Mak Y.S.L.
Makarov V.
Makhene M.
Makinoshima H.
Malakar B.
Maletta M.
Malm S.
Malone K.M.
Mamishi S.
Manfredi M.
Manganelli R.
Mangas K.
Manimekalai S.
Manina G.
Manjari S.
Mao W.
Maqueda M.
Marais S.
Marengo E.
Margolis A.
Maridonneau-Parini I.
Mariller C.
Marino L.
Marinova D.
Marlovits T.
Marriner G.
Martin C.
Martin D.
Martin Z.
Martín C.
Martineau A.
Marwitz S.
Maserumule M.
Matange N.
Mathonsi C.
Mawatwal S.
Maximov G.
Mayer C.
241P
189P
Author/Co-Author
106P
120P
121P
276P
247P
340P
132P
84P
170P
247P, 264P
129P, 130P, 314P
242P
257P, 299P
188P
235P, 304P, 305P
350P
56, 116P, 331P
353P
9
281P
163P
84P, 225P
264P
188P
54
20, 54, 291P
3
219P
12
279P
350P
309P
51
54
321P
298P, 306P
220P
132P
30
15, 165P
60
30, 178P
81P
156P
190P
196P
243P
288P
104P
299P
249P
201P
222P
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
Mazurek J.
Mc Neil M.
Mcbride A.
Mcfadden J.
Mchugh T.
Mc-Hugh D.E.
Mcintosh F.
Mckinney J.
Mcloughlin K.
Mcneil M.R.
M. De La Fuente J.
Meehan C.
Meghdadi H.
Meijgaarden K.E.
Meikle V.
Meiler E.
Mellors T.
Méndez M.
Mendoza-Losana A.
265P
10
353P
102P, 120P, 134P, 279P
23
264P
41, 78P, 79P, 251P
19, 156P
247P
321P
330P
141P, 346P
324P
22
332P
133P
202P
266P
55, 133P, 339P, 350P,
351P, 354P, 355P
Mendum T.
102P, 134P, 295P
Mendy A.
267P
Menezes S.
288P
Mercier I.
46, 298P, 306P
Merker M.
68P
Mery A.
220P
Messens J.
206P
Mi J.
146P
Mikušová K.
54, 127P, 323P
Milla A.
314P
Miller R.
196P
Minato Y.
319P
Minch K.J.
145P
Minnikin D.E.
82P
Miotto P.
108P, 317P
Mirsaeidi M.
324P
Mitra A.
135P
Mizrahi V.
19, 81P, 149P, 156P, 340P
Möckl L.
225P
Modlin S.
76P, 83P
Mohan M.
350P
Molle V.
150P
Mollenkopf H.J.
128P, 157P
Möllmann U.
341P
Mondesert G.
19, 147P
Monk I.
3
Montamat-Sicotte D.
41, 251P
Moody B.
79P, 132P
Moores A.
96P
Moosa A.
345P
Moraña E.J.
306P
Moraski G.
333P
Moreira W.
140P, 312P, 334P
Moreno P.
39
Mori G.
54
Morozova T.
268P
Morris-Jones S.
196P
Movert E.
297P
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
Mukamolova G.
Mukherjee D.
Muller T.L.
Müller R.
Munier-Lehmann H.
Munshi T.
Murugesan D.
Musters R.
Muvva J.
Myllymäki H.
116P, 136P, 254P, 286P
335P
271P
129P
222P
137P
340P
163P
300P
189P
N
Author/Co-Author
150P
322P
36, 138P
299P
247P, 264P
139P, 144P
210P
151P, 250P
281P
242P
219P
37, 146P, 147P,
Nathan C.
154P
Navarro F.
330P
Navas V.H.
106P
Naya H.
4
Nebreda-Mayoral T.
272P
Nedialkova L.
68P
Negatu D.
336P
Němeček J.
323P, 342P
Neo H.
269P
Neres J.
213P
Netea M.
308P
Neyrolles O.
43, 46, 113P, 221P, 298P, 306P
Ngan G.J.Y.
140P
Nicol M.
81P
Niederweis M.
16, 135P, 166P, 332P
Niemann S.
68P, 84P, 346P
Nigou J.
10, 122P, 127P, 221P
Niskanen M.
189P
Noack S.
122P
Noh K.
102P
North E.J.
321P
Notebaart R.
308P
Notelaers K.
172P, 173P
Noursadeghi M.
196P
Novais Bastos H.
276P
Ntoumi F.
84P
Nuermberger E.L.
130P
Nukdee K.
113P
Nur M.
286P
Nurani A.
278P
Nyantakyi S.
334P
Naandicoori V.K.
Naftalin C.
Nagaraja V.
Naicker M.
Nalpas N.
Nambi S.
Nanda R.
Nandi M.
Nandicoori V.
Nanduri R.
Nartey W.
401
O
Obrien S.
Odermatt N.
Ofori-Anyinam B.
O'garra A.
Oh D.
O'hare H.
O'leary S.
Omari S.
Omondi F.
Ong R.T.H.
Oni T.
Oom L.
Oravcova K.
Ordway D.
Orena B.S.
Ortega F.
Osei-Wusu S.
Ostrovsky D.
O'sullivan M.
Otchere I.D.
Ottenhoff T.
Otter A.
Oualim A.
Ouerfelli O.
Ouni R.Y.M.
P
Pacitto A.
Pain A.
Palencia A.
Palló A.
Palmer T.
Palucci I.
Pan M.M.
Pancani E.
Pandey S.
Paquet M.
Parasa V.R.
Pardini M.
Parikka M.
Parish T.
Park S.
Park Y.
Parkhill J.
Parret A.
Pasca M.R.
Pato J.
Paton N.
Pavan F.
Pávek P.
Pawlik A.
Péan C.
Pedre B.
Pellet A.
Penlap Beng V.
Perdih A.
402
Author/Co-Author
73P, 74P
85P
141P
46
77P
35
112P
142P, 349P
244P
160P
81P, 195P, 271P
299P
158P
39
54
354P
66P, 86P, 142P, 349P
228P
112P
66P, 86P, 142P, 349P
22
87P
314P
154P
283P
Author/Co-Author
19
13
55
206P
217P
253P
161P
318P
111P, 164P, 198P, 255P
41
300P
291P
189P
194P
146P
60, 337P
39, 66P, 81P, 86P
15, 113P, 165P, 207P
54
19
322P
132P
323P
106P, 175P, 233P, 304P
288P
206P
314P
84P
211P
Pérez Sánchez I.
Perez-Herran E.
Peters P.
Peterson E.
Petrella S.
Petrone L.
Petrova E.
Petruccioli E.
Pettigrove M.
Pham A.
Pham H.
Phan T.
Phelan J.
Picavet D.
Pidot S.
Pilátová M.
Pillay K.
Pingris K.
Pióro M.
Pissis C.
Pitard B.
Piton J.
Plattner J.J.
Plets E.
Poce G.
Pogenberg V.
Poincloux R.
Poquet Y.
Portela M.
Porter J.
Posey J.
Poulsen A.
Pourakbari B.
Prados-Rosales R.
Prandi J.
Prasad A.
Prideaux B.
Priestman M.
Prieto A.
Pritchard J.
Prosser G.
Pyle C.J.
Pym A.
30, 190P
55, 133P, 350P
172P, 173P, 352P
99P
222P
253P
263P
253P
91P
72P
10
168P
238P
25, 163P
3
288P
299P
306P
121P
222P
234P
213P
350P
105P, 180P
32
113P
298P
221P
215P
3
317P
310P, 343P
188P
191P, 192P
221P
279P
60
143P
69P
7, 223P
338P
49
61, 299P
Q
Author/Co-Author
321P
Quémard A.
R
Rae Sajorda D.
Raffetseder J.
Ragan T.
Ragunathan P.
Rahman M.
Rajaram M.
Rajwani R.
Rakotosamimanana N.
Ramakrishnan L.
Author/Co-Author
101P
171P
132P
208P
131P
49
88P
193P
63, 284P, 302P
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
Rämet M.
189P
Ramirez-Busby S.
91P
Ramon Garcia S.
339P
Ramos A.
276P
Rampersad S.
299P
Ramsheh M.
280P
Ranganathan A.
275P
Rao S.
224P
Raposo-García S.
272P
Rasolofo V.
193P
Ravelli R.
172P, 173P
Ravi J.
20
Rawkins A.
58
Ray P.
337P, 338P, 340P
Rayasam G.
211P
Raynaud-Messina B.
306P
Raze D.
283P
Read K.
340P
Redinger N.
194P
Redmond S.
302P
Reed M.
79P
Reed S.G.
200P
Regad L.
222P
Rego E.H.
161P
Rego H.
11
Rehwinkel J.
46
Reiche M.
156P
Reiling N.
225P, 243P, 245P, 301P
Reimer R.
287P
Remuinan M.
354P
Repnik U.
287P, 296P
Resende M.
270P
Reseqtb Consortium C.
317P
Ressami E.
95P
Reynolds E.
204P
Rhee K.
337P
Riaz T.
103P
Riccardi G.
54
Richard V.
193P
Richard-Greenblatt M.
98P
Richter A.
341P
Riesco A.
96P
Rigouts L.
346P
Ring S.
273P
Riou C.
195P, 244P, 271P
Rittershaus E.
144P
Rivera B.
215P
Rivero-Lezcano O.
272P
Rizal Ganiem A.
89P
Rizzi M.
19, 337P
Robbins K.K.
60
Robello C.
4
Roberts J.
146P, 147P
Robertson B.
307P
Roca Soler F.
302P
Rock F.L.
350P
Rock J.
7, 33
Rockwood N.
51
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
Rodrigues F.
276P
Rodriguez-Beltran J.
69P
Rodriguez-Rincon D.
39
Rodriguez-Rivera F.
226P
Rodwell T.
91P, 317P
Roe J.
196P
Roh J.
323P, 342P
Rohde M.
296P
Rojas A.
69P
Romagnoli A.
253P, 291P
Ronacher K.
23
Ronning D.R.
321P
Rossello J.
215P
Rotkevich M.
73P, 74P
Rottman M.
235P
Rouanet C.
283P
Roubert C.
147P
Rousu J.
23
Roux A.
235P
Rubin E.
7, 11, 21, 107P, 169P, 202P
Rue-Albrecht K.
264P
Ruesen C.
89P, 308P
Rullas J.
355P
Rullas-Trincado J.
133P
Rüsch-Gerdes S.
84P
Russell D.
52
Russell M.
296P
Russell S.
131P
Rustad T.
145P
Ryan L.
292P
Rybniker J.
213P
S
Sacchettini J.
Sachs N.
Saelens J.
Saeys Y.
Saini A.
Saito K.
Sajid A.
Sakwa I.
Sala A.
Sala C.
Salina T.
Salzano G.
Sampson S.
Sander P.
Sans S.
Santos Villarejo A.
Sapriel G.
Saqib M.
Saraiva M.
Sarathy J.
Šarkan M.
Sarkar D.
Sasiain M.C.
Sassetti C.
Author/Co-Author
337P
352P
303P
105P
242P
37, 146P
150P
46
113P
19, 138P, 174P, 213P
268P
318P
97P
28
129P, 130P, 147P
339P
252P
198P
276P
60, 216P, 227P, 227P
127P
148P
298P, 306P
7, 107P, 139P, 144P
403
Sayes F.
304P, 305P
Schaaf K.
277P
Schaible U.
245P, 256P, 287P
Schena E.
68P
Schiebler M.
288P
Schierloh P.
46
Schito M.
317P
Schlesinger L.S.
49
Schlussel S.
119P
Schnappinger D.
7
Schneider B.
273P
Schneider T.
113P
Scholl E.
25
Schoolnik G.
141P
Schubert O.
67P
Schurr E.
258P
Schütze S.
301P
Schwudke D.
243P
Scott M.
158P
Scullion P.
340P
Scutigliani E.
25
Seeger Markus.
167P
Seemann T.
3, 106P
Sefrioui H.
313P
Seldon R.
81P
Seng K.Y.
322P
Serebryakova M.
201P
Serrano I.
330P
Seth A.
114P
Severa M.
291P
Sewgoolam B.
149P
Shah S.
348P
Shahrezaei V.
143P
Shanmugan R.
91P, 93P
Sharma A. 111P, 150P, 164P, 197P, 198P, 289P
Sharma C.
242P
Sharples K.
278P
Sheikh J.A.
198P
Shen L.
220P
Sherman D.R.
145P
Shetty A.
343P
Shi L.
10
Shin E.
184P
Shin S.J.
179P, 185P, 199P, 200P, 236P,
246P, 261P, 274P
Shleeva M.
228P
Shokrgozar M.
183P
Shulgina M.
73P, 74P
Siadat S.
183P
Sidibe A.
84P
Sigal A.
257P, 299P
Sikri K.
151P, 250P
Silve S.
130P
Simeone R.
235P, 305P
Simpson N.
196P
Sina A.H.
324P
Singh A.
99P, 249P, 281P
Singh D.K.
275P
404
Singh H.N.
Singh L.K.
Singh N.
Singh R.
Singh S.
Singh V.
Singh Y.
Singhal A.
Sinigaglia A.
Siroy A.
Sisk D.
Sitaraman R.
Siu G.
Sizemore C.F.
Škovierová H.
Skroch S.
Slizen V.
Smith C.
Smith S.R.
Smyth A.
Sogi K.
Sohst V.
Soldati T.
Soler P.
Solovieva N.
Somersan-Karakaya S.
Sommer R.
Somody L.
Song O.
Song S.
Sørensen K.
S. Osório N.
Soualhine H.
Sougakoff W.
Souriant S.
Sousa J.
Sparrius M.
Speer A.
Sreejit G.
Staack S.
Stacey H.
Stafford S.
Stanley S.
Steigedal M.
Steinhäuser C.
Stendahl O.
Stewart G.
Steyn A.
Steyn A.J.C.
Stinear T.
Stolaříková J.
Stout J.
Stramer B.
Strand T.
Street L.
Stucki D.
Sturegård E.
Subbarao S.
328P
150P
148P
152P, 249P
293P
19, 187P
150P, 289P
269P
291P
172P, 173P
303P
118P
88P
353P
10
299P
153P
14
277P
264P, 356P
44
225P
50, 282P, 295P
174P
73P, 74P
37, 146P
19, 357P
314P
305P
9
229P
276P
187P
72P
306P
276P
315P
135P, 315P
299P
207P
158P
196P
44
170P, 260P
301P
266P
279P, 295P
98P, 131P
248P
3, 106P
323P, 342P, 347P
303P
288P
170P
345P
81P
297P
307P
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
Sun J.
Supply P.
Surkova L.
Suter M.
Sweet G.
Szabó N.
Székely R.
T
166P, 277P
106P
153P
208P
339P
326P
323P
Tagliani E.
Tailleux L.
Takiff H.
Talaat A.
Tan S.
Taylor S.
Tchikov V.
Tekwu E.M.
Teo Y.Y.
Tessema B.
Thakur P.
Tholey A.
Thomas N.
Thomas S.
Thompson C.
Thomson N.R.
Thygesen M.
Tiago Guimarães J.
Tientcheu L.
Tiraby G.
Tischler A.
Tiwari D.
Tiwari P.
Tobin D.
Togun T.
Tomar A.K.
Tønjum T.
Torta F.
Totolian A.
Toulza F.
Trauner A.
Tria G.
Triglia D.
Tripathi D.
Troegeler A.
Trovato A.
Trutneva K.
Tsaliki E.
Tsang P.
Tsodikov O.
Tufariello J.
Turapov O.
Turner S.
Tyagi A.K.
Tyagi J.
Author/Co-Author
317P
46
175P
90P
288P
240P
301P
84P
160P
317P
162P
301P
196P
240P
339P
125P
229P
276P
177P, 267P
221P
290P
242P
152P
303P
177P
328P
69P, 103P
269P
201P
239P
67P
172P, 173P
112P
111P, 114P, 255P
43, 298P
68P
228P
196P
340P
344P
230P
116P, 136P
351P
293P
151P, 250P
U
Ummels R.
Author/Co-Author
13, 168P
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
Upton A.
Ussher J.
Utpatel C.
129P, 130P
278P
68P
V
Author/Co-Author
Valášková L.
323P
Valafar F.
76P, 77P, 83P, 91P, 92P, 93P
Valaskova L.
342P
Valfridsson C.
297P
Van T.
141P
Van Bergen L.
206P
Van Crevel R.
89P, 278P
Van De Weerd R.
163P, 315P
Van Der Sar A.
286P
Van Der Wel N.
25, 163P
Van Der Westhuyzen R.
345P
Van Der Woude A.
163P
Van Helden P.
23
Van Laarhoven A.
89P, 308P
Van Molle I.
206P
Van Stempvoort G.
163P
Van Veen H.
25
Van Wyk N.
94P, 169P, 209P, 229P
Vanderschaeghe D.
180P
Vandewalle K.
105P, 180P
Vasileva E.
201P
Vaubourgeix J.
9, 37, 154P
Vaziri F.
183P
Veerapen N.
122P
Veerasami M.
279P
Verástegui M.
266P
Verbov V.
201P
Vercellone A.
221P
Vergne I.
235P
Vergnolle O.
205P, 230P
Verma D.
39
Verma R.
322P
Vermet H.
19
Vérollet C.
306P
Verrall A.
278P
Verreck F.
298P
Veyrier F.
356P
Veziris N.
72P
Via L.E.
60, 337P
Vieira C.
276P
Vieira J.
276P
Vilcheze C.
155P
Viljoen A.
220P, 233P, 235P, 237P, 252P
Villarreal-Ramos B.
247P, 279P
Villaume S.
220P
Villellas C.
346P
Vinšová J.
326P
Vincent S.
220P
Vinsova J.
347P
Visweswariah S.
104P
Vitoria A.
309P
Vocat A.
19, 331P
Voigt K.
341P
405
Vollmer W.
Vordermeier H.
Vordermeier M.
Voskuil M.
Vouvoungui J.C.
Vuylsteke M.
136P
247P
279P
141P
84P
105P
W
Author/Co-Author
Waddell S.
23, 69P, 96P, 136P
Wagner J.
202P
Wahni K.
206P
Wainwright H.
296P
Walker B.
202P
Walter N.
141P
Walters-Morgan H.
204P
Walzl G.
23
Wampande E.
71P
Wang F.
101P
Wang J.
78P, 79P
Wang R.
17
Wang Y.
269P
Warner D.
19, 81P, 149P, 156P, 340P, 345P
Warren R.
13, 97P
Warrier T.
37, 146P
Webb P.R.
181P
Wenk M.
269P
West N.
3
Wetzlar T.
341P
Wheat W.
10
Wilkinson K.A.
195P
Wilkinson R.
24, 51, 81P, 195P, 244P,
271P, 296P
Willand N.
318P
Williams C.
280P
Williams K.
279P
Williams M.
97P
Wilmanns M.
15, 113P, 165P, 207P
Wiredu A.
231P
Witt K.
196P
Wolschendorf F.
348P
Wooff E.E.
181P
Wu H.
279P
Wu M.L.
157P
Wu Y.
124P
Wyatt P.
340P
X
Xi L.
Xiao H.
Xu H.
Xu W.
Xu Z.
Y
Yadav S.
Yam J.
406
Yam W.C.
Yamada Y.
Yang G.
Yang T.
Yang T.J.
Yari S.H.
Yeboah-Manu D.
Yee M.M.K.
Yimer S.
Yoo J.
Young D.
Yun Y.
Yurchenko A.
Z
Zalutskaya A.
Zech G.
Zelmer A.
Zhang D.
Zhang J.
Zhang M.
Zhang Y.W.
Zheng X.H.
Zhou Y.
Zhu J.H.
Zhuravlev V.
Zimmerman M.
Zoltner M.
Zou H.
Zou W.
Zuccotto F.
Zumla A.
88P
159P, 310P, 343P
154P
334P
130P
183P
66P, 86P, 142P, 349P
160P, 216P
103P
184P
51, 79P, 96P
256P
73P, 74P
Author/Co-Author
153P
129P
194P
147P
130P
174P
124P
124P
55, 350P
124P, 161P
73P, 74P
60
217P
335P
350P
340P
193P
Author/Co-Author
351P
158P
230P
9
32, 176P
Author/Co-Author
328P
232P
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
PARTICIPANT LIST
September 2nd, 2016
ABEL Laurent
Hôpital Necker Enfants Malades
Paris, France
[email protected]
ANDERSEN Peter Lawætz
Statens Serum Institut
Coppenhagen S, Denmark
[email protected]
ADOLF Günther
Austrianni GmbH
Vienna, Austria
[email protected]
ANDREAS Nicholas
Imperial College London
London, United Kingdom
[email protected]
AGARWAL Nisheeth
Translational Health Science and
Technology Institute
Faridabad, India
[email protected]
ANDREU Nuria
London School of Hygiene and
Tropical Medicine
London, United Kingdom
[email protected]
AGUILAR PÉREZ Clara
University of Zaragoza
Zaragoza, Spain
[email protected]
ANWAR Shaheda
London School of Hygiene and
Tropical Medicine
London, United Kingdom
[email protected]
AINHOA Lucia
University of Zaragoza
Zaragoza, Spain
[email protected]
AKABAYOV Barak
Ben-Gurion University of the Negev
Be'er Sheva, Israel
[email protected]
ALBEL Singh
University of Birmingham
Birmingham, United Kingdom
[email protected]
ALEMPARTE Carlos
GlaxoSmithKline
Tres Cantos, Spain
[email protected]
ALLEY Dickon
Anacor Pharmaceuticals
Palo Alto, United States
[email protected]
ALVAREZ ARGUEDAS Samuel
University of Zaragoza
Zaragoza, Spain
[email protected]
ALZARI Pedro
Institut Pasteur
Paris, France
[email protected]
AMORIM FRANCO Tathyana Mar
Albert Einstein College of Medicine
Bronx, United States
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
ARAGAW Wassihun
NUS
Singapore
[email protected]
ARNVIG Kristine
University College London
London, United Kingdom
[email protected]
ARORA Divya
National Institute of Immunology
New Delhi, India
[email protected]
ARRIES Jesmine
Stellenbosch University
Cape Town, South Africa
[email protected]
ARTHUR Michel
UMRS 1138-UPMC INSERM UPD
Paris, France
[email protected]
ASTOLFI ROSADO Leonardo
Vrije Universiteit Brussel
Brussels, Belgium
[email protected]
ATES Louis
Institut Pasteur
Paris, France
[email protected]
AVELINO Asia
Stanford University
Stanford, United States
[email protected]
409
AV-GAY Yossef
University of British Columbia
Vancouver, Canada
[email protected]
BASU Piyali
University of Surrey
Slough, United Kingdom
[email protected]
AZIZ Dinah
National University of Singapore
Singapore
[email protected]
BASU ROY Robin
Imperial College London
London, United Kingdom
[email protected]
BACON Joanna
Public Health England,
National Infection Service
Salisbury, United Kingdom
[email protected]
BAUGHN Anthony
University of Minnesota
Minneapolis, United States
[email protected]
BAILO VERGARA Rebeca
University of Birmingham
Birmingham, United Kingdom
[email protected]
BALLELL Lluis
GSK
Miraflores De La Sierra, Spain
[email protected]
BANAEI ESFAHANI Amir
Institute of Molecular Systems Biology, ETH
Zurich
Zurich, Switzerland
[email protected]
BARBIER Thibault
T.H. Chan Harvard School of Public Health
Boston, United States
[email protected]
BARCZAK Amy
Massachusetts General Hospital
Cambridge, United States
[email protected]
BARISCH Caroline
University of Geneva
Geneva 4, Switzerland
[email protected]
BARKAN Daniel
Hebrew University
Rehovot, Israel
[email protected]
BARRY Clifton
NIH
Bethesda, United States
[email protected]
BASU Nirmalya
Max anck Institute for Molecular Cell Biology
Dresden, Germany
[email protected]
410
BAULARD Alain
Institut Pasteur de Lille
Lille, France
[email protected]
BAUMANN Christoph
Austrianni GmbH
Vienna, Austria
[email protected]
BECKHAM Kate
EMBL Hamburg
Hamburg, Germany
[email protected]
BEHAR Samuel
University of Massachusetts Medical School
Worcester, United States
[email protected]
BEITES Tiago
Weill Cornell Medicine
New York, United States
[email protected]
BELLINZONI Marco
Institut Pasteur
Paris, France
[email protected]
BENABDESSALEM Chaouki
Institut Pasteur de Tunis
Tunis, Tunisia
[email protected]
BENTALEB El Mehdi
Moroccan Foundation for Advanced Science,
Innovation and Research (MAScIR)
Rabat, Morocco
[email protected]
BERNARD Christine
Université Pierre et Marie Curie
Paris, France
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
BERNARD Lucie
CNRS
Toulouse, France
[email protected]
BLAZQUEZ Jesus
Centro Nacional de Biotecnologia
Madrid, Spain
[email protected]
BERNEY Michael
Albert Einstein College of Medice
Bronx, United States
[email protected]
BOECK Lucas
University of Cambridge
Fulbourn, United Kingdom
[email protected]
BERNUT Audrey
CNRS
Montpellier, France
[email protected]
BOISSON-DUPUIS Stéphanie
INSERM Hôpital Necker/Rockefeller University
United States
[email protected]
BESTE Dany
University of Surrey
Guildford, United Kingdom
[email protected]
BONGAERTS Nadine
Institut Cochin
Paris, France
[email protected]
BHATT Apoorva
University of Birmingham
Birmingham, United Kingdom
[email protected]
BOOT Maikel
VU medical center
Amsterdam, The Netherlands
[email protected]
BIRHANU Alemayehu Godana
University of Oslo
Oslo, Norway
[email protected]
BORGERS Katlyn
VIB - Ghent University
Zwijnaarde, Belgium
[email protected]
BISWAS Priyanka
Indian Institute of Science
Bangalore, India
[email protected]
BORITSCH Eva
Institut Pasteur
Paris, France
[email protected]
BITTER Wilbert
Vu Medical Centre Amsterdam
Amsterdam, The Netherlands
[email protected]
BORRELL Sonia
Swiss Tropical and Public Health
Basel, Switzerland
[email protected]
BIUKOVIC Goran
National University of Singapore, Yong Loo Lin
School of Medicine
Singapore
[email protected]
BOSHOFF Helena
National Institutes of Health
Bethesda, United States
[email protected]
BLAISE Mickael
CNRS
Montpellier, France
[email protected]
BLANC Isabelle
SANOFI
Marcy L'etoile, France
[email protected]
BLANC-POTARD Anne
CNRS
Montpellier, France
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
BOTELLA Helene
Weil Cornell Medicine
New York, United States
[email protected]
BOTTAI Daria
University of Pisa
Italy
[email protected]
BOULON Richard
Institut de pharmacologie et
biologie structurale de Toulouse
Toulouse, France
[email protected]
411
BOUTTE Cara
Harvard TH Chan School of Public Health
Somerville, United States
[email protected]
BRANDENBURG Julius
Research Center Borstel
Borstel, Germany
[email protected]
BRENNAN Patrick
Colorado State University
Fort Collins, United States
[email protected]
BRESCIANI Nadia
San Raffaele Scientific Institute
Milan, Italy
[email protected]
BRITES Daniela
Swiss Tropical and Public Health Institute
Basel, Switzerland
[email protected]
BRODIN Priscille
Institut Pasteur de Lille / INSERM U10109
Lille, France
[email protected]
BROSCH Roland
Institut Pasteur
Paris, France
[email protected]
BROSET Esther
Universidad De Zaragoza
Zaragaoza, Spain
[email protected]
BROSSIER Florence
Pitié-Salpêtrière Hospital
Paris, France
[email protected]
BRUCH Eduardo
Institut Pasteur
Paris, France
[email protected]
BRYANT Josephine
University of Cambridge
Cambridge, United Kingdom
[email protected]
BUCHIERI Maria Virginia
Institut Pasteur
Sartrouville, France
[email protected]
412
BUCHRIESER Carmen
Institut Pasteur
Paris, France
BULL Tim
St George's University of London
London, United Kingdom
[email protected]
BUNDUC Catalin
Vrije Universiteit, Amsterdam
Amsterdam, The Netherlands
[email protected]
BURBAUD Sophie
Cambridge University
Cambridge, United Kingdom
[email protected]
BURGERS Wendy
University of Cape Town
Cape Town, South Africa
[email protected]
BURMEISTER Maike
Research Center Borstel
Borstel, Germany
[email protected]
CAMBIER Cj
Stanford
Stanford, United States
[email protected]
CANDEL Sergio
Cambridge University
Cambridge, United Kingdom
[email protected]
CAPTON Estelle
UPMC -Cimi Paris
Paris, France
[email protected]
CARRAT Christophe
IPBS
Toulouse, France
[email protected]
CASCIOFERRO Alessandro
Institut Pasteur
Paris, France
[email protected]
CASTAÑEDA GARCIA Alfredo
Instituto de Biomedicina Sevilla
Sevilla, Spain
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
CAVALIER Jean-François
CNRS
Marseille, France
[email protected]
COLLINS Ben
ETH Zurich
Zurich, Switzerland
[email protected]
CHAE Hansong
Yonsei University
Seoul, South Korea
[email protected]
COMAS Iñaki
Institute of Biomedicine of Valencia
Valencia, Spain
[email protected]
CHARLOTTE Passemar
University of Cambridge
Cambridge, United Kingdom
[email protected]
COMMANDEUR Susanna
VUmc
Weesp, The Netherlands
[email protected]
CHERNYAEVA Ekaterina
St. Petersburg State University
St. Petersburg, Russia
[email protected]
COOPER Andrea
Trudeau Institute/University Leceister
Leicester, United Kingdom
[email protected]
CHIARADIA Laura
Institut de Pharmacologie et
de Biologie Structurale
Toulouse, France
[email protected]
COPPENS Rene
TBVI
Lelystad, The Netherlands
[email protected]
CHINER-OMS Álvaro
University of Valencia
Valencia, Spain
[email protected]
CHIODARELLI Giacomo
GlaxoSmithKline
Tres Cantos, Spain
[email protected]
CHNG Shu Sin
National University of Singapore
Singapore
[email protected]
CORREIA Carolina
University College Dublin
Dublin, Ireland
[email protected]
CORTES Teresa
London School of Hygiene & Tropical Medicine
London, United Kingdom
[email protected]
COSCOLLA Mireia
SwissTPH
Basel, Switzerland
[email protected]
CHOI Hong-Hee
Yonsei University College of Medicine
Seoul, South Korea
[email protected]
COSTA-GOUVEIA Joana
Univ. Lille, CNRS, Inserm, CHU Lille
Institut Pasteur de Lille
Lille, France
[email protected]
CIMINO Mena
Institut Pasteur
Paris, France
[email protected]
COUGOULE Céline
IPBS CNRS UMR5089
Toulouse, France
[email protected]
COHEN-GONSAUD Martin
Centre de Biochimie Structurale
Montpellier, France
[email protected]
COX Jeffery
Berkeley University
San Francisco, United States
[email protected]
COLE Stewart
EPFL
Lausanne, Switzerland
[email protected]
COX Jonathan
Aston University
Birmingham, Uk, United Kingdom
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
413
COYA RABOSO Juan Manuel
Institut Pasteur
Paris, France
[email protected]
CUMMING Bridgette
KwaZulu-Natal Research Institute for
Tuberculosis and HIV
Durban, South Africa
[email protected]
DHIMAN Rohan
National Institute of Technology, Rourkela,
Odisha, India
Rourkela, India
[email protected]
DHINGRA Sakshi
All India Institute of Medical Sciences
Delhi, India
[email protected]
DALLENGA Tobias
Research Center Borstel
Borstel, Germany
[email protected]
DICK Thomas
National University of Singapore
Singapore
[email protected]
DARTOIS Veronique
Rutgers University
Newark, United States
[email protected]
DILLON Nicholas
University of Minnesota
Lakeville, United States
[email protected]
DARWIN Heran
New York University School of Medicine
New York, United States
[email protected]
DODD Claire
The Ohio State University
Columbus, United States
[email protected]
DE ROSSI Edda
University of Pavia
Pavia, Italy
[email protected]
DOORNBOS Kathryn
University of Alabama at Birmingham
Birmingham, United States
[email protected]
DEBOOSERE Nathalie
Institut Pasteur de Lille
Lille, France
[email protected]
DOU Horng-Yunn
National Health Research Institutes
Miaoli County, Taiwan
[email protected]
DEBRÉ Patrice
AP-HP
Paris, France
[email protected]
DUBAR Faustine
Université Lille 1
Villeneuve D'ascq, France
[email protected]
DEMANGEL Caroline
Institut Pasteur
Paris, France
[email protected]
DUBE Jean-Yves
McGill University
Montreal, Quebec, Canada
[email protected]
DEMARET Julie
Queen Mary University of London
Lyon, France
[email protected]
DUBEY Neha
University of Delhi
Delhi, India
[email protected]
DEMOITIE Marie-Ange
GlaxoSmithKline Vaccines
Wavre, Belgium
[email protected]
DUBOIS Violaine
UVSQ
La Celle-Saint-Cloud, France
[email protected]
DERBYSHIRE Keith
Wadsworth Center, New York State
Department of Health
Albany,, United States
[email protected]
DUMA Bogdan
GlaxoSmithKline
Tres Cantos, Spain
[email protected]
414
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
DUMAS Alexia
CNRS
Toulouse, France
[email protected]
EPEMOLU Ola
University of Dundee
Dundee, United Kingdom
[email protected]
DUPIN marilyne
1979
Vaugneray, France
[email protected]
ESKANDARIAN Haig Alexander
École Polytechnique Fédérale de Lausanne
Lausanne, Switzerland
[email protected]
DUPONT Christian
Centre d’études d’agents Pathogènes et
Biotechnologies pour la Santé
Montpellier, France
[email protected]
ETNA Marilena Paola
Istituto Superiore di Sanità
Roma, Italy
[email protected]
DURAN Rosario
Institut Pasteur de Montevideo
Montevideo, Uruguay
[email protected]
EDELSTEIN Paul
University of Pennsylvania
Philadelphia, United States
[email protected]
EHRT Sabine
Weill Cornell Medical College
New York, United States
[email protected]
EL ACHKAR Salam
Institut Pasteur de Lille
Lille, France
[email protected]
EVANGELOPOULOS Dimitrios
The Francis Crick Institute
London, United Kingdom
[email protected]
EVERALL Izzy
Wellcome Trust Sanger Institute
Cambridge, United Kingdom
[email protected]
EYANGOH Sara
Centre Pasteur du Cameroun
Yaoundé, Cameroon
[email protected]
FAVROT Lorenza
Albert Einstein College of Medicine
Bronx, Ny, United States
[email protected]
EL DAKER Sary
K-RITH
Durban, South Africa
[email protected]
FEILCKE Ruth
Martin-Luther-University Halle-Wittenberg,
Germany
Halle (saale), Germany
[email protected]
ELAYATI Karima
TCD
Dublin, Ireland
[email protected]
FESTJENS Nele
VIB - Ghent University
Zwijnaarde, Belgium
[email protected]
ELGHRAOUI Afif
San Diego State University
San Diego, United States
[email protected]
FLO Trude Helen
Norwegian University of Science and
Technology
Trondheim, Norway
[email protected]
ELLA Bhagyaraj
CSIR
Chandigarh, India
[email protected]
ELLIOTT Sarah
University of Minnesota - Twin Cities
Burnsville, United States
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
FLOTO Andres
University of Cambridge
Cambridge, United Kingdom
[email protected]
FOO Shi-Yan Caroline
EPFL
Lausanne, Switzerland
[email protected]
415
FOTOUHI Nader
TB Alliance
New York, United States
[email protected]
GHYS Anne
Janssen Pharmaceuticals
Beerse, Belgium
[email protected]
FREIRE Diana
EMBL-Hamburg
Hamburg, Germany
[email protected]
GICQUEL Brigitte
Institut Pasteur
Paris, France
[email protected]
GAEVAYA Ludmila
Bogomolets National Medical University
Kiev, Ukraine
[email protected]
GIJSBERS ALEJANDRE Abril
The Maastricht Multimodal Molecular
Imaging institute (M4I)
Maastricht, The Netherlands
[email protected]
GAGNEUX Sébastien
Swiss Tropical & Public Health Institute Basel
Basel, Switzerland
[email protected]
GAO Qian
Fudan University
Shangai, China
[email protected]
GARCIA-PELAYO M. Carmen
Animal & ant Health Agency (APHA)
Addlestone, United Kingdom
[email protected]
GARG Rajni
Amity University Gurgaon Haryana
New Delhi, India
[email protected]
GEIBEL Sebastian
University of Wuerzburg
Wuerzburg, Germany
[email protected]
GENGENBACHER Martin
National University of Singapore
Singapore
[email protected]
GENNARO Maria Laura
Rutgers University
Newark, United States
[email protected]
GERRICK Elias
Harvard School of Public Health
Boston, United States
[email protected]
GHANEM Marwan
McGill University
Montreal, Canada
[email protected]
416
GIL Magdalena
Institut Pasteur de Montevideo
Montevideo, Uruguay
[email protected]
GIRAUD GATINEAU Alexandre
Institut Pasteur
Dreux, France
[email protected]
GLENN Sarah
University of Leicester
Leicester, France
[email protected]
GOIG SERRANO Galo Adrián
Instituto de Biomedicina de Valencia - CSIC
Valencia, Spain
[email protected]
GONZALO-ASENSIO Jesús
University of Zaragoza
Zaragoza, Spain
[email protected]
GOPAL Pooja
National University of Singapore
Singapore
[email protected]
GORDON Stephen
University Colloge of Dublin
Ireland
[email protected]
GOUZY Alexandre
Weill Cornell Medecine
New York, France
[email protected]
GRABOWSKA Anna
London School of Hygiene and
Tropical Medicine
London, United Kingdom
peliroja@wp.
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
GRODE Leander
Vakzine Projekt Management GmbH
Hannover, Germany
[email protected]
HAFNEH Nor Azian
University of Leicester
Leicester, United Kingdom
[email protected]
GROESCHEL Matthias
Institut Pasteur
Paris, France
[email protected]
HANANE Issa
Université Paris Sud - Paris Saclay
Courbevoie, France
[email protected]
GRZEGORZEWICZ Anna
Colorado State University
Fort Collins, United States
[email protected]
HARTKOORN Ruben
Institut Pasteur de Lille
Lille, France
[email protected]
GUBELLINI Francesca
Institut Pasteur
Paris, France
[email protected]
HAUPTMANN Matthias
Research Center Borstel
Borstel, Germany
[email protected]
GUERIN Marcelo
CIC bioGUNE
Derio, Spain
[email protected]
HAWRYLUK Natalie
Celgene Global Health
San Diego, United States
[email protected]
GUILHOT Christophe
CNRS, IPBS
Toulouse, France
[email protected]
HE Le
Centre d'immunologie Marseille Luminy
Marseille, France
[email protected]
GUPTA Manish
Jawaharlal Nehru University,
Delhi, India
[email protected]
HEALY Claire
Weill Cornell Medicine
New York, United States
[email protected]
GUR Eyal
Ben-Gurion University of the Negev
Beer Sheva, Israel
[email protected]
HERNANDEZ Johana
University of Surrey
Guildford, United Kingdom
[email protected]
GURUMURTHY Meera
National University of Singapore
Singapore
[email protected]
HERRMANN Jean-Louis
Université de Versailles, Hôpital Poincaré
Versailles, France
[email protected]
GUSHCHIN Vladimir
Federal State Budgetary Institution
Moscow, Russia
[email protected]
HINGLEY-WILSON Suzie
University of Surrey
Guildford, United Kingdom
[email protected]
GUTIERREZ Claude
University Toulouse
Toulouse, France
[email protected]
HOHL Michael
University of Zurich
Zürich, Switzerland
[email protected]
HADIZADEH TASBITI Alireza
Institut Pasteur of Iran
Tehran, Iran
[email protected]
HOUBEN Edith
Vrije Universiteit Amsterdam
Amsterdam, The Netherlands
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
417
HRABALEK Alexandr
Charles University
Hradec Králové, Czech Republic
[email protected]
JANKUTE Monika
University of Birmingham
Birmingham, United Kingdom
[email protected]
HUANG Wei
Institut Pasteur
Paris, France
[email protected]
JAVID Asma
University of Birmingham
Birmingham, United Kingdom
[email protected]
HUDRISIER Denis
CNRS
Toulouse, France
[email protected]
JAVID Babak
Tsinghua University School of Medicine
Beijing, China
[email protected]
HUGONNET Jean-Emmanuel
UMRS 1138-UPMC INSERM UPD
Paris, France
[email protected]
JONG Robyn
UC Berkeley
Berkeley, United States
[email protected]
HUNT Debbie
The Francis Crick Institute
London, United Kingdom
[email protected]
JOSE Ainsa
Universidad de Zaragoza
Zaragoza, Spain
[email protected]
HWA Shi-Hsia
K-RITH
Durban, South Africa
[email protected]
JOSEPHSON Robert
University of Toronto
Toronto, Canada
[email protected]
IAKOBACHVILI Nino
Maastricht University
Maastricht, The Netherlands
[email protected]
JUNG Juyeon
Korea Research Institute of Bioscience and
Biotechnology
Daejeon, South Korea
[email protected]
JABOT-HANIN Fabienne
INSERM
Paris, France
[email protected]
JACKSON Mary
Colorado State University
Fort Collins, United States
[email protected]
JACOBS William R Jr
Albert Einstein College of Medicine
Bronx, United States
[email protected]
JÄGER Franziska
University of Dundee
Dundee, United Kingdom
[email protected]
JAKIMOWICZ Dagmara
University of Wroclaw
Wroclaw, Poland
[email protected].
418
KALSUM Sadaf
Linköping University
Linköping, Sweden
[email protected]
KAMARIZA Mireille
Stanford University
San Francisco, United States
[email protected]
KAMINSKI Lina
Albert Einstein College of Medicine
Bronx, United States
[email protected]
KANEKO Takushi
TB Alliance
Guilford, United States
[email protected]
KANNAN Nisha
NTNU
Trondheim, Norway
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
KANVATIRTH Panchali
University of Birmingham
Birmingham, United Kingdom
[email protected]
KOCH Anastasia
University of Cape Town
Cape Town, South Africa
[email protected]
KAAN Gilla
Bill and Melinda Gates Foundation
Seattle, United States
[email protected]
KOEKEN Valerie
Radboud University Medical Centre
Nijmegen, The Netherlands
[email protected]
KARABANOVICH Galina
Charles University, Faculty of Pharmacy in
Hradec Kralove
Hradec Králové, Czech Republic
[email protected]
KORDULÁKOVÁ Jana
Faculty of Natural Sciences,
Comenius University
Bratislava, Slovakia
[email protected]
KEISER Tracy
Albert Einstein College of Med
Bronx, United States
[email protected]
KOUL Anil
Janssen Pharmaceutica NV
Beerse, Belgium
[email protected]
KESTER Jemila
Harvard University
Cambridge, United States
[email protected]
KRAIGSLEY Alison
NIH/NIAID
North Bethesda, United States
[email protected]
KHANDELIA Arvind Kumar
Nanyang technological University
Singapore
[email protected]
KRÁTKÝ Martin
Charles University in Prague, Faculty of
Pharmacy in Hradec Králové
Hradec Králové, Czech Republic
[email protected]
KHOSRAVI BOROUJENI Azar Dokht
Ahvaz Jundishapur University of Medical
Sciences
Ahvaz, Iran
[email protected]
KREITMANN Louis
Lille University
Paris, France
[email protected]
KIM Hongmin
Yonsei University
Seoul, South Korea
[email protected]
KREMER Laurent
CNRS
Montpellier, France
[email protected]
KIM Jae Myung
Animal and ant Quaratine Service
Gyeongsangbuk-Do, South Korea
[email protected]
KREUTZFELDT Kaj
Weill Cornell Medicine
New York, United States
[email protected]
KIM Woo Sik
Yonsei University College of Medicine
Seoul, South Korea
[email protected]
KRISHNAMOORTHY Gopinath
Max anck Institute for Infection Biology
Berlin, Germany
[email protected]
KLIMESOVA Vera
Charles University
Hradec Kralove, Czech Republic
[email protected]
KUNDU Subhashri
National University of Singapore
Singapore
[email protected]
KNAUL Julia
Vakzine Projekt Managment GmbH
Hannover, Germany
[email protected]
LAANG Hannu
European Commission
Brussel, Belgium
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
419
LABOUDIE Patricia
Sanofi R&D
Marcy L'etoile, France
[email protected]
LASSERRE Moira
Institut Pasteur de Montevideo
Montevideo, Uruguay
[email protected]
LACKNER Gerald
Friedrich Schiller University Jena
Jena, Germany
[email protected]
LAU Alice
University of British Columbia
Vancouver, Canada
[email protected]
LAGRANGE Sophie
Sanofi R&D
Marcy L'etoile, France
[email protected]
LAUGHON Barbara
NIH/NIAID/DHHS/US Government
Bethesda, United States
[email protected]
LAI Rachel
The Francis Crick Institute
London, United Kingdom
[email protected]
LAURSEN Mette
EMBL Hamburg
Hamburg, Germany
[email protected]
LAIR Christine
Sanofi R&D
Marcy L'etoile, France
[email protected]
LE Nguyen-Hung
CNRS IPBS
Toulouse, France
[email protected]
LALVANI Ajit
Imperial College London
London, United Kingdom
[email protected]
LE CHEVALIER Fabien
Institut Pasteur
Paris, France
[email protected]
LAMBERT Linda
National Institute of Allergy, Immunology, and
Transantation, NIH
Bethesda, United States
[email protected]
LE MOIGNE Vincent
Université Versailles St-Quentin-en-YvelinesINSERM U1173
Montigny-Le-Bretonneux, France
[email protected]
LAMERS Meindert
MRC Laboratory of Molecular Biology
Cambridge, United Kingdom
[email protected]
LEFEBVRE Cyril
Institute of Pharmacology and Structural
Biology
Toulouse, France
[email protected]
LAMPRECHT Dirk
K-RITH
Durban, South Africa
[email protected]
LAPA E SILVA Jose Roberto
Federal University of Rio De Janeiro
Rio De Janeiro, Brazil
[email protected]
LARROUY-MAUMUS Gerald
Imperial College London
London, United Kingdom
[email protected]
LARSEN Michelle
Albert Einstein College of Medicine
Bronx, United States
[email protected]
420
LEFRANÇOIS Louise
University of Geneva
Geneva, Switzerland
[email protected]
LELIEVRE Joel
GlaxoSmithKline
Tres Cantos, Spain
[email protected]
LERM Maria
Linköping University
Linköping, Sweden
[email protected]
LERNER Thomas
The Francis Crick Institute
London, United Kingdom
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
LEUNG Vivian
Harvard School of Public Health
Boston, United States
[email protected]
MAHAMED Deeqa
K-RITH
Durban, South Africa
[email protected]
LIENARD Julia
Lund University
Lund, Sweden
[email protected]
MAJLESSI Laleh
Institut Pasteur
France
[email protected]
LIENHARTD Christian
OMS
Geneva, Switzerland
[email protected]
MAKAROV Vadim
A.N. Bach Institute of Biochemistry
Moscow, Russia
[email protected]
LINGE Irina
Central Institute for Tuberculosis
Moscow, Russia
[email protected]
MALM Sven
Research Center Borstel - Leibniz Center for
Medicine and Biosciences
Borstel, Germany
[email protected]
LOUNIS Nacer
Janssen Pharmaceutica
Beerse, Belgium
[email protected]
LOW Jian Liang
National University of Singapore
Singapore
[email protected]
LU Ping
VU University Amsterdam
Amsterdam, The Netherlands
[email protected]
LUGO Geanncarlo
CNRS, IPBS
Toulouse, France
[email protected]
LUKE Alderwick
University of Birmingham
Birmingham, United Kingdom
[email protected]
LUPIEN Andréanne
EPFL
Lausanne, Switzerland
[email protected]
MACHUGH David
University College Dublin
Dublin, Ireland
[email protected]
MAERK Mali
Norwegian University of Science and
Technology
Trondheim, Norway
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
MALONE Kerri
University College Dublin
Dublin, Ireland
[email protected]
MANGANELLI Riccardo
University of Padua
Padova, Italy
[email protected]
MANINA Giulia
Institut Pasteur
Paris, France
[email protected]
MAREKOVIĆ Ivana
University Hospital Centre Zagreb
Zagreb, Croatia
[email protected]
MARTIN Anandi
Ghent University
Gent, Belgium
[email protected]
MARTIN Carlos
University of Zaragoza
Zaragoza, Spain
[email protected]
MARTINEZ Mariano
Institut Pasteur
Paris, France
[email protected]
MASERUMULE Charlotte
Cambridge University
Cambridge, United Kingdom
[email protected]
421
MAZUREK Jolanta
IPBS, CNRS, France and KI, Sweden
Toulouse, France
[email protected]
MIZRAHI Valerie
University of Cape Town
Cape Town, South Africa
[email protected]
MDLULI Khisimuzi
TB Alliance
Brooklyn, United States
[email protected]
MODLIN Samuel
San Diego State University
San Diego, United States
[email protected]
MEIKLE Virginia
University of Alabama at Birmingham
Birmingham, United States
[email protected]
MONICA Monica
GlaxoSmithKline
Tres Cantos, Spain
[email protected]
MEILER Eugenia
GlaxoSmithKline
Tres Cantos, Madrid, Spain
[email protected]
MONTAMAT-SICOTTE Damien
Research Institute of the McGill University
Health Centre
Montreal, Canada
[email protected]
MÉNDEZ Melissa
Universidad Peruana Cayetano Heredia
Lima, Peru
[email protected]
MENDOZA-LOSANA Alfonso
GSK
Tres Cantos, Spain
[email protected]
MENDUM Tom
University of surrey
Guildford, United Kingdom
[email protected]
MERY Alexandre
University Lille1
Villeneuve D'ascq, France
[email protected]
MIKUSOVA Katarina
University of Bratislava
Slovakia
[email protected]
MINNIKIN David
University of Birmingham
Birmingham, United Kingdom
[email protected]
MITERMITE Morgane
University College Dublin
Dublin, Ireland
[email protected]
MITRA Avishek
University of Alabama at Birmingham
Birmingham, United States
[email protected]
422
MOOLLA Nabiela
University of Birmingham
Birmingham, United Kingdom
[email protected]
MORASKI Garrett
Montana State University
Bozeman, United States
[email protected]
MOREIRA Wilfried
National University of Singapore
Singapore
[email protected]
MOYNIHAN Patrick
University of Birmingham
Birmingham, United Kingdom
[email protected]
MUKAMOLOVA Galina
University of Leicester
Leicester, United Kingdom
[email protected]
MUKHERJEE Devika
NUS
Singapore
[email protected]
MUNSHI Tulika
St. George's, University of London
London, United Kingdom
[email protected]
MURPHY Leanne
University College Dublin
Leixlip, Ireland
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
NAGARAJA V
Indian Institute of Science
Bangalore, India
[email protected]
NISKANEN Mirja
University of Tampere
Tampere, Finland
[email protected]
NAMBI Subhalaxmi
Umass medical school
Worcester, United States
[email protected]
NOURSADEGHI Mahdad
University College London
London, United Kingdom
[email protected]
NATHAN Carl
Weill Cornell Medical College
New York, United States
[email protected]
ODERMATT Nina
EPFL
Lausanne, Switzerland
[email protected]
NEGATU Dereje
National University of Singapore
Singapore
[email protected]
OFORI-ANYINAM Nana Akua Boatema
Institute of Tropical Medicine
Antwerpen, Belgium
[email protected]
NEMECEK Jan
Charles University, Faculty of Pharmacy
Hradec Králové, Czech Republic
[email protected]
O'HARE Helen
University of Leicester
Leicester, United Kingdom
[email protected]
NEYROLLES Olivier
CNRS / IPBS
Toulouse, France
[email protected]
OREN Rosenberg
UCSF
San Francisco, United States
[email protected]
NGAN Grace
National University Singapore
Singapore
[email protected]
ORGEUR Mickael
Institut Pasteur
Paris, France
[email protected]
NGUYEN Quang Huy
L'Institut de recherche pour le développement
(IRD)
Montpellier, France
[email protected]
OSEI-WUSU Stephen
Noguchi Memorial Institute for Medical
Research, University of Ghana
Accra, Ghana
[email protected]
NICOD Charlotte
ETH Zürich
Zürich, Switzerland
[email protected]
OSMAN Morwan
University of Cambridge
Cambridge, United Kingdom
[email protected]
NIEDERWEIS Michael
University of Alabama at Birmingham
Homewood, United States
[email protected]
O'SULLIVAN Mary
Trinity College Dublin
Dublin, Ireland
[email protected]
NIEL Van Wyk
CNRS
Montpellier, France
[email protected]
OTCHERE Isaac Darko
Noguchi Memorial Institute for Medical
Research, University of Ghana
Accra, Ghana
[email protected]
NIEMANN Stefan
Research Center Borstel
Borstel, Germany
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
OTTENHOFF Tom
Leiden University Medical Center
Leiden, The Netherlands
[email protected]
423
OTTER Ashley
Royal Veterinary College
London, United Kingdom
[email protected]
PETHE Kevin
Nanyang Technological University
Singapore
[email protected]
OUAKED Nadia
GSK
Rixensart, Belgium
[email protected]
PETRUCCIOLI Elisa
National Institute for Infectiouse Diseases
Lazzaro Spallanzani
Roma, Italy
[email protected]
PALOMINO Juan Carlos
Ghent University
Gent, Belgium
[email protected]
PALUCCI Ivana
università cattolica del sacro cuore
Roma, Italy
[email protected]
PARASA Venkata Ramanarao
Linkoping University
Linkoping, Sweden
[email protected]
PARK Yu Mi
NIH
Bethesda, United States
[email protected]
PARRET Annabel
EMBL Hamburg
Hamburg, Germany
[email protected]
PASCA Maria Rosalia
University of Pavia, Italy
Pavia, Italy
[email protected]
PAWLIK Alexandre
Institut Pasteur
Paris, France
[email protected]
PÉREZ SÁNCHEZ Irene
University of Zaragoza
Zaragoza, Spain
[email protected]
PERRIN Pascale
Université Montpellier
Montpellier, France
[email protected]
PETERS Peter
Maastricht University
Amsterdam, The Netherlands
[email protected]
424
PIETRETTI Danilo
Institute Pharmacologie et Biologie Structurale
IPBS - Centre National de la Recherche
Scientifique
Ramonville St Agne, France
[email protected]
PITON Jérémie
EPFL
Lausanne, France
[email protected]
POJER Florence
EPFL
Lausanne, Switzerland
[email protected]
PRADOS-ROSALES Rafael
CICbioGUNE
Derio, Spain
[email protected]
PRIESTMAN Miles
Imperial College London
London, United Kingdom
[email protected]
PRITCHARD Jennifer
Institute of Structural Molecular Biology
Chelmsford, Essex, United Kingdom
[email protected]
PROSSER Gareth
NIH
Bethesda, United States
[email protected]
PYM Alexander
K-RITH
Durban, South Africa
[email protected]
QUEVAL Christophe
Institut Pasteur
Paris, France
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
QUICK Isadora
INSERM
Paris, France
[email protected]
REGO Hesper
Yale
Boston, United States
[email protected]
RAFFETSEDER Johanna
Linköping University
Linköping, Sweden
[email protected]
REILING Norbert
Research Center Borstel
Borstel, Germany
[email protected]
RAJWANI Rahim
The Hong Kong Polytechnic University
Hong Kong, China
[email protected]
REMUINAN Modesto
GSK
Tres Cantos, Madrid, Spain
[email protected]
RAKOTOSAMIMANANA Niaina
Institut Pasteur de Madagascar
Antananarivo, Madagascar
[email protected]
RESENDE Mariana
I3S/IBMC
Porto, Portugal
[email protected]
RAMAKRISHNAN Lalita
University of Cambridge
Cambridge, United Kingdom
[email protected]
RICCARDI Giovanna
University of Pavia
Pavia, Italy
[email protected]
RAMON GARCIA Santiago
The University of British Columbia
Vancouver, Canada
[email protected]
RICHTER Adrian
Martin Luther Universität Halle Wittenberg
Rossleben, Germany
[email protected]
RAO Saroja
Dayananda Sagar College of Engineering
Bengaluru, India
[email protected]
RIOU Catherine
University of Cape Town
Cape Town, South Africa
[email protected]
RASTOGI Nalin
Institut Pasteur de la Guadeloupe
Abymes, France
[email protected]
RIVERO-LEZCANO Octavio
Comejo Asistencial Universitario de León
Leon, Spain
[email protected]
RAVELLI Raimond
Maastricht University
Maastricht, The Netherlands
[email protected]
ROBERTSON Brian
Imperial College London
London, United Kingdom
[email protected]
RAWKINS Ann
Public Health England
Salisbury, United Kingdom
[email protected]
ROCA SOLER Francisco Jose
University of Cambridge
Cambridge, United Kingdom
[email protected]
RAY Peter
University of Dundee
Stirling, United Kingdom
[email protected]
ROCK Jeremy
Harvard School of Public Health
Boston, United States
[email protected]
REDINGER Natalja
Research Center Borstel
Borstel, Germany
[email protected]
RODRIGO-UNZUETA Ane
University of Cape Town
Cape Town, South Africa
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
425
RODRIGUEZ Daniela
University of Cambridge
Cambridge, United Kingdom
[email protected]
SAITO Kohta
Weill Cornell Medicine
New York, United States
[email protected]
RODRIGUEZ-RIVERA Frances
Stanford University
Stanford, United States
[email protected]
SANCHEZ MONTALVA Adrian
Vall d'Hebron Hospital. Universitat autonoma de
Barcelona
Barcelona, Spain
[email protected]
ROE Jennifer
University College London
London, United Kingdom
[email protected]
ROH Jaroslav
Charles University in Prague, Faculty of
Pharmacy
Hradec Králové, Czech Republic
[email protected]
SANDER Peter
University of Zurich
Zurich, Switzerland
[email protected]
SANS Stéphanie
Sanofi R&D
Marcy L'etoile, France
[email protected]
ROMBOUTS Yoann
IPBS
Toulouse, France
[email protected]
SANTOS Mariana
The Francis Crick Institute
London, United Kingdom
[email protected]
ROUBERT Christine
Sanofi R&D
Marcy L'etoile, France
[email protected]
SANTUCCI Pierre
CNRS
Marseille, France
[email protected]
RUBIN Eric
Harvard TH Chan Scholl of Public Health
Boston, Ma, United States
[email protected]
SARATHY Jickky Palmae
National University of Singapore
Singapore
[email protected]
RUESEN Carolien
Radboud university medical center Nijmegen
Nijmegen, The Netherlands
[email protected]
SAVINO Maria
LABCYTE INC
Sunnyvale, United States
[email protected]
RULLAS Joaquin
GlaxoSmithKline
Tres Cantos, Spain
[email protected]
SAWYER Elizabeth
London School of Hygiene and Tropical
Medicine
London, United Kingdom
[email protected]
RUSSELL David
Cornell University
Ithaca, Ny, United States
[email protected]
RUSTAD Tige
Center for Infectious Disease Research
Seattle, United States
[email protected]
SAELENS Joseph
Duke University
Durham, United States
[email protected]
426
SAYES Fadel
Institut Pasteur
Paris, France
[email protected]
SCHNAPPINGER Dirk
Weill Cornell Medicine
New York, United States
[email protected]
SCHNEIDER Bianca
Research Center Borstel
Borstel, Germany
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
SEEGER Markus
University of Zurich
Zurich, Switzerland
[email protected]
SIMEONE Roxane
Institut Pasteur
France
SERAFINI Agnese
The Francis Crick Institute
London, United Kingdom
[email protected]
SINGH Ramandeep
Translational Health Science and Technology
Institute
Haryana, India
[email protected]
SEWGOOLAM Bevika
University of Cape Town
Cape Town, South Africa
[email protected]
SIROY Axel
Maastricht University
Maastricht, France
[email protected]
SHANAHAN Jonathan
University of Cambridge
Cambridge, United Kingdom
[email protected]
SMYTH Alicia
University College Dublin
Dublin, Ireland
[email protected]
SHARMA Aditya Kumar
Institute of genomic and integrative biology
Delhi, India
[email protected]
SOLDATI Thierry
University of Geneva
Geneva, Switzerland
[email protected]
SHARMA Ashish
Indian Institute of Technology New Delhi
New Delhi, India
[email protected]
SOLER Paloma
EPFL
Lausanne, Switzerland
[email protected]
SHARMA Meenu
Public Health Agency of Canada
Winnipeg, Canada
[email protected]
SOMMER Raphael
EPFL
Lausanne, Switzerland
[email protected]
SHARON Kendall
The Royal Veterinary College
London, United Kingdom
[email protected]
SOMODY Laurence
SANOFI AVENTIS R et D
Marcy L'etoile, France
[email protected]
SHETTY Annanya
National University of Singapore
Singapore
[email protected]
SONG Ok-Ryul
INSERM
Lille, France
[email protected]
SHIN Sung Jae
Yonsei University College of Medicine
Seoul, South Korea
[email protected]
SOOD Sakshi
The Francis Crick Institute
London, United Kingdom
[email protected]
SHLEEVA Margarita
A.N. Bach Institute of Biochemistry
Moscow, Russia
[email protected]
SOTO Ramon
University of Birmingham
Birmingham, United Kingdom
[email protected]
SIKRI Kriti
All India Institute of Medical Sciences
New Delhi, India
[email protected]
SOURIANT Shanti
IPBS, CNRS UMR 5089
Toulouse, France
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
427
SOUSA Jeremy
i3S - Instituto de Investigação e Inovação em
Saúde
Porto, Portugal
[email protected]
SPEER Alexander
VU University Medical Center
Amsterdam, The Netherlands
[email protected]
STAACK Sonja
EMBL Hamburg
Hamburg, Germany
[email protected]
STANLEY Sarah
University of California, Berkeley
Berkeley, United States
[email protected]
STEIGEDAL Magnus
NTNU
Trondheim, Norway
[email protected]
STENDAHL Olle
Linköping University
Linköping, Sweden
[email protected]
STINEAR Timothy
University of Melbourne
Merlbourne, Australia
[email protected]
STRAND Trine
Norwegian University of Science and
Technology
Trondheim, Norway
[email protected]
SUBBARAO Vani
Imperial College
London, United Kingdom
[email protected]
SUN Jim
University of Alabama at Birmingham
Birmingham, United States
[email protected]
SUPY Philip
CNRS
Lille, France
[email protected]
TAILLEUX Ludovic
Institut Pasteur
Paris, France
[email protected]
428
TAKIFF Howard
IVIC/Pasteur
Paris, France
[email protected]
TANVIR Zaid
TB ALLIANCE
New York, United States
[email protected]
TASSINARI Matteo
Institut Pasteur
Paris, France
[email protected]
TELEVANTOS Constantinos
University of Cambridge
Cambridge, United Kingdom
[email protected]
TOMLINSON Gillian
University College London
London, United Kingdom
[email protected]
TROFIMOV Valentin
Insitut Pasteur de Lille
Lille, France
[email protected]
TSODIKOV Oleg
University of Kentucky
Lexington, United States
[email protected]
VALAFAR Faramarz
San Diego State University
San Diego, United States
[email protected]
VALASKOVA Lenka
Faculty of Pharmacy in Hradec Kralove,
Charles University
Hradec Kralove, Czech Republic
[email protected]
VAN DER MEEREN Olivier
GlaxoSmithKline Vaccines
Rixensart, Belgium
[email protected]
VAN DER WEL Nicole
AMC
Amsterdam, The Netherlands
[email protected]
VAN DER WESTHUYZEN Renier
University of Cape Town
Cape Town, South Africa
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
VARELA RAMIREZ Cristian
University of Birmingham
Birmingham, United Kingdom
[email protected]
VINSOVA Jarmila
Faculty of Pharmacy, Charles University
Hradec Kralove, Czech Republic
[email protected]
VASILEVA Elena
Saint-Petersburg Pasteur Institute
Saint-Petersburg, Russia
[email protected]
WADDELL Simon
University of Sussex
Brighton, United Kingdom
[email protected]
VAUBOURGEIX Julien
Weill Cornell Medicine
New York, United States
[email protected]
WAGNER Jeffrey
Harvard T.H. Chan School of Public Health
Boston, United States
[email protected]
VAZIRI Farzam
Institut Pasteur of Iran
Tehran, Iran
[email protected]
WANG Chuan
Fudan University
Shanghai, China
[email protected]
VEGA DOMINGUEZ Perla Jazmin
University of Birmingham
Birmingham, United Kingdom
[email protected]
WARNER Digby
University of Cape Town
Cape Town, South Africa
[email protected]
VERGNOLLE Olivia
Albert Einstein College of Medicine
Bronx, United States
[email protected]
WEHENKEL Anne Marie
Institut Pasteur
Paris, France
[email protected]
VERMA Amit Kumar
University of Cambridge
Cambridge, United Kingdom
[email protected]
WHITWORTH Laura
University of Cambridge
Cambridge, United Kingdom
[email protected]
VERRALL Ayesha
University of Otago
Wellington South, New Zealand
[email protected]
WILKINSON Robert
Imperial College London
United Kingdom
[email protected]
VIA Laura Ellen
NIAID
Bethesda, United States
[email protected]
WILLIAM Conrad
Cambridge University
Cambridge, United Kingdom
[email protected]
VILCHEZE Catherine
Howard Hughes Medical Institute at AECOM
Flushing, New York, United States
[email protected]
WILLIAMS Kerstin
University of Surrey
Guildford, United Kingdom
[email protected]
VILJOEN Albertus
CNRS
Montpellier, France
[email protected]
WILMANNS Matthias
European Molecular Biology Laboratory
Hamburg, Germany
[email protected]
VILLELLAS Cristina
Janssen Pharmaceutica
Beerse, Belgium
[email protected]
WILSON Colin
Drug Discovery and Development Center,
University of Cape Town
Cape Town, South Africa
[email protected]
Tuberculosis 2016 – September 19-23, 2016 – Paris, France
429
WINTER Nathalie
INRA
Nouzilly, France
[email protected]
YAMADA Yoshiyuki
National University of Singapore
Singapore
[email protected]
WIREDU Adwoa
Noguchi Memorial Institute for medical
research, University of Ghana
Greater Accra, Ghana
[email protected]
YEBOAH-MANU Dorothy
Noguchi Memorial Institute for Medical
Research, University of Ghana
Legon, Accra, Ghana
[email protected]
WITT Karolina
QMUL
Feltham, United Kingdom
[email protected]
YEE Michelle
National University of Singapore
Singapore
[email protected]
WOLSCHENDORF Frank
University of Alabama at Birmingham
Birmingham, United States
[email protected]
ZHU Junhao
Tsinghua University
Beijing, China
[email protected]
WU Katherine
Harvard University
Cambridge, Ma, United States
[email protected]
ZHURAVLEV Viacheslav
St.Petersburg Research Institute of
Phthisiopulmonology
Saint-Petersburg, Russia
[email protected]
WU Mulu
National University of Singapore
Singapore
[email protected]
XIAO Han
University of St.Andrews
St.andrews, United Kingdom
[email protected]
ZOUAOUI-FRIGUI Wafa
Institut Pasteur
Paris, France
[email protected]
XU Zhujun
National University of Singapore
Singapore
[email protected]
430
Tuberculosis 2016 – September 19-23, 2016 – Paris, France